Biotechnology of plasma proteins 9781439850268, 1439850267

''Discussing the role of plasma proteins in current biotechnology, this book describes the protein composition

347 76 3MB

English Pages xiii, 446 p.. ; 24 cm [456] Year 2013

Report DMCA / Copyright

DOWNLOAD PDF FILE

Table of contents :

Content: Blood, Plasma, Cells, and Other Biological Fluids Plasma Fractionation and Plasma Protein Products Methods for the Analysis of Plasma and Plasma Protein Fractions Albumin Structure of Albumin In Vitro Chemical Modification of Albumin Albumin as a Carrier Ligand-Binding by Albumin Purification and Characterization of HSA for Clinical Use Biological Activity of Albumin Enzymatic Activity of Albumin Clinical Use of Albumin Albumin as Diagnostic/Biomarker Pharmacokinetics of Albumin Analytical Methods for Albumin Including Use of Albumin as Standard for Analytical Methods Albumin as Excipient Conclusion References Plasma Immunoglobulins Factor VIII and von Willebrand Factor von Willebrand Factor Economic Issues Impacting the Hemophilia Business Conclusions and Future Directions References Plasma Proteinase Inhibitors Antithrombin alpha1-Antitrypsin (alpha1-Antiprotease Inhibitor, SERPINA1) Heparin Cofactor II (SERPIND1) alpha2-Macroglobulin Tissue Factor Pathway Inhibitor alpha2-Antiplasmin C1-Inhibitor (C1-Esterase Inhibitor) Plasminogen Activator Inhibitor-1 Protein C Inhibitor (Plasminogen Activator Inhibitor-3) References Vitamin K-Dependent Proteins Miscellaneous Plasma Proteins Fibrinogen Fibrin Sealant Autologous Fibrin Sealant Fibrinogen and Tissue Soldering Thrombin and Fibrin Foam Fibrinogen Plastics Thrombin Plasminogen and Plasmin Butyrylcholinesterase Fibronectin References
Abstract: ''Discussing the role of plasma proteins in current biotechnology, this book describes the protein composition of human plasma, the fractionation of plasma to obtain therapeutic proteins, and the analysis of these products. It delineates the path from plasma products to recombinant products, and highlights products from albumin, intravenous immunoglobins, and coagulation. It offers a comprehensive review of current techniques for the analysis of proteins including electrophoresis, chromatography, spectrophotometry, and mass spectrometry as well as updates not published since 1975''--Provided by publisher
Recommend Papers

Biotechnology of plasma proteins
 9781439850268, 1439850267

  • 0 0 0
  • Like this paper and download? You can publish your own PDF file online for free in a few minutes! Sign Up
File loading please wait...
Citation preview

Biotechnology of Plasma Proteins Roger L. Lundblad

Biotechnology of Plasma Proteins

P R O T E I N S C I E N C E S E R I E S SERIES EDITOR Roger L. Lundblad Lundblad Biotechnology Chapel Hill, North Carolina, U.S.A.

PUBLISHED TITLES Application of Solution Protein Chemistry to Biotechnology Roger L. Lundblad Approaches to the Conformational Analysis of Biopharmaceuticals Roger L. Lundblad Biotechnology of Plasma Proteins Roger L. Lundblad Chemical Modification of Biological Polymers Roger L. Lundblad Development and Application of Biomarkers Roger L. Lundblad

Biotechnology of Plasma Proteins

Boca Raton London New York

CRC Press is an imprint of the Taylor & Francis Group, an informa business

CRC Press Taylor & Francis Group 6000 Broken Sound Parkway NW, Suite 300 Boca Raton, FL 33487-2742 © 2013 by Taylor & Francis Group, LLC CRC Press is an imprint of Taylor & Francis Group, an Informa business No claim to original U.S. Government works Version Date: 20120615 International Standard Book Number-13: 978-1-4398-5027-5 (eBook - PDF) This book contains information obtained from authentic and highly regarded sources. Reasonable efforts have been made to publish reliable data and information, but the author and publisher cannot assume responsibility for the validity of all materials or the consequences of their use. The authors and publishers have attempted to trace the copyright holders of all material reproduced in this publication and apologize to copyright holders if permission to publish in this form has not been obtained. If any copyright material has not been acknowledged please write and let us know so we may rectify in any future reprint. Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic, mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and recording, or in any information storage or retrieval system, without written permission from the publishers. For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://www. copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-7508400. CCC is a not-for-profit organization that provides licenses and registration for a variety of users. For organizations that have been granted a photocopy license by the CCC, a separate system of payment has been arranged. Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for identification and explanation without intent to infringe. Visit the Taylor & Francis Web site at http://www.taylorandfrancis.com and the CRC Press Web site at http://www.crcpress.com

This work is dedicated to the three Mikes: Grifith, Martinez, and Wood, who taught me more than I ever wanted to know about the plasma biz.

Contents Preface.......................................................................................................................ix Acknowledgments .....................................................................................................xi Author .................................................................................................................... xiii Chapter 1

Blood, Plasma, Cells, and Other Biological Fluids .............................. 1

Chapter 2

Plasma Fractionation and Plasma Protein Products ........................... 19

Chapter 3

Methods for the Analysis of Plasma and Plasma Protein Fractions ......47

Chapter 4

Albumin ............................................................................................. 83 Structure of Albumin ......................................................................... 86 In Vitro Chemical Modiication of Albumin ................................... 101 Albumin as a Carrier ........................................................................ 113 Ligand-Binding by Albumin ............................................................ 121 Puriication and Characterization of HSA for Clinical Use............. 123 Biological Activity of Albumin ........................................................ 125 Enzymatic Activity of Albumin ....................................................... 126 Clinical Use of Albumin .................................................................. 129 Albumin as Diagnostic/Biomarker .................................................. 132 Pharmacokinetics of Albumin ......................................................... 133 Analytical Methods for Albumin Including Use of Albumin as Standard for Analytical Methods................................................. 134 Albumin as Excipient ....................................................................... 136 Conclusion ........................................................................................ 136 References ........................................................................................ 136

Chapter 5

Plasma Immunoglobulins ................................................................. 183

Chapter 6

Factor VIII and von Willebrand Factor ............................................ 233 von Willebrand Factor ...................................................................... 253 Economic Issues Impacting the Hemophilia Business .................... 255 Conclusions and Future Directions .................................................. 257 References ........................................................................................ 257

vii

viii

Chapter 7

Contents

Plasma Proteinase Inhibitors ............................................................ 285 Antithrombin .................................................................................... 285 α1-Antitrypsin (α1-Antiprotease Inhibitor, SERPINA1) ................... 293 Heparin Cofactor II (SERPIND1) .................................................... 301 α2-Macroglobulin ............................................................................. 301 Tissue Factor Pathway Inhibitor .......................................................304 α2-Antiplasmin ................................................................................. 311 C1-Inhibitor (C1-Esterase Inhibitor) ................................................. 315 Plasminogen Activator Inhibitor-1 ................................................... 319 Protein C Inhibitor (Plasminogen Activator Inhibitor-3) ................. 322 References ........................................................................................ 323

Chapter 8

Vitamin K–Dependent Proteins ....................................................... 367

Chapter 9

Miscellaneous Plasma Proteins ........................................................ 401 Fibrinogen ........................................................................................ 401 Fibrin Sealant ...................................................................................402 Autologous Fibrin Sealant ................................................................408 Fibrinogen and Tissue Soldering ......................................................408 Thrombin and Fibrin Foam .............................................................. 410 Fibrinogen Plastics ........................................................................... 411 Thrombin .......................................................................................... 412 Plasminogen and Plasmin ................................................................ 412 Butyrylcholinesterase ....................................................................... 415 Fibronectin ....................................................................................... 415 References ........................................................................................ 417

Preface For some time, I had been wanting to write this book and inally, there was an opportunity. Had I thoroughly understood the task, I would have chosen a less complex topic. The entire book could have been devoted to albumin alone. As indicated at several instances in the text, I was continually realizing how little I knew about some of the topics, despite having worked with plasma proteins for some 50 years, and I hope that the reader will likewise ind some new information. The other point is that plasma fractionation is a mature industry and there are those that promised its demise after the development of recombinant factor VIII some 20+ years ago. Contrary to that prediction, the industry has grown and undergone some consolidation. In addition, a new facility for plasma fractionation has been developed in Brazil and perhaps in other geographies as well. An aggressive program on blood safety has made plasma products safe. While some therapeutics such as factor VIII and factor IX have seen successful recombinant products, other therapeutics such as albumin and intravenous immunoglobulin continue to be dominated by plasma products. Sophisticated methods of analysis, such as mass spectrometry, have increased our understanding of the complexity of plasma, while other works have shown the importance of classic plasma proteins in extravascular function. I hope that the reader inds this work interesting and is encouraged to answer some of the many remaining questions about plasma proteins. Roger L. Lundblad Chapel Hill, North Carolina

ix

Acknowledgments The author is indebted to the editorial staff of CRC Press/Taylor & Francis Group in Boca Raton for their patience and support. In particular, he wants to acknowledge the contributions by Barbara Norwitz, who perhaps merits sainthood, and Jill Jurgensen, to this work. It takes a bit of time for an author to truly understand the importance of the editorial staff in the production of a book. The author is also indebted to the Library of Congress and the Libraries of the University of North Carolina at Chapel Hill for maintaining collections of excellence.

xi

Author Roger L. Lundblad is a native of San Francisco, California. He received his undergraduate education at Paciic Lutheran University and his PhD in biochemistry at the University of Washington. After his postdoctoral work in the laboratories of Stanford Moore and William Stein at The Rockefeller University, he joined the faculty of the University of North Carolina at Chapel Hill. He joined the Hyland Division of Baxter Healthcare in 1990. Currently, Dr. Lundblad works as an independent consultant at Chapel Hill, North Carolina, and writes on biotechnological issues. He is an adjunct professor of pathology at the University of North Carolina at Chapel Hill.

xiii

1

Blood, Plasma, Cells, and Other Biological Fluids

Biotechnology has entered our culture as something that solves many problems in health and agriculture. There is also a belief that biotechnology is something that appeared in the twentieth century. Biotechnology is deined as the use of biological methods and entities such as enzymes and/or organisms to produce a product. Given this broad deinition, biotechnology can range from baking bread, making yogurt, and brewing beer to the use of recombinant DNA technology to produce therapeutic proteins. So, when a “talking head” is pontiicating about the biotechnology industry, it may be about beer, bread, or Betaseron®. A similar confusion exists in the use of the term biopharmaceutical.1 In the context of the current work, biotechnology is used to describe the use of human blood plasma as a starting material for the manufacture of therapeutic products. Where relevant, the production of recombinant products based on plasma proteins is discussed as is the use of a protein as a diagnostic analyte. The use of blood plasma as a source of biomarkers is not discussed in detail, and the reader is referred to other sources.2–4 In my previous books, I had written the irst chapter irst, which seemed to make some sense. As I have matured (a colleague at Iowa suggested that I pass on candles for the birthday cake this year as a contribution to stemming global warming), I have noticed that the irst chapters appear to have been (a) written by someone else (guilt falls on a cat, Callee, who shares my domicile) or (b) written by the author in one of his earlier states of mind. This time, the irst is last; however, the last is not irst. I want to emphasize that, despite having spent most of my professional career working on blood coagulation proteins (there was a brief time of sanity in New York), I really found out how much I did not know about plasma proteins until I started writing this book. There were a number of epiphanies but none for me was larger than inding that substantial quantities of plasma proteins are in the extravascular space; in some instances such as with serpins, there is signiicant function in the extravascular space. An examination of physiology texts5–8 will provide a number of functions for blood, including oxygen transport, nutrient and hormone transport, and temperature regulation. Other functions such as hemostasis are intended for the maintenance of the integrity of the vascular system. Schaller and colleagues9 recently published an excellent book on the molecular and structural biology of the plasma proteins. These researchers developed a list of eight classes of plasma proteins ranging from proteins secreted by hard tissues (classical plasma proteins) to foreign proteins contributed by pathogenic and nonpathogenic organisms. They were then discussed by function, starting with blood coagulation and ending with transport and storage. I was encouraged to see that these workers also had a category for additional proteins, as the current work includes Chapter 9 1

2

Biotechnology of Plasma Proteins

(Miscellaneous Plasma Proteins). With reference to Schaller and colleagues,9 the current work is directed toward the use of “classical plasma proteins” and includes immunoglobulins, which would fall into a bit of a gray zone with their classiication system. I have no quarrel with their classiication and would note that several of their categories such as tissue leakage proteins, aberrant secretions, and receptor ligands are also diagnostic target/biomarkers. It is useful to appreciate that the extensive use of blood for the diagnosis of systemic disorders is a somewhat recent practice and the development/application of biomarkers is an even more recent practice2–4 in the sense that blood has been used in religious rites and furniture manufacture for centuries. Body water is divided into intracellular and extracellular luid.10 The bulk of body water (approximately 70%) is intracellular luid, with the remainder (30%) being extracellular luid. The extracellular luid can be, in turn, allocated between extravascular luid and intravascular luid, which are in dynamic equilibrium.10 Extravascular luid can be further divided into interstitial luid and transcellular luid. Transcellular luid includes luids such as cerebrospinal luid, synovial luid, and ocular luid (aqueous humor of the eye). There is luid and solute exchange between the various compartments of the extravascular luid, but the exchange is variable according to the compartment. The ratio of IgG in cerebrospinal luid to plasma is 0.003, while it is 0.80 for urea, which demonstrates the relative impermeability of the cerebrospinal barrier.11 For comparison, the ratio of IgG concentration between interstitial luid and plasma is 0.52 while albumin is 0.62.12 The exchange of luid between the intravascular luid and the extravascular space is dependent on the physiological state of the individual.13,14 There is also a local variation in distribution.15,16 Bar and coworkers15 observed that insulin stimulated the movement of insulin-like growth factor (IGF) binding protein-1 (IGFBP-1) from the vascular space to the tissue in an isolated beating heart of rat, while there was no effect on IGFBP-2 but a decrease in endothelial cell IGF binding protein. Juweld and coworkers16 observed that while the concentration of albumin is higher than that of IgG in normal tissue, the ratio approaches unity in the inlamed tissue. Reed and Rubin17 suggest that the edema response in inlammation is of functional signiicance in promoting the diffusion of plasma protein into the inlamed tissue. The transcapillary escape rate (transport from the intravascular space to the extravascular space) of albumin, IgG, and IgM increased in angiotensinII-induced hypertension; the relative increase was much higher for IgG and IgM than for albumin.18 Transport from the vascular space depends on endothelial permeability; transport of plasma proteins can occur either by transcellular or by paracellular processes.19,20 As can be surmised from the material cited earlier as well as the material on individual proteins to be presented in later chapters, a substantial portion of a given plasma protein can be found in the interstitial space, as the volume of extravascular luid is two to three times the size of the plasma volume.21,22 Consider, for example, the interstitial luid of skin that contains a substantial amount of albumin with considerable exchange with plasma.23 Binding of drugs to albumin is suggested to improve distribution over the tissues.24 In this study,24 it was found that the diffusion of a drug to deeper tissues after topical application is facilitated by binding to albumin. Other recent studies emphasize the importance of protein binding of drugs as a critical factor in early drug development.25,26

3

Blood, Plasma, Cells, and Other Biological Fluids

The number of unique proteins reported in human blood plasma has increased because of the sophistication of analytical techniques. Thus, while there were “... more than 100 ...” proteins isolated from plasma in 1980,27 the number in 2011 varies from approximately 1000 to 4000 (Table 1.1). The work by Schaller and colleagues was mentioned earlier,9 which emphasized the diversity of contributions TABLE 1.1 Number of Proteins in Various Biological Fluids Fluida

Number

Comment

Reference

Plasma

4590

[139]

Plasma

3654

Plasma

2928

Plasma

>3000

Plasma

697

Plasma Plasma

2698 1175

Plasma

1929

Source of plasma and anticoagulant not provided. Sample depleted of albumin and IgG prior to analysis by high-performance liquid chromatography (HPLC) (reversephase and cation exchange techniques)/mass spectrometry (MS). There are proteins that interact with albumin and/or IgG that are removed in the depletion step Pooled plasma from trauma patients (12); conditions of storage and anticoagulant not provided. Samples depleted of 12 major proteins prior to analysis (stated to be 96% of the total plasma protein mass) Ethylenediaminetetraacetic acid (EDTA) plasma was obtained and depleted of six major proteins prior to analysis. Peptides were obtained by protease digestion and separated by strong cation exchange or reverse-phase technique. Efluent fractions were assayed by mass spectrometry This igure is cited in this chapter. Also, these researchers noted that 10 proteins constituted 90% of the mass in plasma Pooled EDTA plasma from two male subjects; depletion of six most abundant proteins (albumin, transferrin, haptoglobin, α1-antitrypsin, IgA, and IgG);b analysis was done by gel electrophoresis/HPLC/mass spectrometry Number is cited by these researchers The data were collated from four different data sets to obtain 1175 distinct gene products Advanced computational methods were used to analyze combined data sets obtained by various analytical techniques. A total of 20,433 distinct peptides were identiied and used to establish a highly nonredundant set of 1929 proteins with a false discovery rate of 1%

[140]

[141]

[142]

[29]

[143] [144] [145]

(continued)

4

Biotechnology of Plasma Proteins

TABLE 1.1 (Continued) Number of Proteins in Various Biological Fluids Fluida

Number

Comment

Reference

Interstitial luid

525

Tumor interstitial luid from a head-and-neck carcinoma; it suggests that tumor interstitial luid may have a high proportion of tumor-speciic proteins with potential as biomarkers. A total of 208 proteins were common to plasma, 402 common to saliva, and 180 common to an ovarian cell line

[30]

Urine

1543

Use of SDS-PAGE/RP-HPLC with protease digestion of efluent fractions followed by MS analysis. Approximately half the proteins were membrane proteins as determined by Gene Ontology analysis

[146]

Urine

>1500

Many were membrane-bound proteins

[147]

Amniotic luid

842

LC/MS/MS and 2-D SDS PAGE/LC/MS/MS of 16 samples. A total of 36% of the proteins in amniotic luid were also found in plasma

[148]

Cervical– vaginal luidc

150

Two-dimensional HPLC, 2-D electrophoresis with mass spectrometric analysis identiied 150 unique proteins, of which 77 were unique to cervical–vaginal luid while 56 were also found in serum and 17 in amniotic luid

[149]

Perilymph

71

LC/MS/MS used to identify the proteins in human perilymph; 271 proteins identiied with 71 proteins common among the four individual samples

[150]

Saliva

2290

Approximately 27% of the whole saliva proteins were found in plasma. Note that this was whole saliva, not individual glandular (parotid, submaxillary) salivasd

[6]

Synovial luid

135

A total of 135 “high abundance” proteins were identiied in synovial luid. Unique proteins were deduced from mass spectrometric analysis of peptides obtained from the digestion of samples from SDS-PAGE. A total of 18 proteins were found to result from the cutaneous puncture procedure and removed from the reference set

[151]

Cerebrospinal luide

264–2630

As with the analysis of plasma and serum, the number of proteins identiied increased with increasing analytic sophistications. The results suggested considerable individuality as well as signiicant differences in aging

[152–155]

5

Blood, Plasma, Cells, and Other Biological Fluids

TABLE 1.1 (Continued) Number of Proteins in Various Biological Fluids Fluida Brain extracellulare luid

a b

c

d

e

Number 27

Comment 2D-SDS PAGE of a microdialysate showed approximately 160 spots, while MALDITOF-MS provided support for 95 proteins. It is suggested that after consideration of posttranslational modiications and proteolysis, data supported 27 individual proteins

Reference [156]

All luid sources are human. Depletion of the most abundant proteins from plasma has been shown to also remove proteins in addition to the “most abundant proteins.”157 Analytical results can also be inluenced by the storage temperature of plasma158 and the anticoagulant used.159 Cervical–vaginal luid has also been found useful for the measurement of fetal ibronectin and/ or insulin-like growth factor binding protein-1 for the prediction of preterm labor.160 Whole saliva is comprised of secretions from the submaxillary gland, parotid gland, sublingual, and minor salivary glands. In addition, there is contribution, except with edentulous individuals, from gingival crevicular luid, considered to be an ultrailtrated sample of plasma. There are some data obtained from the proteomic analysis of glandular saliva.161,162 Cerebrospinal luid ills the ventricles and subarachnoid space of the brain and the spinal cord, while brain extracellular luid occupies the extracellular space within the brain. The brain extracellular luid is derived from cerebral endothelium and is rigidly controlled by the blood–brain barrier, while cerebrospinal luid is derived from the choroid plexus.163 Cerebrospinal luid is characterized by a much lower protein concentration than plasma, and there is an inverse relationship between the hydrodynamic radius of a protein and the concentration in cerebrospinal luid.

to the plasma proteome. These researchers estimate that there are approximately 500 “true” plasma proteins, which is consistent with the estimates of Frank Putnam years ago.28 Schaller and coworkers9 also suggest that the variance in glycosylation and other aspects of posttranslational processing such as the proteolysis observed with factor VIII described in Chapter 6 contribute to the large number of protein species in plasma. The data cited in Table 1.1 are consistent with the suggestion that as analytical techniques improve, the number of protein species identiied in plasma will increase. The number obtained by Schenk and coworkers29 (697) is close to the estimate of Schaller and coworkers6 but far less than that obtained by other researchers. I did ind it interesting that the number estimated by Schaller and colleagues9 and the value obtained by Schenk and coworkers29 are close to that obtained for interstitial luid (525) by Stone and coworkers.30 The workers showed that there were 208 proteins in interstitial luid, which were also found in plasma. The collection of interstitial luid does present considerable technical challenges.31,32 The relationship of interstitial luid and plasma is important, considering the large amounts of “plasma” proteins found in the extracellular luid. It may well be that while certain proteins such as those involved in hemostasis may have a primary function inside the

6

Biotechnology of Plasma Proteins

vascular system to protect the primary functions of oxygen and nutrient transport, other plasma proteins may have more critical roles outside of the vascular bed. Considering the above then, what criteria did I use for inclusion? The major criterion was established intravascular function with demonstrated commercial interest in a therapeutic product. Having said that, there are two outlying proteins— α1-antitrypsin and heparin cofactor 2—where the major function is outside of the vascular bed. In the case of α1-antitrypsin, local application in the lung is the effective therapeutic mode (Chapter 7), not a systemic application, while with heparin cofactor 2, a function is yet to be deined, although it is most likely within the interstitial space. In addition to α1-antitrypsin and heparin cofactor 2, it is most likely that antithrombin and immunoglobulins function in the extravascular space. Prothrombin (and thrombin) are somewhat unique in extravascular function.33,34 There are several proteins that I considered and discarded as candidates for Chapter 9.* These include the following: • Haptoglobin is a glycoprotein in plasma, which is responsible for binding free hemoglobin and returning the hemoglobin (and iron) to the liver to prevent the loss of hemoglobin by urine.35,36 Fucosylated haptoglobin is suggested as a biomarker for pancreatic cancer.37 • α1-Acid glycoprotein (orosomucoid) is an acidic glycoprotein with a mass of approximately 43 kDa.38,39 The function of α1-acid glycoprotein has not been described, but there is a suggestion of immunomodulation.40 α1-Acid glycoprotein is an acute phase protein.41 • Vitronectin, also known as serum spreading factor, is an adhesive protein that is found in both the plasma and the extracellular matrix.42–44 The major function of vitronectin appears to be outside of the vascular system. There are studies on the interaction of vitronectin with urokinase receptor45,46 and plasminogen activator inhibitor-147 as well as suggestion of its importance in bacterial pathogenesis.48 • Ceruloplasmin is a plasma protein responsible for the binding and transport of copper.49,50 While a function other than copper transport has not been identiied, ceruloplasmin has been suggested to have a role in oxidation/ reduction.51 There has been speciic interest in the role of ceruloplasmin in brain function.52,53 Ceruloplasmin has a long history of use in the diagnosis of copper homeostasis such as Wilson’s disease54 and is more recently associated with cardiovascular risk.54,55 • Prekallikrein, also known as Prekallikrein or Fletcher factor, together with high-molecular-weight kininogen, also known as Fitzgerald factor, participates with factor XII and factor XI in the contact phase of blood coagulation.56,57 Prekallikrein has a molecular mass of 88 kDa and a concentration in plasma of approximately 30 mg/L,58 which is the precursor of plasma *

Most of these concepts were thrust upon me in one of my former lives by well-meaning individuals who considered such concepts to be potential commerical products. One learns rapidly that good (and even) great science may not be a good commercial product but a good commercial product does require good science.

Blood, Plasma, Cells, and Other Biological Fluids

kallikrein. Plasma kallikrein catalyzes the formation of kinins from highmolecular-weight kininogen.59 Prekallikrein deiciency is rare and, while associated with a prolonged partial thromboplastin time, is not associated with a bleeding diathesis.60,61 The presence of prekallikrein activator (factor XIIa) in therapeutic plasma fractions can result in hypotension.62–64 It should be noted that plasma kallikrein is an enzyme that is different from tissue kallikrein.65–67 It has been suggested that “oversulfated” heparin/oversulfated chondroitin sulfate68 can “activate” prekallikrein to kallikrein, which then activates prothrombin.69 Other studies have shown that “oversulfated” chondroitin sulfate can result in kinin formation presumably through the activation of prekallikrein.70 • High-molecular-weight kininogen, also known as Fitzgerald, Williams, or Flaujeac factor, is a component of the contact activation system56,57 and a precursor of kinins such as bradykinin.59 High-molecular-weight kininogen has a molecular weight of 120 kDa and is present in plasma at a concentration of 80 mg/L.58 Excess production of kinins from high-molecularweight kininogen can result in hereditary angioenema (deiciency of C1 inhibitor)71 and vasculitis.72 • Transferrin is the plasma protein responsible for the binding and transport of iron, although its function is also suggested in the immune system.73 Transferrin and transferrin receptors are targets for chemotherapy.73–75 Transferrin with ferritin is responsible for the homeostatic regulation of iron,76 and its deiciency can cause clinical issues.77 Deiciency can arise from a variety of causes, including liver disease, but the congenital deiciency is quite rare. The normal level of transferrin in blood is 2–3 gm/L with a half-life of 8 days. Therapeutic applications have been suggested for both the holoprotein and the apoprotein,78 but none have achieved clinical application. The recombinant protein has been produced and is being considered for a range of applications, including atransferrinemia, age-related macular degeneration, ischemia/reperfusion injury, diabetes, and bacterial infection.79 • IGFBPs are a family of proteins that bind and, thus, regulate the activity of IGFs.80–82 There are two forms of IGF, IGF-I and IGF-II, which are bound primarily to IGFBP-3 in plasma, which is associated with a glycoprotein, acid-labile subunit (ALS) yielding a complex with a mass of approximately 150 kDa.82,83 IGFBP can be isolated from the Cohn Fraction IV of human plasma.84 It is likely that IGFBPs have functions other than the regulation of IGF,83 but there is no indication for a therapeutic product. The multiplicity of IGFBPs also confounds the development of a therapeutic product. The various IGFBPs do have somewhat different roles in the regulation of IGFs. IGFBP-3 is responsible for binding the two major forms of IGF in plasma, IGF-I and IGF-II,83 while IFG-I concentration varies widely and has been demonstrated to inhibit IGF function in both in vitro and in vivo studies.85 An additional complication for a therapeutic product is the posttranslational modiication of IGFBP-1, in particular phosphorylation, associated with function.86–90 While an opportunity does not exist for a therapeutic

7

8

Biotechnology of Plasma Proteins

preparation of IGFBP, IGFBP-3,91 IGFBP-2,92 and IGFBP-793 have been suggested as therapeutic targets. • Lipoproteins and apolipoproteins are of great importance in the transport of lipids.94,95 It is important to distinguish between high-density lipoproteins and low-density lipoproteins (HDL and LDL) for determining cardiovascular risk.96–100 There was some interest in the use of lipoproteins for drug delivery,101 and recombinant lipoproteins have been developed for drug delivery as well as for possible therapeutic application in sepsis and atherosclerosis.102 Subsequent work suggested that lipoproteins (HDL or LDL) were of little value in delivering chemotherapeutic drugs to tumor cells in culture.103 More recent work104 has demonstrated the use of a fusion protein between a HDL and a protein transduction domain, TAT,105 to deliver doxirubin into tumor cells. The use of HDL and/or apolipoprotein A-I (the principal protein component of HDL) as a therapeutic approach to atherosclerosis has been suggested.106 Technical approaches suggested by Shah and coworkers106 include enhancing HDL synthesis, decreasing the clearance of apolipoprotein A-I, parenteral administration of apolipoprotein A-I, and peptide mimetic based on apolipoprotein A-I. • β2-Microglobulin is an example of a shed protein as described by Schaller and colleagues,9 as it is not secreted from either a solid tissue such as liver or a lymphoid cell nor is it leaked from damaged cells such as enzymes from cardiac tissue or liver tissue. β2-Microglobulin is present in all nucleated cells, where it is associated with major histocompatibility complex (MHC) class 1 and CD1 complex.107–110 β2-Microglobulin has a molecular mass of approximately 11 kDa and is present in plasma at a low concentration (1–3 mg/L) and can form amyloid in kidney failure. The reader is directed to an excellent review by Heegaard.111 The concentration of β2-microglobulin is elevated in a variety of conditions.112–118 The fractionation of human blood plasma can be considered to be a mature industry, with the basic technology, alcohol fractionation, dating back at least to 70 years. Many of the products described in the current work have been approved biologics for more than 60 years. The challenge that I faced in assembling both the chapters that follow and the brief list mentioned earlier is in distinguishing between proteins in which the major function is considered to occur within the vascular space and those proteins that are transported for primary function at an extravascular site. The distinction between these two categories is not as clear as one would like. Some proteins such as factor VIII, α2-macroglobulin, and the von Willebrand factor appear to function primarily within the vascular system in the absence of disease, while heparin cofactor 2 is considered to function primarily in the extravascular space. Chang and coworkers found less than 1% (plasma level is 100%) factor V, factor VIII, or the von Willebrand factor in synovial luid.119 These researchers did ind substantial amounts of antithrombin (74%) and α2-macroglobuln (13%); prothrombin was present at a concentration of 21%, while factor IX was present at a concentration of 10%. Miller and coworkers120 found substantial amounts of plasma protein in lymph including ibrinogen; ibrinogen antigen was substantially higher

Blood, Plasma, Cells, and Other Biological Fluids

9

than functional activity, and there were substantial amounts of D-dimer. These researchers also observed higher factor VIIa activity in lymph than in plasma; factor VII activity was lower in lymph (35%) than in plasma (110%). Other researchers have reported substantial amounts of ibrinogen in interstitial luids.121–123 Worm reported on the concentration of albumin, transferrin, IgG, and α2-macroglobulin in skin suction blister luid obtained from patients with skin disease compared to a control population.124 The ratio of the concentration of plasma albumin to interstitial luid was lower in the patient population (0.46 vs. 0.55); the ratios of the concentration of IgG (0.39 vs. 0.43), transferrin (0.45 vs. 0.48), and α2-macroglobulin (0.22 vs. 0.21) were closer to the control group. Similar results were observed by other researchers.125 Thrombin/thrombin-like activity has been demonstrated to function in the extravascular space,126–128 but the synthesis of the prothrombin has also been demonstrated in the extravascular space,129 suggesting that extravascular synthesis of prothrombin may be a signiicant factor, as well as extravasation.130 Jacob and coworkers130 suggest in their work on protein C that there is physiological heterogeneity of the vascular barrier where a particular plasma protein will be present at a discrete vascular site. Local differences in vascular permeability have been discussed previously with respect to insulin15 and local accumulation of IgG at focal sites of inlammation.16 Clearly, vascular permeability is a complex area with a variety of mechanisms.131 There is also an active extravascular ibrinolytic system,132 which is discussed in Chapters 7 and 9. López and Nowak133 suggested that a small derivative of hirudin, dipetarudin, showed rapid distribution over the extravascular space where it could inhibit thrombin; thrombin has been suggested to promote tumor growth.134 In subsequent work, López and coworkers modiied dipetarudin with polyethylene glycol.135 The monosubstituted derivative had pharmacokinetics similar to that of the unmodiied dipetarudin, while the disubstituted derivative demonstrated much slower clearance to the extravascular compartment. The aforementioned information suggests that, at least with our current information, there are some plasma proteins such as factor VIII and the von Willebrand factor (Chapter 6) in which the intravascular function is clear. Many, if not all, of the other classical plasma proteins appear to have signiicant functions in the extravascular space, and such functions may have regional speciicity. It is clear that more work is needed in this area to understand the factors inluencing the distribution between intravascular space and extravascular space. As shown by the dipetarudin results cited earlier,135 PEGylation may have a major inluence on the distribution over the extravascular space in addition to the effects on renal clearance as part of its effect on pharmacokinetics. Human plasma has been a valuable source of biological products for the past 70 years, and the information gathered from the development of plasma proteins has proved vital to the development of recombinant therapeutic proteins. Some of the plasma-derived therapeutics such as factor VIII and factor IX have had clear paths to clinical success, while others, most notably the serpins, have had a more dificult path to clinical success. It is my opinion that the concept of biological degeneracy, as elaborated by Gerry Edelman and Joe Gally,136 can provide insight into why the application of the therapeutics has been so dificult. The modulation of plasminogen activator inhibitors can be a useful target for RNAi therapeutics.137,138

10

Biotechnology of Plasma Proteins

REFERENCES 1. Rader, R.A., (Re)deining biopharmaceutical, Nature Biotechnol. 26, 743–751, 2008. 2. Lundblad, R.L., Development and Application of Biomarkers, CRC Press/Taylor & Francis, Boca Raton, FL, 2011. 3. Simpson, R.J. (ed.), Serum/Plasma Proteomics Methods and Protocols, Humana Press/ Springer, New York, 2011. 4. Surinova, S., Schiess, R., Hüttenhain, R., et al., On the development of plasma protein biomarkers, J. Proteome Res. 10, 5–16, 2011. 5. Berne, R.M., Levy, M.N., Keoppen, B.M., and Stanton, B.A. (eds.), Physiology, 4th edn., Blood, Chapter 20, pp. 315–324, Mosby, St.Louis, MO, 1998. 6. Berne, R.M., Levy, M.N., Keoppen, B.M., and Stanton, B.A. (eds.), Physiology, 5th edn., Blood, Chapter 20, pp. 315–324, Mosby, St.Louis, MO, 2004. 7. Costanzo, L.S., Physiology, Saunders/Elsevier, Philadelphia, 2006. 8. Sircar, S., Principles of Medical Physiology, Thieme, Stuttgart, Germany, 2008. 9. Schaller, J., Gerber, S., Kämpfer, U., Lejon, S., and Trachsel, O., Human Plasma Protein Structure and Function, John Wiley & Sons, Hoboken, NJ, 2008. 10. Altman, P.L. and Dittmer, D.S., Blood and Other Body Fluids, Federation of American Societies for Experimental Biology, Washington, DC, 1961. 11. Johanson, C.E., Stopa, E.G., and McMillan, P.N., The blood–cerebrospinal luid barrier: Structure and functional signiicance, Meth. Mol. Biol. 686, 101–131, 2011. 12. Poulson, H.L., Interstitial luid concentrations of albumin and immunoglobulin G in normal man, Scand. J. Clin. Lab. Invest. 34, 119–122, 1974. 13. Wittels, P., Gunga, H.C., Kirsch, K., et al., Fluid regulation during prolonged physical strain with water and food deprivation in healthy, trained men, Weiner Klin. Wochenschr. 108, 788–794, 1996. 14. Mack, G.W., Yang, R., Hargens, A.R., et al., Inluence of hydrostatic pressure gradient on regulation of plasma volume after exercise, J. Appl. Physiol. 85, 667–675, 1998. 15. Bar, R.S., Boes, M., Clemmons, D.R., et al., Insulin differentially alters transcapillary movement of intravascular IGFBP-1, IGFBP-2 and endothelial cell IGF-binding proteins in the rat heart, Endocrinology 127, 497–499, 1990. 16. Juweld, M., Strauss, H.W., Yaoita, H., et al., Accumulation of immunoglobulin G at focal sites of inlammation, Eur. J. Nucl. Med. 19, 159–165, 1992. 17. Reed, R.K. and Rubin, K., Transcapillary exchange: Role and importance of the interstitial luid pressure the extracellular matrix, Cardiovasc. Res. 87, 211–217, 2010. 18. Parving, H.H., Nielsen, S.L., and Lassen, N.A., Increased transcapillary escape rate of albumin, IgG, and IgM during angiotensin-II-induced hypertension in man, Scand. J. Clin. Lab. Invest. 34, 111–118, 1974. 19. Mehta, D. and Malik, A.B., Signaling mechanisms regulating endothelial permeability, Physiol. Rev. 86, 279–367, 2006. 20. Komarova, Y. and Malik, A.B., Regulation of endothelial permeability via paracellular and transcellular pathways, Annu. Rev. Physiol. 72, 463–493, 2010. 21. Poulsen, H.L., Jensen, H.Æ., and Parving, H.-H., Extracellular luid volume determined by a single injection of inulin in men with untreated essential hypertension, Scand. J. Clin. Lab. Invest. 37, 691–697, 1977. 22. Harper, H.A., Review of Physiological Chemistry, 15th edn., Water and metabolism, Chapter 19, p. 422, Lange Medical Publishers, Los Altos, CA, 1975. 23. Holliday, M.A., Extracellular luid and its proteins: Dehydration, shock, and recovery, Pediatr. Nephrol. 13, 989–995, 1999. 24. Dancik, Y., Anissimov, Y.G., Jepps, O.G., and Roberts, M.S., Convective transport of highly plasma protein bound drugs facilitates direct penetration into deep tissues after topical application, Br. J. Clin. Pharmacol. 73, 564–578, 2012.

Blood, Plasma, Cells, and Other Biological Fluids

11

25. Howard, M.L., Hill, J.J., Galluppi, G.R., and McLean, M.A., Plasma protein binding in drug discovery and development, Comb. Chem. High Throughput Screen. 13, 170–187, 2010. 26. Liu, X., Chen, C., and Hop, C.E., Do we need to optimize plasma protein and tissue binding in drug discovery?, Curr. Top. Med. Chem. 11, 450–466, 2011. 27. Schwick, H.-G. and Haupt, H., Chemistry and function of human plasma proteins, Angew. Chem. Int. Ed. 19, 87–99, 1980. 28. Putnam, F.W., Alpha, beta, gamma, omega—The Structure of the plasma proteins, in The Plasma Proteins. 2nd edn., ed. F.W. Putnam, Volume IV, Chapter 2, pp. 45–166, Academic Press, New York, 1984. 29. Schenk, S., Schoenhals, G.J., de Souza, G., and Mann, M., A high conidence, manually validated human protein reference set, BMC Med. Genomics 1, 41, 2008. 30. Stone, M.D., Odland, R.M., McGowan, T., et al., Novel in situ collection of tumor interstitial luid from a head and neck squamous carcinoma reveals a unique proteome with diagnostic potential, Clin. Proteomics 6, 75–82, 2010. 31. Olszeski, W.L., Collection and physiological measurements of peripheral lymph and interstitial luid in man, Lymphology 10, 137–145, 1977. 32. Sloop, C.H., Dory, L., and Roheim, P.S., Interstitial luid lipoproteins, J. Lipid Res. 28, 225–237, 1987. 33. Yamazaki, Y., Shikamoto, Y., Fukudome, K., et al., Fibroblasts, glial, and neuronal cells are involved in extravascular prothrombin activation, J. Biochem. 126, 655–661, 1999. 34. Conzen, P. and Becker, B.F., Perspectives in microvascular luid handling: Does the distribution of coagulation factors in human myocardium comply with plasma extravasation in venular coronary segments?, J. Vasc. Res. 48, 219–226, 2011. 35. Wassell, J., Haptoglobin: Function and polymorphism, Clin. Lab. 46, 547–552, 2000. 36. Levy, A.P., Asleh, R., Blum, S., et al., Haptoglobin: Basic and clinical aspects, Antioxid. Redox. Signal. 12, 293–304, 2010. 37. Miyoshi, E., Shinzaki, S., Moriwaki, K., and Matsumoto, H., Identiication of fucosylated haptoglobin as a novel tumor marker for pancreatic cancer and its possible application for a clinical diagnostic test, Meth. Enzymol. 478, 153–164, 2010. 38. Fournier, T., Medjoubi-N, N., and Porquet, D., Alpha-1-acid glycoprotein, Biochim. Biophys. Acta 1482, 157–171, 2000. 39. Hochepied, T., Berger, F.G., Baumann, H., and Libert, C., α1-Acid glycoprotein: An acute phase protein with inlammatory and immunomodulating properties, Cytokine Growth Factor Rev. 14, 25–34, 2003. 40. Miranda-Ribera, A., Lecchi, C., Bronzo, V., et al., Down-regulation effect of alpha1-acid glycoprotein on bovine neutrophil degranulation, Comp. Immunol. Microbiol. Infect. Dis. 33, 291–306, 2010. 41. Zsila, F., Chaperone-like activity of the acute-phase component human serum alpha-1acid glycoproein: Inhibition of thermal- and chemical-induced aggregation of various proteins, Bioorg. Med. Chem. Lett. 20, 1205–1209, 2010. 42. Preissner, K.T. and Seiffert, D., Role of vitronectin and its receptors in haemostasis and vascular remodeling, Thromb. Res. 89, 1–21, 1998. 43. Schvartz, I., Seger, D., and Shaltiel, S., Vitronectin, Int. J. Biochem. Cell Biol. 31, 539– 544, 1999. 44. Wechsler-Reya, R.J., Caught in the matrix: How vitronectin controls neuronal differentiation, Trends Neurosci. 24, 680–682, 2001. 45. Madsen, C.D. and Sidenius, N., The interaction between urokinase receptor and vitronectin in cell adhesion and signalling, Eur. J. Cell Biol. 87, 617–629, 2008. 46. Huai, Q., Zhou, A., Lin, L., et al., Crystal structures of two human vitronectin, urokinase and urokinase receptor complexes, Nat. Struct. Mol. Biol. 15, 422–423, 2008.

12

Biotechnology of Plasma Proteins

47. Konstantinides, S., Schäfer, K., and Loskutoff, D.J., Do PAI-1 and vitronectin promote or inhibit neointima formation? The exact role of the ibrinolytic system in vascular remodeling remains uncertain, Arterioscler. Thromb. Vasc. Biol. 22, 1943–1945, 2002. 48. Singh, B., Su, Y.C., and Riesbeck, K., Vitronectin in bacterial pathogenesis: A host protein used in complement escape and cellular invasion, Mol. Microbiol. 78, 545–560, 2010. 49. Hellman, N.E. and Gitlin, J.D., Ceruloplasmin metabolism and function, Annu. Rev. Nutr. 22, 439–458, 2002. 50. Healy, J. and Tipton, K., Ceruloplasmin and what it might do, J. Neural Transm. 114, 777–781, 2007. 51. Burkitt, M.J., A critical overview of the chemistry of copper-dependent low density lipoprotein oxidation: Roles of lipid hydroperoxides, α-tocopherol, thiols, and ceruloplasmin, Arch. Biochem. Biophys. 394, 117–135, 2001. 52. Qian, Z.M. and Ke, Y., Rethinking the role of ceruloplasmin in brain iron metabolism, Brain Res. Rev. 35, 287–294, 2001. 53. Vassiliev, V., Harris, Z.L., and Zatta, P., Ceruloplasmin in neurodegenerative diseases, Brain Res. Rev. 49, 633–640, 2005. 54. Mak, C.M. and Lam, C.W., Diagnosis of Wilson’s disease: A comprehensive review, Crit. Rev. Clin. Lab. Sci. 45, 263–290, 2008. 55. Tang, W.H., Wu. Y., Hartiala, J., et al., Clinical and genetic association of serum ceruloplasmin with cardiovascular risk, Arterioscler. Thromb. Vasc. Biol. 32, 516–522, 2012. 56. Schmaier, A.H., Assembly, activation, and physiologic inluence of the plasma kallikrein/ kinin system, Int. Innunopharmacol. 8, 161–165, 2008. 57. Saito, H., Studies on Fletcher trait and Fitzgerald trait. A rare chance to disclose body’s defense reactions against injury, Thromb. Haemost. 104, 867–874, 2010. 58. Vogler, E.A. and Siedlecki, C.A., Contact activation of blood–plasma coagulation, Biomaterials 30, 1857–1869, 2009. 59. Coleman, R.W. and Bagdasarian, A., Human kallikrein and prekallikrein, Methods 45, 303–327, 1976. 60. Girolami, A., Scarparo, P., Candeo, N., and Lombardi, A.M., Congenital prekallikrein deiciency, Expert Rev. Hematol. 3, 685–695, 2010. 61. Nakao, T., Yamane, T., Katagami, T., et al., Severe prekallikrein deiciency due to a homozygous Trp499stop nonsense mutation, Blood Coagul. Fibrinolysis 22, 337–339, 2011. 62. Alving, B.M., Hojima, Y., Pisano, J.J., et al., Hypotension associated with prekallikrein activator (Hageman-Factor fragments) in plasma protein fractions, N. Engl. J. Med. 299, 66–76, 1978. 63. Snape, T.J., Grifith, D., Vallet, L., and Wesley, E.D., The assay of prekallikrein activator in human blood products, Develop. Biol. Standard. 44, 115–120, 1979. 64. Georgakopoulos, T., Bertolini, J., and Hayes, T.K., The effect of salt concentration on the prekallikrein activator assay, Biologicals 38, 178–179, 2010. 65. Colman, R.W., Plasma and tissue kallikrein in arthritis and inlammatory bowel disease, Immunopharmacology 43, 103–108, 1999. 66. Lima, A.R., Alves, F.M., Angelo, P.F., et al., S1’ and S2’ subsite speciicities of human plasma kallikrein and tissue kallikrein 1 for the hydrolysis of peptides derived from the bradykinin domain of human kininogen, Biol. Chem. 389, 1487– 1494, 2008. 67. Feener, E.P., Plasma kallikrein and diabetic macular edema, Curr. Diab. Rep. 10, 270– 275, 2010. 68. Beni, S., Limtiaco, J.F., and Larive, C.K., Analysis and characterization of heparin impurities, Anal. Bioanal. Chem. 399, 527–539, 2011.

Blood, Plasma, Cells, and Other Biological Fluids

13

69. Qian, Y., Pan, J., Zhou, X., et al., Oversulfated heparin by-products induce thrombin generation in human plasmas through contact system activation, Clin. Appl. Thromb. Hemost. 16, 244–250, 2010. 70. Adam, A., Montpas, N., Keire, D., et al., Bradykinin forming capacity of oversulfated chondroitin sulfate contaminated heparin in vitro, Biomaterials 31, 5741–5748, 2010. 71. Kaplan, A.P., Enzymatic pathways in the pathogenesis of hereditary angioedema: The role of C1 inhibitor therapy, J. Allergy Clin. Immunol. 126, 918–925, 2010. 72. Kahn, R. and Karpman, D., Kinin system activation in vasculitis, Acta Paediatr. 100, 950–957, 2011. 73. Macedo, M.R. and de Sousa, M., Transferrin and the transferrin receptor of magic bullets and other concerns, Inlamm. Allergy Drug Targets 7, 41–52, 2008. 74. Daniels, T.F., Delgado, T., Rodriguez, J.A., et al., The transferrin receptor part I: Biology and targeting with cytotoxic antibodies for the treatment of cancer, Clin. Immunol. 121, 144–158, 2006. 75. Daniels, T.F., Delgado, T., Helguera, G., and Penichet, M.L., The transferrin receptor part II: Targeted delivery of therapeutic agents into cancer cells, Clin. Immunol. 121, 159–176, 2006. 76. Muñoz, M., Garcia-Erce, J.A., and Remache, A.F., Disorders of iron metabolism. Part 1: Molecular basis of iron homeostasis, J. Clin. Pathol. 64, 281–286, 2011. 77. Harju, E., Clinical pharmacokinetics of iron preparations, Clin. Pharmacokinet. 17, 69–89, 1989. 78. Gomme, P.T. and McCann, K.B., Transferrin: Structure, function, and potential therapeutic actions, Drug Discov. Today 10, 267–273, 2005. 79. Brandsma, M.E., Jevnikar, A.M., and Ma, S., Recombinant human transferrin: Beyond iron binding and transport, Biotechnol. Adv. 29, 230–238, 2011. 80. Chan, K. and Spencer, E.M., General aspects of insulin-like growth factor binding proteins, Endocrine 7, 95–97, 1997. 81. Collett-Solberg, P.F. and Cohen, P., Genetics, chemistry, and function of the IGF/IGFBP system, Endocrine 12, 121–136, 2000. 82. Kelly, K.M., Schmidt, K.E., Berg, L., et al., Comparative endocrinology of the insulinlike growth factor binding protein, J. Endocrinol. 173, 3–18, 2002. 83. Holly, J. and Perks, C., The role of insulin-like growth factor binding proteins, Neuroendocrinology 83, 154–160, 2006. 84. Martin, J.L. and Baxter, R.C., Insulin-like growth factor-binding protein from human plasma. Puriication and characterization, J. Biol. Chem. 261, 8754–8760, 1986. 85. Lee, P.D., Giudice, L.C., Conover, C.A., and Powell, D.R., Insulin-like growth factor binding protein-1: Recent indings and new directions. Proc. Soc. Exp. Biol. Med. 216, 319–357, 1997. 86. Frost, R.A., Bereket, A., Wilson, T.A., et al., Phosphorylation of insulin-like growth factor binding protein in patients with insulin-dependent diabetes meillitus and severe trauma, J. Clin. Endocrinol. Metab. 78, 1533–1535, 1978. 87. Wojnar, M.M., Fan, J., Frost, R.A., et al., Alterations in the insulin-like growth factor system in trauma patients, Am. J. Physiol. 268, R970–R977, 1995. 88. Sakai, K., Busby, W.H., Jr., Clarke, J.B., and Clemmons, D.R., Tissue transglutaminase facilitates and the polymerization of insulin-like growth factor-binding protein-1 (IGFBP-1) and leads to loss of IGFBP-1’s ability to inhibit insulin-like growth factor-Istimulated protein synthesis, J. Biol. Chem. 276, 8740–8745, 2001. 89. Kabir-Salmani, M., Shimizu, Y., Sakai, K., and Iwashita, M., Post translational modiications of decidual IGFBP-1 by steroid hormones in vitro, Mol. Hum. Reprod. 11, 667–671, 2005.

14

Biotechnology of Plasma Proteins

90. Dolcini, L., Sala, A., Campagnoli, M., et al., Identiication of the amniotic luid insulinlike growth factor binding protein-1 phosphorylation sites and propensity to proteolysis of the isoforms, FEBS J. 276, 6033–6046, 2009. 91. Cheung, C.W., Taylor, P.J., Kirkpatrick, C.M., et al., Therapeutic value of orally administered silibinin in renal cell carcinoma: Manipulation of insulin-like growth factor binding protein-3 levels, BJU Int. 100, 438–444, 2007. 92. So, A.I., Levitt, R.J., Eigl, B., et al., Insulin-like growth factor binding protein-2 is a novel therapeutic target associated with breast cancer, Clin. Cancer Res. 14, 6944–6954, 2008. 93. Nousbeck, J., Ishida-Yamamoto, A., Bidder, M., et al., IGFBP7 as a potential therapeutic target in psoriasis, J. Invest. Dermatol. 131, 1767–1770, 2011. 94. Smith, L.C., Pownall, H.J., and Gotto, A.M., Jr., The plasma lipoproteins: Structure and metabolism, Annu. Rev. Biochem. 47, 731–777, 1978. 95. Fredrickson, D.S., Plasma lipoproteins and apolipoproteins, Harvey Lect. 68, 185–237, 1974. 96. Goldstein, J.L. and Brown, M.S., The low-density lipoprotein pathway and its relation to atherosclerosis, Annu. Rev. Biochem. 46, 897–930, 1977. 97. Scanu, A.M., Plasma lipoproteins and coronary heart disease, Ann. Clin. Lab. Sci. 8, 79–83, 1978. 98. Levi, R.I. and Rifkin, B.M., The structure, function and metabolism of high-density lipoproteins: A status report, Circulation 62, IV4–IV8, 1980. 99. Sniderman, A., McQueen, M., Contois, J., et al., Why is non-high-density lipoprotein cholesterol a better marker of the risk of vascular disease than low-density lipoprotein cholesterol?, J. Clin. Lipidol. 4, 152–155, 2010. 100. Lund-Katz, S. and Phillips, M.C., High density lipoprotein structure–function and role in reverse cholesterol transport, Subcell. Biochem. 51, 183–227, 2010. 101. van Berkel, Th.J.C., Drug targeting: Application of endogenous carriers for site-speciic delivery of drugs, J. Control. Release 24, 145–155, 1993. 102. Rensen, P.C.N., de Vruch, R.L.A., Kuiper, J., et al., Recombinant lipoproteins: Lipoproteinlike lipid particles for drug targeting, Adv. Drug Deliv. Rev. 47, 251–276, 2001. 103. Kader, A. and Pater, A., Loading anticancer drugs into HDL as well as LDL has little effect on properties of complexes and enhances cytotoxicity to human carcinoma cells, J. Control. Release 80, 29–44, 2002. 104. Murakami, T., Wijagkanalan, W., Hashida, M., and Tsuchida, K., Intracellular drug delivery by genetically engineered high-density lipoprotein nanoparticles, Nanomedicine 5, 867–879, 2010. 105. Musacchio, T. and Torchilin, V.P., Recent development in lipid-based pharmaceutical nanocarriers, Front. Biosci. 16, 1388–1412, 2011. 106. Shah, P.K., Kaul, S., Nilsson, J., and Cercek, B., Exploiting the vascular protective effects of high-density lipoprotein and its apolipoprotein. An idea whose time for testing is coming, Part II, Circulation 104, 2498–2502, 2001. 107. Groves, M.L. and Greenberg, R., β2-Microglobulin and its relationship to the immune system, J. Dairy Sci. 65, 317–325, 1982. 108. Tatake, R.J., Ferrone, S., and Zeff, R.A., The role of beta-2 microglobulin in temperature-sensitive and interferon-gamma-induced exocytosis of HLA class I molecules, Transplantation 54, 395–403, 1992. 109. Rammensee, H.G., Falk, K., and Rötzschke, O., MHC molecules as peptide receptors, Curr. Opin. Immunol. 5, 35–44, 1993. 110. Ohta, Y., Shiina, T., Lohr, R.L., et al., Primordial linkage of β2-microglobulin to the MHC, J. Immunol. 186, 3563–3571, 2011. 111. Heegaard, N.H.N., β2-Microglobulin: From physiology to amyloidosis, Amyloid 16, 151–173, 2009.

Blood, Plasma, Cells, and Other Biological Fluids

15

112. Matos, A.C., Durão, M.S., Jr., and Pacheco-Silva, A., Serial beta-2-microglobulin measurement as an auxilliary method in the eaerly diagnosis of cytomegalovirus infection in renal transplant patients, Transplant. Proc. 36, 894–895, 2004. 113. Gross, M., Top, I., Laux, I., et al., Beta-2-microglobulin is an androgen-regulated secreted protein elevated in serum of patients with prostate cancer, Clin. Cancer Res. 13, 1979–1986, 2007. 114. Hofstra, J.M., Deegens, J.K., Willems, H.L, and Wetzels, J.F., Beta-2-microglobulin is superior to N-acetyl-β-glucosamidase in predicting prognosis in idiopathic membranous nephropathy, Nephrol. Dial. Transplant. 23, 2546–2551, 2008. 115. Kim, H.A., Jeon, J.Y., Yoon, J.M., and Suh, C.H., Beta-2-microglobulin can be a disease activity marker in systemic lupus erythematosus, Am. J. Med. Sci. 339, 337–340, 2010. 116. Mink, S.R., Hodge, A., Agus, D.B., et al., Beta-2-microglobulin expression correlates with high-grade prostate cancer and speciic defects in androgen signaling, Prostate 70, 1201–1210, 2010. 117. Amighi, J., Hoke, M., Miekusch, W., et al., Beta-2-microglobulin and the risk for cardiovascular events in patients with asymptomatic carotid atherosclerosis, Stroke 42, 1826–1833, 2011. 118. Annweiler, C., Bataille, R., Ferrière, N., et al., Plasma beta-2-microglobulin as a marker of frality in older adults: A pilot study, J. Gerontol. A. Biol. Sci. Med. Sci. 66A, 1077– 1079, 2011. 119. Chang, P., Aronson, D.L., Borenstein, D.G., and Kessler, C.M., Coagulant proteins and thrombin generation in synovial luid: A model for extravascular coagulation, Am. J. Hematol. 50, 79–83, 1995. 120. Miller, G.J., Howarth, D.J., Attield, J.C., et al., Haemostatic factors in human peripheral afferent lymph, Thromb. Haemost. 83, 427–432, 2000. 121. Reeve, E.B., Stephens, A., and Carlson, T.H., Experimenal studies of the ibrinogen response to hemorrhage, Am. J. Physiol. 237, H504–H513, 1979. 122. Hedin, A. and Hahn, R.G., Voume expansion and plasma protein clearance during intravenous infusion of 5% albumin and autologous plasma, Clin. Sci. 108, 217–224, 2005. 123. Mendez, I.Z.R., Shi, Y., HogenEsch, H., and Hem, S.L., Potentiation of the immune response to non-adsorbed antigens by aluminum-containing adjuvants, Vaccine 25, 825– 833, 2007. 124. Worm, A.-M., Exchange of macromolecules between plasma and skin interstitium in extensive skin disease, J. Invest. Dermatol. 76, 489–492, 1981. 125. Anvar, M.D., Khiabani, H.X., Lande, K., et al., The concentration of protein-compounds in interstitial tissue of patients with chronic critical limb ischemia and oedema, Vasa J. Vasc. Dis. 30, 14, 2001. 126. Xi, G., Reiser, G., and Keep, R.F., The role of thrombin and thrombin receptors in ischemic, hemorrhagic and traumatic brain injury: Deleterious or protective?, J. Neurochem. 84, 3–9, 2003. 127. Naldini, A., Morena, E., Belotti, D., et al., Identiication of thrombin-like activity in ovarian cancer associated ascites and modulation of multiple cytokine networks, Thromb. Haemost. 106, 705–711, 2011. 128. Yamazaki, Y., Shikamoto, Y., Fukudome, K., et al., Fibroblasts, glial, and neuronal cells are involved in extravascular prothrombin activation, J. Biochem. 126, 655–661, 1999. 129. Dihanich, M., Kaser, M., Reinhard, E., et al., Prothrombin mRNA is expressed by cells of the nervous system, Neuron 6, 575–581, 1991. 130. Jacob, M., Chappel, D., Stoeckelhuber, M., et al., Perspectives in microvascular luid handling: Does the distribution of coagulation factors in human myocardium comply with plasma extravasation in venular coronary segments?, J. Vasc. Res. 48, 219–226, 2011.

16

Biotechnology of Plasma Proteins

131. Dvorak, H.F., Vascular permeability to plasma, plasma proteins, and cells: An update, Curr. Opin. Hematol. 17, 225–229, 2010. 132. Chung, S.I., Lee, S.Y., Uchino, R., and Carmassi, F., Factors that control extravascular ibrinolysis, Semin. Thromb. Hemost. 22, 479–488, 1996. 133. López, M.L. and Nowak, G., Special pharmacokinetics of dipetarudin suggests a potential antitumor activity of this thrombin inhibitor, Anti-Cancer Drugs 15, 145–149, 2004. 134. Zigler, M., Kamiya, T., Brantley, E.C., et al., PAR-1 and thrombin: The ties that bind the microenvironment to melanoma metastasis, Cancer Res. 71, 6561–6566, 2011. 135. López, M., Koehler, A., and Nowak, G., Biochemical and pharmacokinetic characterization of two PEGylated variants of dipetarudin, Thromb. Haemost. 102, 454–459, 2009. 136. Edelman, G.M. and Gally, J.A., Degeneracy and complexity in biological systems, Proc. Natl. Acad. Sci. USA 98, 13763–13768, 2001. 137. Chen, D., Lin, Q., Box, N., et al., SKI knockdown inhibits human melaonoma tumor growth in vivo, Pigment Cell Melanoma Res. 22, 761–772, 2009. 138. Nishioka, N., Matsuoka, T., Yashiro, M., et al., Plasminogen activator inhibitor-1 RNA interference suppresses gastric cancer metastasis in vivo, Cancer Sci. 103, 228–232, 2012. 139. Shen, Y., Kim, J., Strittmatter, E.F., et al., Characterization of the human blood plasma proteome, Proteomics 5, 4034–4045, 2005. 140. Liu, T., Qian, W.-J., Gritsenko, M.A., et al., High dynamic range characterization of the trauma patient plasma proteome, Mol. Cell. Proteomics 5, 1899–1913, 2006. 141. Liu, X., Valentine, S.J., Plasencia, M.D., et al., Mapping the human plasma proteome by SCX-LC-IMS-MS, J. Am. Soc. Mass Spectrom. 18, 1249–1264, 2007. 142. Garbett, N.C., Miller, J.J., Jenson, A.B., and Chaires, J.B., Calorimetric analysis of the plasma proteome, Sem. Nephrol. 27, 621–626, 2007. 143. Loo, J.A., Yan, W., Ramachandran, P., and Wong, D.T., Comparative human salivary and plasma proteome, J. Dent. Res. 89, 1016–1023, 2010. 144. Anderson, N.L., Polanski, M., Pieper, R., et al., The human plasma proteome, Mol. Cell. Proteomics 3, 311–326, 2004. 145. Farrah, T., Deutsch, E.W., Oemnn, G.S., et al., A high-conidence human plasma proteome reference set with estimated concentrations in PeptideAtlas, Mol. Cell. Proteomics 10:M100.006353, 2011. 146. Adachi, J., Kumar, C., Zhang, Y.L., et al., The human urinary proteome contains more than 1500 including a large proportion of membrane proteins, Genome Biol. 7, R80, 2006. 147. Wiley, D.W. and Merchant, M.L., Proteomic methods for biomarker discovery in urine, Sem. Nephrol. 27, 584–596, 2007. 148. Cho, C.-K.J., Shan, S.J., Winsor, E.J., and Diamandis, E.P., Proteomic analysis of human amniotic luid, Mol. Cell. Proteomics 6, 1406–1415, 2007. 149. Dasari, S., Pereira, L., Reddy, A.P., et al., Comprehensive proteomic analysis of human cervical–vaginal luid, J. Proteome Res. 6, 1258–1268, 2007. 150. Lysaght, A.C., Kao, S.Y., Paulo, J.A., et al., Proteome of human perilymph, J. Proteome Res. 10, 3845–3851, 2011. 151. Gobezie, R., Kho, A., Krastins, B., et al., High abundance synovial luid proteome: Distinct proiles in health and osteoarthritis, Arthritis Res. Ther. 9, R26, 2007. 152. Wenner, B.R., Lovell, M.A., and Lynn, B.C., Proteomic analysis of human ventricular cerebrospinal luid from neurologically normal, elderly subjects using two-dimensional LC-MS/MS, J. Proteome Res. 3, 97–103, 2004. 153. Zheng, J., Goodlett, D.R., Peskind, E.R., et al., Quantitative proteomic analysis of age-related changes in human cerebrospinal luid, Neurobiol. Aging 26, 207–227, 2005.

Blood, Plasma, Cells, and Other Biological Fluids

17

154. Pan, S., Zhu, D., Quinn, J.F., et al., A combined dataset of human cerebrospinal luid proteins identiied by multi-dimensional chromatography and tandem mass spectrometry, Proteomics 7, 469–473, 2007. 155. Schutzer, S.E., Liu, T., Natelson, B.H., et al., Establishing the proteome of normal human cerebrospinal luid, PLoS One 5, e10980, 2010. 156. Maurer, M.H., Proteomics of brain extracellular luid (ECF) and cerebrospinal luid (CSF), Mass Spectrom. Rev. 29, 17–28, 2010. 157. Yadev, A.K., Bhardwaj, G., Basak, T., et al. A systematic analysis of eluted fractions of plasma post immunoafinity depletion: Implications in biomarker discovery, PLOS ONE 6, e24442, 2011. 158. Lee, D.H., Kim, J.W., Jeon, S.Y., et al., Proteomic analysis of the effect of storage temperature on human serum, Ann. Clin. Lab. Sci. 40, 61–70, 2010. 159. Kim, H.J., Kim, M.R., So, E.J., and Kim, C.W., Comparison of proteomes in various human plasma preparations by two-dimensional gel electrophoresis, J. Biochem. Biophys. Meth. 70, 619–625, 2007. 160. Cooper, S., Lange, I., Wood, S., et al., Diagnostic accuracy of rapid phIGFBP-1 or predicting preterm labor in symptomatic patients, J. Perinatol. (in press). 161. Walz, A., Stühler, K., Wattenberg, A., et al., Proteome analysis of glandular parotid and submandibular–sublingual saliva in comparison to whole human saliva by twodimensional gel electrophoresis, Proteomics 6, 1631–1639, 2006. 162. Quintana, M., Palicki, O., Lucchi, G., et al., Inter-individual variability of protein patterns in saliva of healthy adults, J. Proteomics 72, 822–830, 2009. 163. Segal, M.B., Extracellular and cerebrospinal luids, J. Inher. Metab. Dis. 16, 617–638, 1993.

2

Plasma Fractionation and Plasma Protein Products

The various plasma-derived proteins described in the current work are frequently included in the ield of transfusion medicine.1–7 It is also clear that the development of blood collection and storage method was essential to the provision of plasma for fractionation. It is useful then to briely consider the history of transfusion medicine as a preamble to plasma fractionation and plasma protein products. The late Bernard Ficarra compiled an excellent collection of essays on the history of medicine,8 including one chapter on the history of transfusion medicine.9 Ficarra noted both the historical importance of blood in human sacriice and the custom of oral blood consumption as a therapeutic. Considering the current interest in the oral delivery of protein therapeutics,10,11 the oral consumption of blood is an early example of this technology. The oral administration of protein therapeutics for hemophilia was described in the 1930s.12,13 Early work14 on the clinical characterization of the Cohn fractionation products showed that oral albumin was not as effective as intravenous albumin in maintaining the nitrogen balance in human subjects. While there were earlier attempts at transfusion such as that described for Pope Innocent VIII in 1492, Ficarra9 described the seminal role of Francis Felli in the discovery of blood transfusion in 1654 followed by the demonstration of the effectiveness of venipuncture using a quill by Christopher Wren in 1656. Notwithstanding these early observations, James Blondell, an English physician, is usually given the credit for the irst successful blood transfusion in 1818 for the treatment of an obstetrical hemorrhage. The irst use of blood transfusion for the treatment of hemophilia was reported by Lane in 1840.15 The second half of the nineteenth century saw considerable advances in the use of blood transfusion in a variety of clinical situations with a remarkable number of positive outcomes. The great majority of the early work on blood transfusion was performed in England and has been the subject of an excellent review by McLaughlin.16 This early work showed that the transfer of blood from a healthy donor to a patient had therapeutic value, but there were major technical obstacles to the general use of transfusion medicine. Early work had deined the species speciicity of blood transfusion, but blood serotyping in human subjects had to wait for development until 1907.17 In addition to the blood type compatibility issue, there were two other major technical problems that required resolution: the coagulation (or fermentation) of blood, which meant that the donor and the recipient could not be separated in space (location), and the lability of the blood, which meant that it was dificult to separate the donor and the recipient in time. Blood clots when it is removed from the circulatory system, and it is suggested that the endothelial lining of the vascular provides a nonthrombogenic surface. An immense amount of effort has been expended over 19

20

Biotechnology of Plasma Proteins

the past 50 years in an attempt to develop a nonthrombogenic surface with limited success.18,19 The porcelain and metal containers that were used in the 1800s did not allow the collection of blood without the formation of a clot. Early attempts to preserve blood in a liquid state involved using the method of deibrillation (deibrination; frequently, a wire whip not unlike those used in cooking was used) of blood to remove ibrin.16 While deibrillation was somewhat effective, there was considerable use of tubes and other devices for physically connecting a donor and a recipient. There was some use of parafin-coated containers for the collection of blood for transfusion,20 which allowed the limited separation of a donor and a recipient. The use of phosphate to prevent blood coagulation was introduced by Hicks in 1869.21 Phosphate salts were used for some time as an anticoagulant without an understanding of the mechanism, as it was not until sometime later that the role of calcium ions in blood coagulation was suggested by several groups,22,23 and even later when citrate was observed to serve as an anticoagulant,24 permitting the physical separation of a donor and a recipient. The use of citrate as an anticoagulant was extremely useful under battleield conditions in World War I,25 most likely resulting in the irst transfusion service. Robertson did state that parafin coating was not required for blood collection, although he emphasized that the time between the collection and the transfusion should be kept to a minimum to ensure the safety and eficacy of the product. The direct use of serum will be discussed later in this chapter. The discovery of the anticoagulant properties of citrate solved one of the two aforementioned problems—the one of location. The issue of time remained a problem, awaiting a solution. As with the alcohol fractionation of plasma, World War I also played an important role. While Edwin Cohn (see below) was a logical candidate to drive the alcohol fractionation, it could be argued that Peyton Rous was an unlikely individual26 to have made the seminal observation on the effect of saccharides on red blood cell viability. Rous, at the Rockefeller Institute for Medical Research, established that, at least with the technology available during 1915–1918, there was no substitute for red blood cells in transfusion medicine. Rous continued to do some work in this area for several years but eventually returned to his work on the role of viruses in tumor development, resulting in the discovery of the Rous sarcoma virus and a Nobel prize.27 Rous and Turner28 reported in 1916 that the presence of glucose resulted in an extended in vitro viability of red blood cells. Subsequent work on the physiology of the red blood cells29 resulted in the development of the ACD (acid–citrate–dextrose) anticoagulant.30 More details on this work can be obtained from Mollison’s excellent review on the use of citrate and glucose in early transfusion medicine.31 The citrate/glucose method permitted the development of blood banks (Cook County, 193632)* and subsequently the plasma fractionation industry. Early blood banks were a little like early inancial banks in that the recipient and family/friends could make “deposits” to an “account” from which blood could be withdrawn. As with inancial banks, blood banks have changed over time, with a large volunteer *

There is mention of a blood bank during World War I and then later during the Spanish Civil War, both of which predate the Chicago blood bank. It has also been reported that the Mayo Clinic in Rochester, Minnesota, established a blood bank in 1935.

Plasma Fractionation and Plasma Protein Products

21

population used as donors; rarely are “paid” donors used for blood banks. With some exceptions,* blood banks operate separately from the plasma fractionation industry. Blood banks do offer a variety of products, including packed red blood cells, platelet concentrates, whole blood, fresh plasma, fresh frozen plasma, and cryoprecipitate.33 Blood is an expensive commodity whose cost may increase due to a variety of challenges,34 including increased screening, storage, and transfusion costs. An excellent study35 on the cost of blood transfusions in Sweden published in 2005 indicated a cost of €702 for a 2-unit allogeneic red blood cell transfusion and €598 for a 2-unit autologous red blood cell transfusion (both surgery patients). Similar costs are reported for Greece36 and the United Kingdom.37 A recent study by Shander and coworkers38 reported the costs of blood transfusion at four hospitals in the United States; the per unit cost of red blood cell transfusions to surgical patients ranged from US$ 522 to 1083 (mean US$ 761±294). While blood is a inite resource, there is little competition between blood banks* and the plasma fractionation industry for “raw material.” The study also suggested that biotechnology will permit the production of some blood factors currently derived from plasma,39 such as factor VIII, factor IX, and antithrombin, which might drive down the demand for plasma; however, there does not appear to be a viable alternative for intravenous immunoglobulin at the time of this writing (November, 2011). Speculation regarding the future of plasma-derived biopharmaceutical products is presented below. There had been sporadic attempts in the early twentieth century to identify unique fractions from blood for the treatment of disorders such as hemophilia (see Chapter 6), but such efforts were suspended for various reasons. It is fair to say that World War II was the driver of large-scale fractionation of plasma in the United States, England, and Germany. The blood plasma program was part of a much larger program on the use of blood in World War II, which is described in some detail in a volume from the Ofice of the Surgeon General of the Department of Army.40 Students of history as well as students of product development and product management would be well served by reading the preface by Brigadier General Douglas B Kendrick. Several points that deserve consideration some 50 years later have been stated. First, unless effort is made, lessons learned with respect to a response to an unusual situation, in this case the treatment of battleield casualties, are easily lost. Second, supporting administrative infrastructure is not readily regained when lost. Finally, the provision of blood and blood products requires a separate administrative structure, which explains why hospitals have a separate transfusion service. The use of serum as a therapeutic deserves mention. Serum was used early as a vehicle for passive immunization41 and has been largely replaced by plasma-derived intravenous immunoglobulin fraction or, in some cases, monoclonal antibody42 (Chapter 5). Prior to the resolution of the anticoagulation problem discussed previously, serum was seen as an effective treatment modality.43 Even today, albumin is frequently referred to as serum albumin, relecting its early characterization from a *

The American Red Cross collects blood that is used for red cells, platelets, and fresh frozen plasma. In addition, it recovers plasma from red cell preparations, which is used for plasma fractionation (see http://www.redcross.org). The plasma fractionation process mostly uses plasma collected by plasmapheresis at donor centers.

22

Biotechnology of Plasma Proteins

serum, rather than a plasma, source.44 Current literature frequently refers to the serum concentration of a protein therapeutic.45 The use of serum did present some problems,46 but its use continued through the 1930s.47 The infusion of serum can produce thrombosis, but such an effect is markedly enhanced by stasis.48 The literature during this period of time could be confusing as discussed by Strumia and coworkers,47 noting that the terms “serum” and “plasma” were frequently used interchangeably in the literature; Sapan and Lundblad have noticed that this practice has continued.49 Some plasma products (see Chapter 8) such as Autoplex could be considered serum protein derivatives based on analysis,50 as could be the recombinant factor VIIa.51 The irst part of the twentieth century saw a transition from donor-to-donor transfusion to the collection of blood from the donor as separate from the administration of blood to the recipient based on the development of anticoagulants. The next issue to be addressed was the container for collection and storage of whole blood. Parafin-coated containers were used in the later part of the nineteenth century, and sterilized glass bottles were introduced in the 1920s for the collection of blood but were replaced some 20–40 years later by plastic bags.52 Lozner and Taylor53 published a study in 1942 describing the “activating” effect of glass surfaces on blood coagulation when compared to parafin-coated tubes or lusteroid (nitrocellulose) tubes. It is fair to say that the transition from glass containers to plastic containers is as important as the ACD anticoagulant for the development of the plasma fractionation industry. Dr. Carl B. Walter, Dr. Walter Murphy, and colleagues at Harvard Medical School54–56 are given the credit for the development of the plastic bag for blood transfusion, working with Fenwal Laboratories (later part of Baxter-Travenol). As a historical note, the late Dale Smith led the commercial development of plastic bags for transfusion at Baxter. In the original study,54 Walter described a procedure for the collection of blood into a plastic bag through a small ion-exchange (sulfonated polystyrene) column that removed calcium and other divalent cations that were required for coagulation.* The blood collected in this manner was stable for 10 days, but a transition to collection in acid–citrate–dextrose preserving red cell function was accomplished in 1952.55,57 Plastic bags were originally composed of polyvinyl54,58 with phthalate as a plasticizer; other plastics and plasticizers have been developed.59 The use of phthalates as a plasticizer has created concern59,60 and remains controversial till now.61,62 Phthalate-free polyvinyl bags are currently available. Plasma fractionation as such came into its own as a process with the leadership of Professor E.J. Cohn during World War II40 and has evolved into an international business.63–66 Quite simply, the Cohn fractionation process67,68 uses the combined effect of pH, ionic strength, and organic solvent (solvent polarity) on the solubility of proteins69–71 to obtain several fractions (Table 2.1). The effect of alcohol on protein solubility has been known for some time, as has the effect of ionic strength on the effect *

The author has used a similar method to collect blood and observed differences when compared to the blood collected in EDTA.296 Other researchers have noted an effect of citrate other than calcium binding on blood coagulation.297 Chelating agents will, of course, remove cations other than calcium from whole blood with still largely unknown consequences. However, given the history of blood safety, it is unlikely that the use of citrate has a deleterious effect; there was an early report on the adverse effects of citrate.298

23

Plasma Fractionation and Plasma Protein Products

TABLE 2.1 Cohn Fractions from Human Plasmaa Cohn Fractionb I

Conditionsc 8% ethyl alcohol; pH 7.2; I = 0.14/−3°C

II

Contentsd Fibronectin217–225 ADAMTS 13 (vWF-cleaving protease)226 Glycosaminoglycans are also found in Cohn Fraction IV-1 and in smaller amounts in Cohn Fraction II + III 227 C1r subunit of the irst component of complement228,229 Factor VIIIe von Willebrand Factorf Fibrinogeng Immunoglobulin G230

IIIh

25% ethyl alcohol; pH 6.9; I = 0.09/−5°C

Mannan-binding lectin protein231,232 Eighth component of complement233 Basic proteins234 α2-Macroglobulini Factor IX235 H-icolin (Hakata antigen)236 P-icolin (P35)237 Apolipoprotein A-I and A-IIj

IVk

18% ethyl alcohol; pH 5.2; I = 0.09/−5°C (IV-1) 40% ethyl alcohol; pH 5.8; I = 0.09/−5°C (IV-4)

Antithrombin238,239 Apotransferrin (IV-4)240,241 Butyrylcholinesterase (IV-4)242 Protein C (IV-1)243 Ceruloplasmin (IV-1)l Basic somatomedin (IV-1)244,245 α1-Antiproteinase inhibitorm Protein C246,247 Support of cell culturen Insulin-like growth factor activity248 Insulin-like growth factor binding protein (SmBP)249,250 Immunosuppressive activity251,252 Inhibitor of acid lipase253 α-Galactosidase a254 Acid-activated plasma kallikrein255 Vitamin D–binding protein (Gc globulin)256,257 Lipoproteino Lipid258 Haptoglobin259 Plasminogenp (continued)

24

Biotechnology of Plasma Proteins

TABLE 2.1 (Continued) Cohn Fractions from Human Plasmaa Cohn Fractionb Vq VIr

Conditionsc 40% ethyl alcohol; pH 4.8; I = 0.11/−5oC

Contentsd Albumin260,261 Serine–threonine rich galactoglycoproteins

VIIt a

b

c

d

e

f

g h

i

j

The Cohn Fraction designation is taken from the Cohn Fraction Method 6 with the understanding that various manufacturers have added modiications that are proprietary in nature. Such modiications could include the combination of II + III into a single step and subsequent modiications of step IV, which eliminates the need for a IV-4 precipitation step. Fibrinogen, the von Willebrand factor, and factor VIII are now processed from the cryoprecipitate step.81,160,262 See footnote a and also see http://www.sanquin.nl/Sanquin-eng/sqn_From_blood_to_medicine.nsf/All/Plasma-Fractionation—Medicines-Derived-From-Plasma.html. From the Cohn Method 6 process as presented by Pennell.160 The conditions shown for Fraction III are for II + III. This list can be, at best, considered partial, as there is some disagreement regarding the deinition of plasma protein and the discovery of many proteins since the development of proteomic technology for protein identiication. The dificulty here, as discussed in the text, is the deinition, as many of the new, trace proteins may not it within the classical deinition of Putman.263 The separation between fractions is not absolute, and a protein or protein fraction (i.e., immunoglobulin) can be “spread” over several fractions, with a major quantity in one fraction. Such overlap is common with most precipitation puriication technologies such as ammonium sulfate.264 As an example, apolipoprotein C-II can be found in Fraction V.265 In the classic process of Cohn fractionation, factor VIII is found in the Cohn Fraction I, and this was an early source of factor VIII therapeutic products.266 The introduction of the cryoprecipitate step removed the bulk of factor VIII, ibrinogen, and the von Willebrand factor. The von Willebrand factor is present in the Cohn Fraction I but is removed in the cryoprecipitation step as is the situation with factor VIII. When factor VIII is puriied from the cryoprecipitate, it would appear that a unique form of the von Willebrand factor can be associated with factor VIII after subsequent puriication.267 See Chapter 6 for further discussion on this issue. Fibrinogen is one of the best-known constituents of the Cohn Fraction I.268,269 The Cohn Fraction III may be a historic footnote except for research purposes. I know of no commercial plasma fractionation process that yields a discrete Fraction III; processes have been optimized for the recovery of starting material for intravenous immunoglobulin and products such as α1-antitrypsin. In another development, the cryoprecipitate procedure has markedly decreased the value of a discrete Cohn Fraction I. More recent work on the puriication of IgG from plasma shows puriication from Fraction I + II + III.270 Thus, the older literature describes the puriication of factor IX from the Cohn Fraction III.271 More recent studies have used the Cohn Fraction IV-1 as a source of prothrombin complex concentrate.272 See Chapter 8 for additional details on prothrombin complex concentrates. There have been various suggestions for the therapeutic use of α2-macroglobulin over the last several decades, including its use as a transport vehicle.273,274 There have been suggestions for the use of apolipoprotein A-I as a therapeutic.275,276

Plasma Fractionation and Plasma Protein Products

25

TABLE 2.1 (Continued) Cohn Fractions from Human Plasmaa k

l

m

n

o

p q

r

s t

Fraction IV is taken to include material derived from Fraction IV-1 and Fraction IV-4. Fraction IV is considered to contain α-globulins,277 while β-globulins and γ-globulins are found in Fractions I–II–III. Fraction IV products have never been the economic drivers for plasma fractionation (see text), and the Cohn Fraction was considered a waste fraction from which albumin could be recovered.278 I would note that, at one point in time, the green color of the IV-1 paste was used (anecdotally) as a critical product attribute.279 α1-Proteinase inhibitors have previously been described as α1-antitrypsin.280,281 Somatomedin is also known as insulin-like growth factor, and there is less reference in the current work to somatomedin and more to insulin-like growth factor. The ability of serum to support cell culture activity is well accepted. Certain Cohn Fractions have been studied for their ability to support cell culture. Bovine Cohn IV-1 was found to substitute for bovine serum in the growth of Giardia lamblia. It was of interest that only goat, horse, and bovine Cohn IV-1 could support the growth of Giardia lamblia; agglutinating antibodies inhibitory factors in canine, human, rabbit, and rat fractions.282–284 The Cohn Fraction IV also stimulated the production of NGF from murine L cells.285 Earlier work had suggested that the Cohn Fraction IV could replace albumin as a cell culture supplement.286 Lipoprotein (β-lipoprotein) is also described as being present in the Cohn Fraction I + II + III,287,288 the Cohn Fraction III,289 and the Cohn Fraction III-O.290 Plasminogen has also been obtained from the Cohn Fraction III.291,292 The Cohn fractionation process is also applied to other species, most notably, bovine plasma. In particular, the Cohn Fraction V from bovine plasma (bovine serum albumin) has been used in cell culture.293,294 Fraction VI is derived from the supernatant fraction from the Cohn V precipitation step and has been poorly characterized. As described by Kendrick40 in Chapter XIII, Fraction VI “consists of the large amount of salts, especially citrates, and the small amount of protein left in the mother liquor following the removal of these various precipitates…” Peptides and acidic proteins have been obtained from this material. Found in the supernatant fraction from the Cohn V precipitation.295 A Cohn Fraction VII is mentioned by Pennell.160

of alcohol.72 Cohn was trained as a physical chemist at the University of Chicago and was a member of the Department of Physical Chemistry at Harvard Medical School for many years. During his long tenure at Harvard, he worked with other outstanding scientists such as John Edsall69 and George Scatchard.*,73 Cohn’s early work with E.J. Henderson concerned the physical chemistry of seawater74 and later on the physical *

A consideration of the contributions of biomedical scientists to World War II provides interesting stories. A number of scientists like Edsall, Scatchard, Hans Neurath, Peter Medawar, Erwin Chargaff, William Stein, Stanford Moore, and others made largely unknown contributions to the national effort and then moved on after the war to take up distinguished academic careers, sometimes in areas quite disparate from their work during World War II.

26

Biotechnology of Plasma Proteins

chemistry of bread.75 This latter work was done in collaboration with the Department of Defense and Professor Cohn was Lieutenant Cohn. While Cohn was interested in other problems during the period from 1920 to the mid-1930s, he never lost his primary interest in proteins76–80 and was well positioned to work on plasma fractionation during World War II.40 The original Cohn process has gone through modiications over the years, and alternative methods of fractionation have been developed.81 Despite advances in separation technology, the initial step in plasma fractionation remains ethanol fractionation. Kistler and Friedl82 have summarized the technical and economic advantages of ethanol fractionation. Most of the recent changes in plasma fractionation are intended to improve the yields of speciic fractions; as such, most of the technical information is proprietary in nature. The original process has been modiied by various manufacturers such that a cryoprecipitate fraction83,84 is removed and a Fraction I may or may not be obtained as a separate fraction. Fraction II and Fraction III may be obtained in a single step, and Fractions I–II–III may also be obtained in a single step following the cryoprecipitation step. Fraction IV may be obtained as Fraction IV-1 and Fraction IV-4. Fraction V is mostly albumin. Fraction VI is derived from the supernatant fraction obtained from the Fraction V step. Fraction VI has been poorly characterized, and there are only a few papers on this fraction.85–88 Plasma protein fraction is a plasma fraction similar to albumin.89–91 The processing of plasma into various products is not dissimilar to the fractionation of crude oil into various products. In a discussion of the economics of plasma fractionation, Burnouf92 noted the analogy of the reining process of oil where various derivative fractions and subfractions such as propane and butane are used to make fuel gas, light naphtha for gasoline, gas oils for lubricants, and residue for asphalt.93,94 Curran94 divides the petroleum industry into four separate technology and business areas: extraction of crude oil, transportation of oil to reinery, the reinery process, and marketing. Similarly, the plasma industry is divided into four areas: donor centers, refrigerated transport to a manufacturing process, the fractionation process, and distribution and sales. The reader is also directed to a chapter by Professor Angela Creager (Princeton) in Private Science.95 Professor Creager provides a unique view of the development of the plasma business at an important time in the ield of science in the United States. It was a time when there was considerable concern about the involvement of companies in plasma and the role of patents. Therefore, I strongly encourage anyone who is interested in the current state of commercial biotechnology to read this work by Professor Creager. The Cohn fractionation process was developed for the production of albumin, although the other proteins, most notably immune serum globulins (Chapter 5) and ibrin foam/ibrin ilm (Chapter 9), together with thrombin, were considered to be of value in 1945.96 The growth of the plasma fractionation industry slowed down for a period of time after World War II, with albumin (see Chapter 4) being the dominant product.97,98 There was some work on the development of factor VIII products (see Chapter 6) and intramuscular immunoglobulin (see Chapter 5).99,100 Albumin continued to be the economic driver for the fractionation of plasma until the 1960s, when the development of intermediate-purity factor VIII concentrates101 established hemophilia treatment as the economic driver for plasma fractionation (see Chapter 6). The subsequent development of recombinant factor VIII preparations in the 1980s, combined with the recognition of the value of intravenous immunoglobulin (IVIG)

Plasma Fractionation and Plasma Protein Products

27

in immunomodulation, resulted in IVIG replacing factor VIII as the economic driver for the plasma fractionation business.64 Other products derived from plasmas that were developed in the 1970s included the prothrombin complex concentrates, which have been largely replaced by single-factor IX concentrates from either plasma or recombinant sources. Activated prothrombin complex concentrates (see Chapter 8), while still in use, have been replaced by recombinant factor VIIa products, which are also being used for factor VII replacement in vitamin K–antagonist intoxication and liver disease. As an aside, it is the author’s sense that there are a large number of commercial entities seeking to capitalize on the use of factor VIIa as a general hemostatic agent quite separate from the aforementioned uses (see Chapter 8). The end of World War II resulted in the dismantling of the government infrastructure established for plasma fractionation during the conlict. During the past 50 years, a number of private companies have been involved in commercial plasma fractionation, including CSL in Australia; Grifols (Probitas), Immuno, Octapharm, Sanguin, and Behring in Europe; and Hyland, Cutter, Armour, and others in the United States. There are also efforts at the national level for fractionation aimed at ensuring self-suficiency for blood and blood products.63–66 This includes a new fractionation, plant being built in Brazil.65 Curling and Bryant64 have presented an excellent review of the plasma product industry in 2005; the reader is also referred to an article by Burnouf102 published in the same time period. Burnouf notes that the production of plasma-based therapeutics has never met the demand. Having said that, Burnouf also notes that it will be necessary to develop both new products and new indications for existing therapeutic products. The manufacture of biological products from human blood plasma has always been a challenging proposition. First, there is the issue of supply of raw material. Blood plasma for fractionation is usually obtained from commercial sources using paid donors.* This occurs at donor centers that are owned by either the fractionator or another company dedicated to plasma collection. Plasma, as a raw material, contributes to about 50% of the cost of the manufacturing process,64 with the *

I am embarrassed to say that after the current manuscript was sent to the publisher, I became aware of a book entitled Blood, Plasma and Plasma Proteins: A Unique Contribution to Modern Healthcare (ed. J.L. Valverde, IOS Press, Amsterdam, Netherlands, 2006). This book is an extraordinary collection of contributions on blood and plasma fractionation. The emphasis is more on the business side including plasma procurement and manufacturing. Victor Grifols, contribution entitled “Financing plasma proteins: Unique challenges” emphasizes the importance of IVIG and albumin as drivers of plasma fractionation. IVIG products require approximately 20 million liters of plasma, albumin slightly less, while plasma factor VIII requires half that amount, with much less required to factor IX. This disparity is suggested to relect the availability of recombinant factor VIII and factor IX. The related chapters by Peter Rankin (“Perilous economics of the plasma protein therapeutics industry”) and Thierry Burnouf (“Plasma proteins: Unique biopharmaceuticals—unique economics”) also discuss this product pricing and the related problem of cost allocation. A somewhat related issue, selfsuficiency, is discussed by Valverde (“The political dilemma of blood and plasma derivatives”). The issue of volunteer, nonremunerated donors versus paid donors is a complicated problem. As a result, the United States is self-suficient because of paid donors, while there are few other countries that can be self-suficient with volunteer donors. Valverde does suggest that the production and distribution of puriied plasma proteins obtained via plasma fractionation is a global business. It then follows that the regulation of the plasma fractionation is in desperate need of harmonization as discussed by von Hoegen and Gustafson in their chapter entitled “The importance of greater regulatory harmonization.”

28

Biotechnology of Plasma Proteins

cost balanced between acquisition, including storage and transport, and qualiication. The HIV tragedy of the 1980s and other viral cases have presented issues with blood-derived therapeutics.103 A substantial portion involves analytical costs including the cost of nucleic-based assays for virus testing as part of an overall strategy to ensure safety against viruses. It is noted that the methodology for viral testing continues to develop,104 and it is possible that multiplexed methods will reduce the cost of testing and increase the safety against viruses. The risk of infection resulting from a blood transfusion is estimated to be 1 in 1,000,000 for a unit of blood in the case of hepatitis C virus (HCV), 1 in approximately 300,000 in the case of hepatitis B virus (HBV), and approximately 1 in 3,000,000 in the case of HIV.105 Now these are single-unit odds, so these cannot be directly applied to the risk with the chronic use of a biological such as factor VIII for hemophilia A but might be reasonable for an acute-use product such as antithrombin. For a better sense of risk, I recommend the reader a book106 by James Walsh, which discusses how risk affects everyday life. Walsh’s book was published in 1998 and, as such, odds will have changed, and the following are presented to put the aforementioned odds in perspective. First, the lifetime odds of being struck by lightning are 1 in 30,000; death by excessive alcohol consumption, 1 in 100; and death by motor accidents, 1 in 60. I will grant you that these numbers are not directly comparable to the blood safety numbers, but they are useful in driving the analysis of risk rather than coping with the fog of uncertainty. Notwithstanding the safety of human blood in most geographies107 and determined efforts to guarantee safety in others,108 there is a tendency to emphasize the production of recombinant protein in protein-free cell culture systems.109,110 This approach appears to be based on a concept of zero-risk rather than risk versus beneit/cost. In the case of variant Creutzfeldt–Jakob disease, the theoretical potential of epidemic discussed in 1999111,112 never materialized.113,114 While the potential for blood transfusion via cellular elements exists, transmission via protein therapeutics has not been demonstrated.114 Aversion to plasma-derived therapeutics would appear to be based more on perceived uncertainty rather than on a deined risk. In the case of nonblood plasma substitutes for albumin, there are issues quite separate from the basic colloid substitution issue.115 As cost management becomes an increasing concern in health care, there will be an increased use of evidence-based medicine in the use of blood products.116–120 As an example, the oldest plasma protein therapeutic, albumin, and its use is still a topic of discussion,117,120 which is complicated by issues with therapeutic substitutes.115 In addition, it would appear that therapeutic effectiveness must be established by the plasma-derived product in which the recombinant product would be regarded as a follow-on biologic or biosimilar product.121 It is my opinion that there are products derived from plasma, such as albumin and IVIG, which (1) have strong safety records and (2) cannot be duplicated by recombinant DNA technology, in the case of IVIG (see Chapter 5). In the case of albumin, while recombinant albumin has been produced,122,123 the economics do not appear to be favorable for reasonable competition for the plasma-derived product; this could change with the use of novel recombinant product systems such as tobacco124 or algae.125 When recombinant factor VIII was produced by several companies in the 1980s, there was some thought that all therapeutic proteins would be produced by recombinant DNA technology, and there was considerable effort to

Plasma Fractionation and Plasma Protein Products

29

remove all proteins from the cell culture media and as excipients in the inal drug product. However, as of 2010, of the many intrinsic plasma proteins, only recombinant forms of factor IX, the von Willebrand factor, thrombin, and antithrombin have been licensed as biologicals in the United States. As noted earlier, recombinant albumin remains under development, as does α1-antitrypsin.126–128 The point that I am trying to make is that biologicals derived from human blood are safe within the reasonable limits of risk. The paramount issue regarding the current use and future development of plasmaderived biologicals relates to the risk of infection from a known or unknown pathogen. I trust that the reader understands that I do consider this a serious issue but that the risk of infection should be considered in light of therapeutic value and societal cost. The HIV tragedy described earlier has been a learning experience for the plasma industry and has resulted in the development of objective and subjective screening techniques for donors and donated blood. The increased appreciation of zoonotic disease emphasizes the importance of the unknown pathogens in products obtained from blood.129–133 The risk from the transfusion of blood is known (see above), and the risk from puriied protein fractions obtained from blood is less than that for whole blood134–139 with the removal by various processing steps demonstrated by various researchers.140–145 It is not unreasonable to suggest, at least for the sake of argument, that the risk of a plasma-derived therapeutic should be balanced with the cost of the plasma-derived product as compared to a recombinant product. The question then arises as to how much risk will the society accept and at what cost, which is a question of signiicance across all sectors of health care.146–148 As an example, recombinant factor VIII products are available at a substantial premium compared to plasma-derived factor VIII with essentially equivalent therapeutic equivalence.* Mantovani and colleagues149 presented an excellent study on the complex nature of treatment choice in hemophilia showing the combined importance of safety against viruses, inhibitor development, and infusion frequency. These researchers noted that product choice, when there is cost discrepancy between therapeutically equivalent products, is important when resources are limited. Other researchers have also provided an insight into this issue.150 Outcome analysis is used for other therapeutic approaches for determining the value,151 and it is clear that regardless of geography and reimbursement processes, resource allocation will be an increasing problem in health care.152 I would be remiss if I do not mention the current issue between recombinant human thrombin and bovine plasma–derived thrombin when there is a strong effort by supporters of bovine thrombin to preserve market share.153 Here, the issue is the development of antibodies against the bovine proteins, with little thought given to diseases caused by potential pathogens from bovine sources, such as variant Creutzfeldt–Jakob disease, which has driven the development of “protein-free” therapeutics. Robert154 argues that plasma fractionation will increase at a greater rate if intravenous immunoglobulin is approved for use in the case of Alzheimer disease. I do not *

As an example, a study conducted by a group of Italian researchers published in 2005299 using data from 2002 reported that the cost for a second-generation recombinant product (albumin-free formulation) was some 140% of a highly puriied plasma-derived product.

30

Biotechnology of Plasma Proteins

necessarily agree with this speciic conclusion (see Chapter 5) but do see an increase in the use of intravenous immunoglobulin (again see Chapter 5). There are a variety of potential indications for intravenous immunoglobulins in immunomodulation.155 Since it is unlikely that polyclonality is required for immunomodulation indications, there would be a pathway to a second-generation therapeutic product from the fractionation of an intravenous immunoglobulin active pharmaceutical ingredient. A similar argument can be made for infectious diseases in which it is clear that donor plasma could be selected for action against speciic pathogens,156,157 suggesting that donor plasmas could be preselected for the presence of antibodies of value in infectious diseases, as it is likely that a maximum therapeutic effect will be obtained from a polyclonal antibody preparation (see Chapter 5). It is of some interest that, considering the studies on the presence of antibodies for measles virus in intravenous immunoglobulin cited previously,156 one of the irst applications of human immune serum globulin was in the treatment of measles.158 The Cohn fractionation procedure was developed well in advance of the various separation technologies that are currently available for commercial biotechnology in the twenty-irst century. A perspective may be obtained by considering reviews by Taylor in 1953159 and a bit later by Pennell in 1960.160 Taylor159 reviews the state-of-the-art protein puriication during the mid-twentieth century. The available techniques were based on differential solubility, physical methods such as ultracentrifugation, preparative electrophoresis (free-boundary electrophoresis), and adsorption/elution from insoluble salts as well as the use of partition chromatography. Partition chromatography was developed by Gordon and coworkers161 in 1943 to separate amino acids and found application for proteins in the work by Martin and Porter in 1951.162 Adsorption chromatography for proteins on silica gel was reported by Shepard and Tiselius in 1949.163 The latter paper163 is prescient of hydrophobic interaction chromatography. The use of chromatography was in the earliest stage of development when Cohn and colleagues developed the alcohol fractionation scheme, and it would be some 40 years before Michael Grifith and colleagues in the Hyland Division of Baxter Healthcare applied immunoafinity chromatography for the puriication of plasma factor VIII,164 and other groups applied it for albumin and IgG.*,165–170 Lihme and colleagues171 have recently described the use of expanded bed chromatographic systems for the fractionation of plasma. I would be remiss if I do not mention some additional considerations about protein puriication. Protein puriication can be pursued for different goals as discussed by Linn.172 In the case of plasma fractionation, the goal is maximal recovery of the product with as few impurities as possible and no contaminants. Here, purity is relative, as impurities may be tolerated as long as they do not present a safety risk. As an example, consider the development of ultrapure factor VIII preparations (see Chapter 6). Here, the goal was not as much to obtain pure factor VIII as it was to eliminate the ibrinogen, which was a substantial impurity in therapeutic factor VIII preparations in the 1970s.173–175 The problem is that the *

It is important to understand that the introduction of column chromatography into a manufacturing environment based on iltration and centrifugation for separation technologies is a challenging proposition.300

Plasma Fractionation and Plasma Protein Products

31

half-life of ibrinogen is approximately 80 h,176 which is substantially longer than the 12–16 h half-life of factor VIII (see Chapter 6). It is critical to keep an account of the puriication and maintain a record.177 If one is willing to learn from the past (see Preface in Kendrick40) and do a rigorous analysis of development costs,178 it is possible to argue for the development of new plasma protein therapeutics. Admittedly, this is dependent on the unique nature of intravenous immunoglobulins, allowing most of the cost to be borne by those products.* Four issues impinge on the future of plasma-derived biopharmaceutical products. First, the emergence of another “HIV-like” pathogen would be devastating for the industry, limiting the market to only absolutely unique biopharmaceuticals such as intravenous immunoglobulin. Second, new indications for existing products such as the use of intravenous immunoglobulin in Alzheimer disease would increase plasma demand.179 Third, there is potential for the development of new products.92,180 This could be done with a change to the basic Cohn fractionation process, thereby eliminating any inluence on the licensure of existing products. A recent example is the development of α1-antitrypsin from the Cohn Fraction IV.181,182 Where there is a change in process that might inluence a downstream product, for example, a change in the processing of the Cohn Fraction I–III to maximize intravenous immunoglobulin yields might inluence the Cohn IV process, advanced characterization technologies,183 combined with insight gained through the considerations of biosimilars184,185 and Quality-by-Design (QbD),186 should facilitate the approval of changes. Fourth is the potential for the establishment of new markets for the existing products. Curling and Bryant, in their review of the plasma fractionation industry in 2005,64 observed that the developing economies represent an underserved market for all biopharmaceuticals, including the plasma-derived biologicals. The use of plasma-derived biologicals in developing economies raises the perennial issue of the self-suficiency of resources.187–190 Blood could be collected in the local geography and processed either locally or in another geography using contract manufacturing. The reader is referred to an article by Farrugia,62 which discusses the international movement of plasma and contract manufacturing. Human blood plasma for fractionation into products can be separated on the basis of the source.191–193 “Recovered” plasma is obtained from whole blood after the separation of red blood cells, while “source” plasma is collected from donors by plasmapheresis.† When I did not have any direct knowledge, I had assumed that plasmapheresis had been invented as a process either during World War II or shortly thereafter. Therefore, it was a bit of a surprise to the author‡ that plasmapheresis was developed in 1913 by a group from Johns Hopkins194 who coined the term using , the Greek αφαίρεσις (removal or withdrawal). Plasmapheresis allowed the repeated use of qualiied donors in commercial plasma collection centers.61 There are some *





In the spirit of full disclosure, the author (1) does not have a business background and (2) knows little, if anything, about cost allocation in commercial plasma fractionation. The term apheresis is frequently used in place of plasmapheresis. Plasmapheresis is used to describe the removal of plasma, with the cellular elements returned to the donor. Apheresis is a term used to describe the removal of speciic elements such as platelets or granulocytes, with the return of red blood cells and plasma to the donor.301 Apheresis is frequently used as a therapeutic process.302 An informal survey of colleagues suggested that the author is not alone in this matter.

32

Biotechnology of Plasma Proteins

differences in the quality of source plasma as compared to recovered plasma, but these differences do not inluence the product safety.156,191–193,195–197 One example of quality difference can be illustrated by the studies of Audet and coworkers,156 who found higher titers of measles antibody in recovered plasma versus source plasma, while Planitzer and colleagues196 found higher titers of echovirus antibody in source plasma as opposed to recovered plasma. It is of interest to note that plasmapheresis was introduced in the United Kingdom as a method for harvesting hyperimmune plasma for various indications.195 It can be argued that “local” plasma is invaluable in relecting the local immune experience,198,199 which would then be critical in guarding against local pathogens200 such as the H1N1 virus.201,202 Not all geographies have the same quality of plasma collection,108 and there may be different standards for the processing and storage of human plasma; there are differences between EU standards and U.S. standards for human plasma.62 There are, however, WHO standards for the collection and processing of blood and blood products, which could be used for harmonization.203–205 The differences between the U.S. and EU standards63 are concerned with screening (to the extent that the author has information, all manufacturers use the EU standards) and processing and storage temperatures (which center on the eutectic point of human plasma). There are signiicant economic issues in considering local versus imported materials.65 For example, in Brazil, the cost of imported plasma protein products for 2006 was more than US$ 300,000,000, approximately half of which was for intravenous immunoglobulin. This has encouraged the development of a new organization, Hemobrás,206 in Brazil for plasma fractionation. It is assumed that the new facility will use the existing technology that combines the Cohn fractionation and column chromatography. Considering some of the above discussion, newcomers to the fractionation business would ind it useful if they start “from scratch” using new technologies. This might be of particular value when the product might be used only within the geography where it is generated and would not have the burden of following the regulatory norms of the United States or the European Union. Chromatography of intact plasma is complicated by the viscosity of the starting material and the associated tendency to clog the ilter. The use of expanded bed chromatography207–209 offers a solution to these problems, and it has been used by Lihme and coworkers for the puriication of therapeutic proteins from plasma.171 A complementary approach would incorporate the cryoprecipitate step and process the cryopoor plasma. The cryoprecipitate can be used as it is presently used for therapeutic products,210–212 while the cryopoor plasma can be processed by chromatography to yield therapeutic protein preparations such as C-1 inhibitor,213 factor IX,214 factor VIIa,215 and prothrombin complex.216 The latter study216 used the supernatant fraction from Cohn I rather than cryosupernatant fraction.

REFERENCES 1. Hillyer, C.D., Transfusion Medicine and Hemostasis: Clinical and Laboratory Aspects, Elsevier, Burlington, MA, 2009. 2. Rossi, C.E. and Simon, T.L., Rossi’s Principles of Transfusion Medicine, WileyBlackwell, Chichester, 2009.

Plasma Fractionation and Plasma Protein Products

33

3. Perkins, J.G., Cap, A.P., Weiss, B.M., et al., Massive transfusion and nonsurgical hemostatic agents, Crit. Care Med. 36(7 Suppl.), S325–S339, 2008. 4. Caudill, J.S., Nichols, W.L., Plumhoff, E.A., et al., Comparison of coagulation factor XIII content and concentration in cryoprecipitate and fresh-frozen plasma, Transfusion 49, 765–770, 2009. 5. Yazer, M.H., Triulzi, D.J., Hassett, A.C., and Kiss, J.E., Cryoprecipitate prepared from plasma frozen within 24 hours after phebotomy contains acceptable levels of ibrinogen and VIIIC, Transfusion 50, 1014–1018, 2010. 6. Wade, C.E., Eastridge, B.J., Jones, J.A., et al., Use of recombinant factor VIIa in US military casualties for a ive-year period, J. Trauma 69, 353–359, 2010. 7. Shehata, N., Palda, V.A., Meyer, R.M., et al., The use of immunoglobulin therapy for patients undergoing solid organ transplantation: An evidence-based practice guideline, Transfus. Med. Rev. 24 (Suppl. 1), S7–S27, 2010. 8. Ficarra, B.J., Essays on Historical Medicine, Froben Press, New York, 1948. 9. Ficarra, B.J., Essays on Historical Medicine, Chapter 4, pp. 94–130, Froben Press, New York, 1948. 10. O’Hagan, D.T., Palin, K.J., and Davis, S.S., Intestinal absorption of proteins and macromolecules and immunological response, Crit. Rev. Ther. Drug Carrier Syst. 4, 197–220, 1988. 11. Muller, G., Oral delivery of protein drugs: Driver for personalized medicine, Curr. Issues Mol. Biol. 13, 13–24, 2010. 12. Eley, R.C., Green, A.A., and McKhann, C.F., The use of a blood coagulant extract from the human placenta in the treatment of hemophilia, J. Pediatr. 8, 135–147, 1936. 13. Bendien, W.M. and van Crevald, S., Investigations on hemophilia, J. Dis. Children 54, 713–725, 1937. 14. Eckhardt, R.D., Lewis, J.H., Murphy, T.L., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XXXIV. Comparative studies on the nutritive value of orally and intravenously administered human serum albumin in man, J. Clin. Invest. 27, 119–134, 1948. 15. Lane, S., Successful transfusion of blood, Lancet I, 185–188, 1840. 16. McLaughlin, G., The British contribution to blood transfusion in the nineteenth century, Brit. J. Anaesth. 31, 503–516, 1959. 17. Oberman, H.A., The crossmatch. A brief historical perspective, Transfusion 21, 645– 651, 1981. 18. Waterhouse, A., Yin, Y., Wise, S.G., et al., The immobilization of recombinant human tropoelastin on metals using a plasma-activated coating to improve the biocompatibity of coronary stents, Biomaterials 31, 8332–8340, 2010. 19. Soletti, L., Nieponice, A., Hong, Y., et al., In vivo performance of a phospholipid-coated bioerodable elastomeric graft for small-diameter vascular applications, J. Biomed. Mater. Res. A 96, 436–448, 2011. 20. Hutchins, P., History of blood transfusion: A bicentennial look, Surgery 64, 685–700, 1968. 21. Hicks, J.B., Cases of transfusion with some remarks on a new method of performing the operation, Guys Hospital Reports Series 3, 14, 1–14, 1869. 22. Hammersten, O., Untersuchungen ueber die Faserstoffgerinnung, Novae Acta Regiae Societatis Sceintiarum, Uppsala Series III, 10, 1–130, 1875. 23. Arthus, N.M. and Pagès, C., Nouvelle théorie chimique de la coagulation du sang, Arch. Physiol. Norm. Pathol. Paris 5th series, 2, 739–746, 1890. 24. Wright, A.E., On a method of determining the conditions of blood coagulability for clinical and experimental purposes, and on the effect of administration of calcium salts in haemophilia and actual or threatened haemorrhage, Brit. Med. J. II, 223–225, 1893.

34

Biotechnology of Plasma Proteins

25. Robertson, L.G., A method of citrated blood transfusion, Brit. Med. J. I, 477–479, 1918. 26. http://nobelprize.org/nobel_prizes/medicine/laureates/1966/rous.html, accessed Jan 20, 2011. 27. Javier, R.T. and Butel, J.S., The history of tumor virology, Cancer Res. 68, 7693–7706, 2008. 28. Rous, P. and Turner, J.R., The preservation of living red blood cells in vitro. I. Methods of preservation, J. Exp. Med. 23, 219–238, 1916. 29. Dawson, R.B., Preservation of red blood cells for transfusion, Hum. Pathol. 14, 213– 217, 1983. 30. Button, L.N. and Kevy, S.V., The development and acceptance of the anticoagulant, CPD, Vox. Sang. 48, 122–125, 1985. 31. Mollison, P.L., The introduction of citrate as an anticoagulant for transfusion and of glucose as red cell preservative, Brit. J. Haematol. 108, 13–18, 2000. 32. Fantus, B., The therapy of the Cook County Hospital, J. Am. Med. Assoc. 109, 128–131, 1937. 33. Hillyer, C.D., Silberstein, L.E., Ness, P.M., Anderson, K.C., and Roback, J.D. (eds.), Blood Banking and Transfusion Medicine, 2nd edn., Churchill Livingstone/Elsevier, Philadelphia, 2007. 34. Toner, R.W., Pizzi, L., Leas, B., et al., Cost to hospitals of acquiring and processing blood in the US: A survey of hospital-based blood banks and transfusion services, Appl. Health Econ. Health Policy 9, 29–37, 2011. 35. Glenngård, A.H., Persson, U., and Söderman, C., Costs associated with blood transfusions in Sweden—The societal cost of autologous and perioperative RBC transfusion, Transfusion Med. 15, 295–306, 2005. 36. Kanavos, P., Yfantopoulos, J., Vandoros, C., and Politis, C., The economics of blood: Gift of life or a commodity, Int. J. Technol. Assess. Health Care 22, 338–343, 2006. 37. Agrawal, S., Davidson, N., Walker, M., et al., Assessing the total costs of blood delivery to hospital oncology and haematology patients, Curr. Med. Res. Opin. 33, 1903–1909, 2006. 38. Shander, A., Hofmann, A., Ozawa, S., et al., Activity-based costs of blood transfusions in surgical patients at four hospitals, Transfusion 50, 753–765, 2010. 39. Prowse, C.V., Alternatives to human blood and blood resources, Vox. Sang. 74(Suppl. 2), 21–28, 1998. 40. Kendrick, D.B. (ed.), Blood Program in World War II, Superintendent of Documents, U.S. Government Printing Ofice, Washington, DC, 1964. 41. Eibl, M.M., History of immunoglobulin replacement, Immunol. Allergy Clin. North Am. 28, 737–764, 2008. 42. Dunman, P.M. and Nesin, M., Passive immunization as prophylaxis: When and where will this work?, Curr. Opin. Pharmacol. 3, 486–496, 2003. 43. Mann, F.G., Further study of surgical shock, J. Am. Med. Assoc. 71, 1184–1188, 1918. 44. Foster, J.F., Plasma albumin, in The Plasma Proteins, Vol. 1, ed. F.W. Putnam, Chapter 6, pp. 179–239, Academic Press, New York, 1960. 45. Stecenko, A.A. and Brigham, K.L., Gene therapy progress and prospects: Alpha-1antitrypsin, Gene Ther. 10, 95–99, 2003. 46. Brodie, T.G., The immediate actions of an intravenous injection of blood-serum, J. Physiol. 26, 48–71, 1900. 47. Strumia, M.M., Wagner, J.A., and Monaghan, J.F., The intravenous use of serum and plasma, fresh and preserved, Ann. Surg. 111, 623–629, 1940. 48. Wessler, S., Reimer, S.M., and Freiman, D.G., Thrombosis inducing factors in serum, Thromb. Diath. Haemorh. (Suppl. 1), 201–210, 1959. 49. Sapan, C.V. and Lundblad, R.L., Considerations regarding the use of blood samples in the proteomic identiication of biomarkers for cancer diagnosis, Cancer Genomics Proteomics, 33, 227–231, 2006.

Plasma Fractionation and Plasma Protein Products

35

50. Lundblad, R.L., Bergstrom, J. De Vreker, R., et al., Measurement of active coagulation factors in Autoplex-T with colorimetric active site-speciic assay technology, Thromb. Haemost. 80, 811–815, 1998. 51. Persson, E., Bolt, G., Steenstrup, T.D., and Ezban, M., Recombinant coagulation factor VIIa—From molecular to clinical aspects of a versatile haemostatic agent, Thromb. Res. 125, 483–489, 2010. 52. Diamond, L.K., History of blood banking, JAMA 193, 40–44, 1965. 53. Lozner, E.L. and Taylor, F.W.L., The effect of foreign surfaces on blood coagulation, J. Clin. Invest. 21, 241–246, 1942. 54. Walter, C.B., A new technic for collection, storage, and administration of unadulterated whole blood, Surg. Forum. 1950, 483–490, 1950. 55. Walter, C.B. and Murphy, W.P., Jr., A closed gravity technique for the preservation of whole blood in ACD solution utilizing plastic equipment, Surg. Gynecol. Obstet. 94, 687–692, 1952. 56. Button, L.N., Bernhard, W.F., and Walter, C.W., In vitro oxygenation of fresh whole blood for arterial perfusion, AMA Arch. Surg. 75, 183–187, 1957. 57. Sack, T., Gibson, J.G., and Buckley, E.S., The preservation of whole ACD blood collected, stored, and transfused in plastic containers, Surg. Gynecol. Obstet. 95, 113–119, 1952. 58. Sommer, A.J. and Koster, B., Studies on blood stored in plastic bags, Am. J. Clin. Pathol. 23, 818–827, 1953. 59. Carmen, R., The selection of plastic materials for blood bags, Transfusion Med. Rev. 7, 1–10, 1993. 60. Marcel, Y.L., Determination of di-2-ethylhexylphthalate levels in human blood plasma and cryoprecipitates, Environ. Health Perspect. 3, 119–121, 1973. 61. Myhre, B.A., Toxicological quandry of the use of bis(2-diethylhexyl)phthalate (DEHP) as plasticizer for blood bags, Ann. Clin. Lab. Sci. 18, 131–140, 1988. 62. Luban, N., Rals-Bahraml, K., and Short, B., I want to say to you—Just one word— “Plastics.” Transfusion 46, 503–506, 2006. 63. Farrugia, A., International movement of plasma and plasma contracting, Dev. Biol. (Basal), 120, 85–96, 2005. 64. Curling, J. and Bryant, C., The plasma fractionation industry. New opportunities to move forward?, Bioprocess International, March, pp. 18–27, 2005. 65. Farrugia, A., Evers, T., Falcon, P.-F., et al., Plasma fractionation issues, Biologicals 37, 88–93, 2009. 66. Cheraghali, A.M. and Abolghasemi, H., Improving availability and affordability of plasma-derived medicines, Biologicals 38, 81–86, 2010. 67. Cohn, E.J., Strong, L.E., Hughes, W.L., Jr., et al., Preparation and properties of serum and plasma proteins. IV. A system for the separation into fractions of the protein and lipoprotein components of biological tissues and luids, J. Am. Chem. Soc. 68, 459–475, 1946. 68. Cohn, E.J., The separation of blood into fractions of therapeutic value, Ann. Int. Med. 26, 341–352, 1947. 69. Edsall, J.T., The plasma proteins and their fractionation, Adv. Prot. Chem. 3, 383–479, 1947. 70. Brand, E. and Edsall, J.T., The chemistry of the plasma proteins and amino acids, Annu. Rev. Biochem. 16, 223–272, 1947. 71. Hughes, W.L., Interstitial proteins: The proteins of blood plasma and lymph, in The Proteins, eds. H. Neurath and K. Bailey, Vol. II, Part B., Chapter 21, pp. 663–754, Academic Press, New York, 1953. 72. Loeb, J., Proteins and the Theory of Colloidal Behavior, Chapter XX, McGraw-Hill, New York, 1924.

36

Biotechnology of Plasma Proteins

73. Scatchard, G., Edwin J. Cohn and protein chemistry, Vox. Sang. 17, 37–44, 1969. 74. Henderson, L.J. and Cohn, L.J., The equilibrium between acids and bases in sea water, Proc. Natl. Acad. Sci. USA 2, 618–622, 1916. 75. Cohn, E.J. and Henderson, L.J., The physical chemistry of bread making, Science 48, 501–505, 1918. 76. Cohn, E.J., Study on the physical chemistry of proteins I. The solubility of certain proteins at their isoelectric point, J. Gen. Physiol. 4, 697–722, 1922. 77. Cohn, E.J., McMeekin, T.L., Edsall, J.T., and Weare, J.H., Studies on the physical chemistry of amino acids, peptides and related substances II. The solubility of α-amino acids in water and alcohol–water mixtures, J. Am. Chem. Soc. 56, 2270–2282, 1934. 78. Cohn, E.J., McMeekin, T.L., Ferry, J.D., and Blanchard, M.H., The physical chemistry of amino acids, peptides, and related substances. XII. Interaction between dipolar ions in aqueous solutions, J. Phys. Chem. 43, 169–188, 1939. 79. Cohn, E.J., McMeekin, T.L., Oncley, J.L., et al., Preparation and properties of serum and plasma proteins. I. Size and charge of proteins separating upon equilibrium across membranes with ammonium sulfate solutions of controlled pH, ionic strength, and temperature, J. Am. Chem. Soc. 62, 3386–3393, 1940. 80. Cohn, E.J., Luetscher, J.A., Jr., Oncley, J.L., et al., Preparation and properties of serum and plasma proteins. III. Size and charge of proteins separating on equilibrium across membranes with ethanol–water mixtures of controlled pH, ionic strength and temperature, J. Am. Chem. Soc. 62, 3396–3400, 1940. 81. Curling, J.M. (ed.), Methods of Plasma Fractionation, Academic Press, London, 1980. 82. Kistler, P. and Friedl, H., Ethanol precipitation, in Methods of Plasma Fractionation, ed. J.M. Curling, Chapter 1, pp. 1–15, Academic Press, London, 1980. 83. Tanaka, K., Shigueoka, E.M., Sawatani, E., et al., Puriication of human albumin by the combination of the method of Cohn with liquid chromatography, Braz. J. Med. Biomed. Res. 31, 1383–1388, 1998. 84. Brown, P., Rohwer, R.G., and Dunstan, B.C., The distribution of infectivity in blood components and plasma derivatives in experimental models of transmissable spongiform encephalopathy, Transfusion 38, 810–818, 1998. 85. Covaci, A., Laub, R., Di Giambattista, M., et al., Polychlorinated biphenyls and organochlorine pesticides are eliminated from therapeutic factor VIII and immunoglobulin concentrates and reduced in albumin by plasma fractionation, Organohalogen Compounds 52, 172–175, 2001. 86. Burgi, W. and Schmid, K., Preparation and properties of zinc-α2-glycoprotein of normal human plasma, J. Biol. Chem. 236, 1066–1074, 1961. 87. Polet, H. and Spieker-Polet, H., Mechanism of the growth-promoting effect of serum albumin on concanavalin A-activated lymphocytes: Protective effect of the plasma proteins, J. Immunol. 117, 1275–1281, 1976. 88. Papp, A.C., Hai, E.R., and Wu, K.K., Binding of prostacyclin by plasma glycoproteins, Prostaglandins 30, 1057–1068, 1985. 89. Hink, J.H., Jr., Pappenhagen, A.R., Lundblad, J., and Johnson, F.F., Plasma protein fraction (human). Physical and clinical properties after storage for 7–8 years, Vox. Sang. 18, 527–541, 1970. 90. Vogelaar, E.F., Brummelhuis, H.G., Beentjes, S.P., and Krijnen, H.W., Contributions to the optimal use of human blood. I. Analysis and optimalization of the production of plasma protein fraction (PPF), Vox. Sang. 23, 481–492, 1972. 91. Lane, R.S. and Vallet, L., Human albumin and plasma protein fraction, Lancet 323 (8388), 1245–1246, 1984. 92. Burnouf, T., Plasma proteins: Unique biopharmaceuticals—Unique economics, Pharmaceutical Policy & Law 7, 209–218, 2005–2006.

Plasma Fractionation and Plasma Protein Products

37

93. Lynk, E.L., On the economics of the oil reining industry in the United Kingdom, Appl. Econ. 18, 113–126, 1986. 94. Curran, L.M., Waste minimization procedures in the petroleum industry, J. Hazard. Mater. 29, 189–197, 1992. 95. Creager, A.N.H., Biotechnology and blood: Edwin’s Cohn’s plasma fractionation project, 1941–1945, in Private Science, ed. A. Thackery, pp. 39–62, University of Pennsylvania Press, Philadelphia, 1998. 96. Anon, Byproducts of plasma fractionation. General consideration, in Blood Programs in World War II, ed. D.B. Kendrick, Chapter 13, pp. 350–369, Superintendent of Documents, U.S. Government Printing Ofice, Washington, DC, 1964. 97. Finlayson, J.S., Therapeutic plasma fractions and plasma fractionation, Sem. Thromb. Hemost. 6, 1–11, 1979. 98. Palmer, J.W., The evolution of large-scale plasma fractionation in the United States, in Proceedings of the Workshop on Albumin, DHEW Publication (NIH)76-925, U.S. Government Printing Ofice, Washington, DC, 1975. 99. Bodian, D., Experimental studies on passive immunization against poliomyelitis. I. Protection with human gamma globulin against intramuscular inoculation, and combined passive and active immunization, Am. J. Hyg. 54, 132–143, 1951. 100. Janeway, C.A., Rosen, F.S., Merla, E., and Alper, C.A., The Gamma Globulins, Little, Brown, Boston, MA, 1966. 101. Sharrer, I. and Becker, T., Products used to treat hemophilia: Evolution of treatment for hemophilia A and B, in Textbook of Hemophilia, eds. C.A. Lee, E.E. Berntorp, and W.K. Hoots, Blackwell, Malden, MA, 2005. 102. Burnouf, T., Plasma proteins: Unique biopharmaceuticals—Unique economics, Pharmaceutical Policy and Law 7, 209–218, 2005/2006. 103. Hoots, W.K., Transfusion-transmitted diseases: History of epidemics (focus on HIV), in Textbook of Hemophilia, eds. C.A. Lee, E.E. Berntorp, and W.K. Hoots, Chapter 35, pp. 200–206, Blackwell, Malden, MA, 2005. 104. Fournier-Wirth, C., Jaffrezic-Renault, N., and Coste, J., Detection of blood-transmissible agents: Can screening be miniaturized?, Transfusion 50, 2032–2045, 2010. 105. Dwyre, D.M., Fernando, L.P., and Holland, P.V., Hepatitis B, hepatitis C and HIV transfusion-transmitted infections in the 21st century, Vox. Sang. 100, 92–98, 2011. 106. Walsh, J., The Odds, Silver Lake Publishing, Los Angeles, CA, 1998. 107. Dodd, R., Managing the microbiological safety of blood for transfusion: A US perspective, Future Microbiol. 4, 807–818, 2009. 108. Emmanuel, J.C., Material & equipment, procurement & maintenance: Impact on blood safety, Biologicals 38, 78–80, 2010. 109. Key, N.S. and Negrier, C., Coagulation factor concentrates: Past, present, and future, Lancet 370, 439–448, 2007. 110. Grillberger, L., Kreil, T.R., Nasr, S., and Reiter, M., Emerging trends in plasma-free manufacturing of recombinant protein therapeutics expressed in mammalian cells, Biotechnol. J. 4, 186–201, 2009. 111. Collinge, J., Variant Creutzfeldt–Jakob disease, Lancet 354, 317–324, 1999. 112. Turner, M., The impact of new-variant Creutzfeldt disease on blood transfusion practice, Brit. J. Haematol. 106, 842–850, 1999. 113. Lefrère, J.J. and Hewitt, P., From mad cows to sensible blood transfusion: The risk of prion transmission by labile blood components in the United Kingdom and in France, Transfusion 49, 797–812, 2009. 114. Turner, M.L. and Ludlam, C.A., An update on the assessment and management of the risk of transmission of variant Creutzfeldt–Jakob disease by blood and plasma products, Brit. J. Haematol. 144, 14–23, 2009.

38

Biotechnology of Plasma Proteins

115. Boldt, J., Safety of nonblood plasma substitutes: Less frequently discussed issues, Eur. J. Anaesthesiol. 27, 495–500, 2010. 116. Jairath, V., Hearnshaw, S., Brunskill, S.J., et al., Red cell transfusion for the management of upper gastrointestinal haemorrhage, Cochrane Database Syst. Rev. (9), CD006613, 2010. 117. Holm, C., Resuscitation in shock associated with burns. Tradition or evidence-based medicine?, Resuscitation 44, 157–164, 2000. 118. Zimmerman, J.L., Use of blood products in sepsis: An evidence-based review, Crit. Care Med. 32(11 Suppl.), S542–S547, 2004. 119. Vamvakas, E.C. and Blajchman, M.A., Blood still kills: Six strategies to further reduce allogeneic blood transfusion-related mortality, Transfus. Med. Rev. 24, 77–124, 2010. 120. Farrugia, A., Albumin usage in clinical medicine: Tradition or therapeutic?, Transfus. Med. Rev. 24, 53–63, 2010. 121. Roger, S.D. and Goldsmith, D., Biosimilars: It’s not as simple as cost, J. Clin. Pharmacy Therapeutics 33, 456–464, 2008. 122. Kobayashi, K., Summary of recombinant human serum albumin development, Biologicals 34, 55–59, 2006. 123. Chuang, V.T. and Otagiri, M., Recombinant human serum albumin, Drugs Today (Barc) 43, 547–561, 2007. 124. Tremblay, R., Wang, D., Jevnikar, A.M., and Ma, S., Tobacco, a highly eficient green bioreactor for production of therapeutic proteins, Biotechnol. Adv. 28, 214–221, 2010. 125. Specht, E., Miyake-Stoner, S., and Mayield, S., Micro-algae come of age as a platform for recombinant protein product, Biotechnol. Lett. 32, 1373–1383, 2010. 126. Karnaukhova, E., Ophir, Y., and Golding, B., Recombinant human alpha-1-proteinase inhibitor: Toward therapeutic use, Amino Acids 30, 317–332, 2006. 127. Tonelli, A.R. and Brantly, M.L., Augmentation therapy in alpha-1-antitrypsin deicieny: Advances and controversies, Ther. Adv. Respir. Dis. 4, 289–312, 2010. 128. Agarwal, S., Jha, S., Sanyal, I., and Amla, D.V., Expression and puriication of recombinant human alpha-1-proteinase inhibitor in and its single amino acid substituted variants in Escherichia coli for enhanced stability and biological activity, J. Biotechnol. 147, 64–72, 2010. 129. Tang, J.W., Shetty, N., Lam, T.T., et al., Emerging, novel, and known inluenza virus infections in humans, Infect. Dis. Clin. North Am. 24, 603–617, 2010. 130. Roess, A.A., Galan, A., Kitces, E., et al., Novel deer-associated parapoxvirus infection in deer hunters, N. Engl. J. Med. 363, 2621–2627, 2010. 131. Leiby, D.A., Transfusion-transmitted Babesia spp.: Bull’s-eye on Babesia microti, Clin. Microbiol. Rev. 24, 14–28, 2011. 132. Altizer, S., Bartel, R., and Han, B.A., Animal migration and infectious disease risk, Science 331, 296–302, 2011. 133. Burnouf, T. and Radosevich, M., Reducing the risk of infection from plasma products: Speciic preventative strategies, Blood Rev. 14, 94–110, 2000. 134. Cai, K., Gierman, T.M., Hotta, J., et al., Ensuring the biologic safety of plasma-derived therapeutic proteins—Detection, inactivation, and removal of pathogens, BioDrugs 19, 79–96, 2005. 135. Burdick, M.D., Pifat, D.Y., Petteway, S.R., and Cai, K., Clearance of prions during plasma protein manufacture, Transfus. Med. 20, 57062, 2006. 136. Poelsler, G., Berting, A., Kindermann, J., et al., A new liquid intravenous immunoglobulin with three dedicated virus reduction steps: Virus and prion reduction capacity, Vox. Sang. 94, 184–192, 2008. 137. Agjaie, A., Pourfathollah, A.A., Bathaie, S.Z., et al., Structural study on immunoglobulin G solution after pasteurization with and without stabilizer, Transfus. Med. 18, 62–70, 2008.

Plasma Fractionation and Plasma Protein Products

39

138. Roberts, P.L., Lloyd, D., and Marshall, P., Virus inactivation in a factor VIII/VWF concentrate treated using solvent/detergent procedure based on polysorbate 20, Biologicals 3, 26–31, 2009. 139. Jeong, E.K., Sung, H.M., and Kim, I.S., Inactivation and removal of inluenza A virus H1N1 during the manufacture of plasma derivatives, Biologicals 38, 652–657, 2010. 140. Prince, A.M., Piët, M.P., and Horowitz, B., Effect of Cohn fractionation conditions on infectivity of the AIDS virus, New. Engl. J. Med. 314, 386–387, 1986. 141. Mitra, G., Wong, M.F., Mozen, M.M., et al., Elimination of infectious retroviruses during preparation of immunoglobulins, Transfusion 26, 394–397, 1986. 142. Yei, S., Yu, M.W., and Tankersley, D.L., Partitioning of hepatitis C virus during CohnOncley fractionation of plasma, Transfusion 32, 824–828, 1992. 143. Bos, O.J., Sunyé, D.G., Nieuweboer, C.E., et al., Virus validation of pH 4-treated human immunglobulin products by the Cohn fractionation process, Biologicals 26, 267–276, 1998. 144. Horwith, G. and Revie, D.R., Eficacy of viral clearance methods used in the manufacture of activated prothrombin complex concentrate: Focus on AUTOPLEX T, Haemophilia 5(Suppl. 3), 19–23, 1999. 145. Gregori, L., Maring, J.-A., MacAuley, C., et al., Partitioning of TSE infectivity during ethanol fractionation of human plasma, Biologicals 32, 1–10, 2004. 146. Brown, S.A., Aledort, L.M., et al., Economic Challenges in haemophilia, Haemophilia 11, 64–72, 2005. 147. Plosker, G.L. and Lyseng-Williamson, K.A., Atorvastatin: A pharmacoeconomic review of its use in the primary and secondary prevention of cardiovascular events, Pharmacoeconomics 25, 1031–1053, 2007. 148. Bodrogi, J. and Kaló, Z., Principles of pharmacoeconomics and their impact on strategic imperatives of pharmaceutical research and development, Brit. J. Pharmacol. 159, 1367–1373, 2010. 149. Mantovani, L.G., Monzini, M.S., Mannucci, P.M., et al., Differences between patients’, physicians’ and pharmacists’ preferences for treatment products in haemophilia: A discrete choice experiment, Haemophilia 11, 589–597, 2005. 150. O’Connor, A.M., Bennett, C.L., Stacey, D., et al., Decision aids for people facing health treatment or screening decisions, Cochrane Database Syst. Rev. (3), CD001431, 2009. 151. Colquitt, J.L., Jones, J., Tan, S.C., et al., Ranibizumab and pegaptanib for the treatment of age-related macular degeneration: A systematic review and economic evaluation, Health Technol. Assess. 12, iii–207, 2008. 152. Burkart, J., The future of peritoneal dialysis in the United States: Optimizing its use, Clin. J. Am. Soc. Nephrol. 4(Suppl. 1), S125–S131, 2009. 153. Bhandari, M., Ofosu, F.A., Mackman, N., et al., Safety and eficacy of Thrombin-JMI: A multidisciplinary expert group consensus, Clin. Appl. Thromb. Hemost. 17, 39–45, 2011. 154. Robert, P., Global plasma demand in 2015, Pharm. Policy Law 11, 359–367, 2009. 155. Sapan, C.V., Reisner, H.M., and Lundblad, R.L., Antibody Therapy (IVIG): Evaluation of the use genomics and proteomics for the study of immunomodulation therapeutics, Vox. Sang. 92, 197–205, 2007. 156. Audet, S., Virata-Theimer, M.L., Beeler, J.A., et al., Measles–virus–neutralizing antibodies in intravenous immunoglobulin, J. Infect. Dis. 194, 781–789, 2006. 157. Sullivan, J.S., Selleck, P.W., Downton, T., et al., Heterosubtypic anti-avian H5N1 inluenza antibodies in intravenous immunoglobulins from globally separate populations protect against H5N1 infection in cell culture, J. Mol. Genet. Med. 3, 217–224, 2009. 158. Stokes, J., Jr., Maris, E.P., and Gellis, S.S., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XI. The use of concentrated normal human serum gamma globulin (human immune serum globulin) in the prophylaxis and treatment of measles, J. Clin. Invest. 23, 531–549, 1944.

40

Biotechnology of Plasma Proteins

159. Taylor, J.F., The isolation of proteins, in The Proteins, eds. H. Neurath and K. Bailey, Vol. 1, Pt. B., Chapter 1, pp. 1–85, Academic Press, New York, 1953. 160. Pennell, R.B., Fractionation and isolation of puriied components by precipitation methods, in The Plasma Proteins, ed. F.W. Putman, Chapter 2, pp. 9–50, Academic Press, New York, 1960. 161. Gordon, A.H., Martin, A.J.P., and Synge, R.L.M., Partition chromatography in the study of protein constituents, Biochem. J. 37, 79–86, 1943. 162. Martin, A.J.P. and Porter, R.B., The chromatographic fractionation of ribonuclease, Biochem. J. 49, 215–218, 1951. 163. Shepard, C.C. and Tiselius, A., The chromatography of proteins. The effect of salt concentration on the adsorption of proteins to silica gel, Discuss. Faraday Soc. 7, 275–283, 1949. 164. Addiego, J.E., Jr., Gomperts, E., Liu, S.L., et al., Treatment of hemophilia A with a highly puriied factor VIII concentrate prepared by anti-FVIIIc immunoafinity chromatography, Thromb. Haemost. 67, 19–27, 1992. 165. Fourcart, J., Saint-Blancard, J., Girot, P., and Boschetti, E., Préparation de l’albumine et des immunoglobulines G par fractionnment chromatographique direct du plasma humain sur DEAE et CM—Trisacryl M, Rev. Franc. Trans. Immunohematol. 25, 7–17, 1982. 166. Berglöf, J.H., Eriksson, S., and Curling, J.M., Chromatographic preparation and in vitro properties of albumin from human plasma, J. Appl. Biochem. 5, 282–292, 1983. 167. Tayot, J.L., Tardy, M., Gattel, P., et al., Large scale use of Spherosil ion exchangers in plasma fractionation, Dev. Biol. Stand. 67, 15–24, 1987. 168. Li, G., Stewart, R., Conlan, B., et al., Puriication of human immunoglobulin G: A new approach to plasma fractionation, Vox. Sang. 83, 332–338, 2002. 169. Buchacher, A. and Iberer, G., Puriication of intravenous immunoglobulin G from human plasma: Aspects of yield and virus safety, Biotechnol. J. 1, 148–163, 2006. 170. Wolf, M., Kronenberg, H., Dodds, A., et al., A safety study of Albumex 5, a human albumin solution produced by ion exchange chromatography, Vox. Sang. 70, 198–202, 1996. 171. Lihme, A., Hansen, M.B., Anderson, I.V., and Burnouf, T., A novel core fractionation process of human plasma by expanded bed adsorption chromatography, Anal. Biochem. 399, 102–109, 2010. 172. Linn, S., Strategies and considerations for protein puriication, Meth. Enzymol. 463, 9–19, 2009. 173. Lopaciuk, S., Latallo, Z.S., Bykowska, K., et al., Separation of human antihemophilic factor (AHF, factor VIII) from ibrinogen by means of deibrase, Folia Haematol. 102, 671–678, 1975. 174. Allain, J.P., Verroust, F., and Soulier, J.P., In vitro and in vivo characterization of factor VIII preparations, Vox. Sang. 38, 68–80, 1980. 175. Austen, D.E. and Smith, J.K., Factor VIII fractionation on aminohexyl Sepharose with possible reduction in hepatitis B antigen, Thromb. Haemost. 48, 46–48, 1982. 176. Manco-Johnson, M.J., Dimichele, D., Castaman, G., et al., Fibrinogen Concentrate Study Group. Pharmacokinetics and safety of ibrinogen concentrate, J. Thromb. Haemost. 7, 2064–2069, 2009. 177. Burgess, R.R., Preparing a puriication summary table, Meth. Enzymol. 463, 29–34, 2009. 178. Farid, S.S., Process economics of industrial monoclonal antibody manufacture, J. Chromatogr. A 848, 8–18, 2007. 179. Dodel, R., Neff, F., Noelker, C., et al., Intravenous immunoglobulins as a treatment for Alzheimer’s disease: Rationale and current evidence, Drugs 70, 513–528, 2010.

Plasma Fractionation and Plasma Protein Products

41

180. Farrugia, A., Trialing plasma protein therapies for rare disorders: Thinking outside the box, Pharm. Policy Law 11, 345–352, 2009. 181. Chen, S.X., Hammond, D.J., Lang, J.M., and Lebing, W.R., Puriicaton of α1 proteinase inhibitor from human plasma fraction IV-1 by ion exchange chromatography, Vox. Sang. 74, 232–241, 1998. 182. Zimmerman, T.P., Yield improvement for manufacture of α1-proteinase inhibitor, Vox. Sang. 91, 309–315, 2006. 183. Lundblad, R.L., Approaches to the Conformational Analysis of Biopharmaceuticals, CRC Press, Boca Raton, FL, 2010. 184. Jelkmann, W., Biosimilar epoetins and other “follow-on” biologics: Update on the European experiences, Am. J. Hematol. 85, 771–780, 2010. 185. Wang, Y.M. and Chow, A.T., Development of biosimilars—Pharmacokinetic and pharmacodynamic considerations, J. Biopharm. Stat. 20, 46–61, 2010. 186. de Val, I.J., Kontorvavdi, C., and Nagy, J.M., Towards the implementation of quality by design to the production of therapeutic monoclonal antibodies with desired glycosylation patterns, Biotechnol. Prog. 26, 1505–1527, 2010. 187. Bocciardo, L., Martinengo, M., Ardenghi, D., et al., Plasma derivatives and strategies for reaching self-suficiency in Liguria: The role of the Transfusion Medicine Service of the Gaslini Institute, Blood Transfus. 5, 85–92, 2007. 188. Dyer, C., “Human tragedy” was due to delay in achieving national self suficiency in blood products, BMJ 333, b808, 2009. 189. Park, Q., Kim, M.J., Lee, J., et al., Plasma fractionation in Korea: Working towards selfsuficiency, Korean J. Hematol. 45, 3–5, 2010. 190. Rautonen, J., Self-suficiency, free trade and safety, Biologicals 38, 97–99, 2010. 191. Pusey, G., Dash, C., Garrett, M., et al., Experience of using human albumin solutions 4.5% in 1195 therapeutic plasma exchanges, Transfus. Med. 20, 244–249, 2010. 192. Bianco, C., Impact of source plasma standards on recovered plasma, Dev. Biol. (Basal) 120, 97–100, 2005. 193. Laub, R., Baurin, S., Timmerman, D., et al., Speciic protein content of pools of plasma for fractionation from different sources: Impact of frequency of donations, Vox. Sang. 99, 220–231, 2010. 194. Abel, J.J., Rowntree, L.G., and Turner, B.B., Plasma removal with the return of corpuscles (plasmapheresis), J. Pharmacol. Exp. Ther. 5, 625–641, 1913–1914. 195. Smith, J.K., Quality of plasma for fractionation: Does it matter?, Transfus. Sci. 15, 343– 350, 1994. 196. Planitzer, C.B., Farcet, M.R., Schiff, R.I., et al., Neutralization of different echovirus serotypes by individual lots of intravenous immunoglobulin, J. Med. Virol. 83, 305–310, 2011. 197. Geng, Y., Wu, C.G., Bhattacharayya, S.P., et al., Parvovirus B18 DNA in Factor VIII concentrates: Effects of manufacturing procedures and B19 screening by nucleic acid testing, Transfusion 47, 883–889, 2007. 198. Matejtschuk, P., Chitwick, Y., Prince, A., et al., A direct comparison of the antigenspeciicity antibody proile of intravenous immunoglobulins derived by US and UK donor plasma, Vox. Sang. 83, 17–22, 2002. 199. Farcet, M.R., Planitzer, C.B., Stein, O., et al., Hepatitis A virus antibodies in immunoglobulin preparations, J. Allergy Clin. Immunol. 125, 198–202, 2010. 200. Luke, T.C., Casadevall, A., Watowich, S.J., et al., Hark back: Passive immunotherapy for inluenza and other serious infections, Crit. Care Med. 38(4 Suppl.), e66–e73, 2010. 201. Hung, I.F., To, K.K., Lee, C.K., et al., Effect of clinical and virological parameters on the level of neutralizing antibody against pandemic inluenza A virus H1N1 2009, Clin. Infect. Dis. 51, 274–279, 2010.

42

Biotechnology of Plasma Proteins

202. Wong, H.K., Lee, C.K., Hung, I.F., et al., Practical limitations of convalescent plasma collection: A case scenario in pandemic preparation for inluenza A (H1N1) infection, Transfusion 50, 1967–1971, 2010. 203. Anon, The Collection, Fractionation, Quality Control and Use of Blood and Blood Products, WHO, Geneva, Switzerland, 1981. 204. Anon, Recommendations for production, control, and regulation of human plasma for fractionation, WHO Technical Report Series No. 941, 2007. 205. Greening, D.N., Glenister, K.M., Sparrow, R.L., and Simpson, R.J., International blood collection and storage: Clinical use of blood products, J. Proteomics 73, 386–393, 2010. 206. http://www.hemobras.gov.br, accessed Aug 20, 2010. 207. Chase, H.A., Puriication of proteins by adsorption chromatography in expanded beds, Trends Biotechnol. 12, 296–303, 1994. 208. Kaufmann, M., Unstable proteins: How to subject them to chromatographic separations for puriication procedures, J. Chromatogr. B 699, 347–369, 1997. 209. Anspach, F.D., Curbelo, D., Hartmann, R., et al., Expanded-bed chromatography in primary protein puriication, J. Chromatogr. A 865, 129–144, 1999. 210. Burnouf-Radosevich, M. and Burnouf, T., Chromatographic preparation of a therapeutic highly puriied von Willebrand factor concentrates from human cryoprecipitate, Vox. Sang. 62, 1–11, 1992. 211. Branović, K., Gebauer, B., Trescec, A., and Benko, B., Characterization of F VIII concentrates produced by two methods incorporating double virus inactivation, Appl. Biochem. Biotechnol. 69, 99–111, 1998. 212. Mazurier, C., Poulle, M., Samor, B., et al., In vitro study of a triple-secured von Willebrand factor concentrate, Vox. Sang. 84, 54–64, 2003. 213. Poulle, M., Burnouf-Radesovich, M., and Burnouf, T., Large-scale preparation of highly puriied human C1-inhibitor for therapeutic use, Blood Coagul. Fibrinolysis 5, 543–549, 1994. 214. Hoffer, L., Schwinn, H., and Josić, D., Production of highly puriied clotting factor IX by a combination of different chromatographic methods, J. Chromatogr. A 844, 119– 128, 1999. 215. Tomokiyo, K., Yano, H., Imamura, M., et al., Large-scale production and properties of human plasma-derived activated factor VII concentrate, Vox. Sang. 84, 54–64, 2003. 216. McCann, K.B., Gomme, P.T., Wu. J., et al., Evaluation of expanded bed adsorption chromatography for extraction of prothrombin complex from Cohn supernatant I, Biologicals 36, 227–233, 2008. 217. Maurer, L.M., Tomasini-Johansson, B.R., and Mosher, D.F., Emerging roles of ibronectin in thrombosis, Thromb. Res. 125, 287–291, 2010. 218. Bar, L., Malko, O., Naboichenko, E., and Nur, I., The binding of ibrin sealant to collagen is inluenced by the method of puriication and the cross-linked ibrinogen–ibronectin (heteronectin) content of the ‘ibrinogen’ component, Blood Coag. Fibrinolysis 16, 111–117, 2005. 219. Hughes, C., Thomas, K.B., Schiff, P., et al., Effect of delayed blood processing on the yield of factor VIII in cryoprecipitate and factor VIII concentrate, Transfusion 28, 566– 570, 1988. 220. Mazurier, C., Samor, B., Deromeuf, C., and Goudemand, M., The role of ibronectin in factor VIII/von Willebrand factor cryoprecipitation, Thromb. Res. 37, 651–658, 1985. 221. Reilly, I.T., McVerry, B.A., and Mackie, M.J., Fibronectin in blood products—An in vitro and in vivo study, J. Clin. Pathol. 36, 1377–1381, 1983. 222. Robbins, A.B., Doran, J.E., Reese, A.C., and Mansberger, A.R., Jr., Clinical response to cold insoluble globulin replacement in a patient with sepsis and thermal injury, Am. J. Surg. 142, 636–638, 1981.

Plasma Fractionation and Plasma Protein Products

43

223. Wood, G., Rucker, M., Davis, M.W., et al., Interaction of plasma ibronectin with selected cryoglobulins, Clin. Exp. Immunol. 40, 358–364, 1980. 224. Saba, T.M. and Cho, E., Reticuloendothelial systemic response to operative trauma as inluenced by cryoprecipitate or cold-insoluble globulin therapy, J. Reticuloendothel. Soc. 26, 171–186, 1979. 225. Stathakis, N.E. and Mosesson, M.W., Interactions among heparin, cold-insoluble globulin and ibrinogen in formation of the heparin-precipitable fraction of plasma, J. Clin. Invest. 60, 855–865, 1977. 226. Aronson, D.L., Krizek, D.M., and Rick, M.E., von Willebrand factor-cleaving protease content of Cohn plasma fractions, Vox. Sang. 85, 123, 2003. 227. Cecchi, F., Ruggiero, M., Cappelletti, R., et al., Improved method for analysis of glycosaminoglycans in glycosaminoglycan/protein mixtures: Application in Cohn-Oncley fractions of human plasma, Clin. Chim. Acta 376, 142–149, 2007. 228. Haupt, H. and Baudner, S., Isolation and crystallization of the C1r subunit of the irst component of human complement, Hoppe Seylers Z. Physiol. Chem. 362, 1147–1150, 1981. 229. Bing, D.H., Andrews, J.W., Morris, K.M., et al., Puriication of the subcomponents C1q, C1(−)r and C1(−)s of the irst component from Cohn Fraction I by afinity chromatography, Prep. Biochem. 10, 269–296, 1980. 230. Aghaie, A., Pourfatollah, A.A., Bathaie, S.Z., et al., Preparation, puriication and virus inactivation of intravenous immunoglobulin from human plasma, Hum. Antibodies 19, 1–6, 2010. 231. Kilpatrick, D.D., Isolation of human mannan binding lectin, serum amyloid P component and related factors from Cohn fraction III, Transfus. Med. 7, 289–294, 1997. 232. Laursen, I., Houen, G., Højrup, P., et al., Second-generation nanoiltered plasma-derived mannan-binding lectin product: Process and characteristics, Vox. Sang. 92, 338–350, 2007. 233. Steckel, E.W., York, R.G., Monahan, J.B., and Sodetz, J.M., The eighth component of human complement. Puriication and physicochemical characterization of its unusual subunit structure, J. Biol. Chem. 255, 11997–12005, 1980. 234. Suzuki, K. and Schmid, K., Basic proteins of Cohn fraction 3 of human plasma, Arch. Biochem. Biophys. 123, 421–422, 1968. 235. Monahan, J.B. and Sodetz, J.M., Identiication of human Cohn fraction III as a useful source for the simultaneous puriication of FIX and FX zymogens, Thromb. Res. 19, 743–755, 1980. 236. Masushita, M., Kuraya, M., Hamasaki, N., et al., Activation of the lectin complement pathway by H-icolin (Hakata antigen), J. Immunol. 168, 3502–3506, 2002. 237. Kilpatrick, D.C., Fujita, T., and Matsushita, M., P35, an opsonic lectin of the icolin family, in human blood from neonates, normal adults, and recurrent miscarriage patients, Immunol. Lett. 67, 109–112, 1999. 238. Wickerhause, M., Williams, C., and Mercer, J., Development of large scale fractionation methods VII. Preparation of antithrombin III concentrate, Vox. Sang. 36, 281– 293, 1979. 239. Hoffman, D.L., Puriication and large-scale preparation of antithrombin III, Am. J. Med. 87, 23S–26S, 1989. 240. von Bonsdorff, L., Tölo, H., Lindeberg, E., et al., Development of a pharmaceutical apotransferrin product for iron-binding therapy, Biologicals 29, 27–37, 2001. 241. Ascione, E., Muscariello, L., Maiello, V., et al., A simple method for large-scale puriication of plasma-derived apo-transferrin, Biotechnol. Appl. Biochem. 57, 87–95, 2010. 242. Saxena, A., Sun, W., Fedorko, J.M., and Doctor, B.P., Prophylaxis with human serum butyrylcholinesterase protects guinea pigs exposed to multiple lethal doses of soman or VX, Biochem. Pharmacol. 81, 164–169, 2011.

44

Biotechnology of Plasma Proteins

243. Lee, J.J., Bruley, D.F., and Kang, K.A., Manipulation of the afinity between protein and metal ions by imidazole and pH for metal afinity puriication of protein C from Cohn fraction IV-1. Adv. Exp. Med. Biol. 614, 93–100, 2008. PubMed PMID: 18290318. 244. Svoboda, M.E., Van Wyk, J.J., Klapper, D.G., et al., Puriication of somatomedin-C from human plasma: Chemical and biological properties, partial sequence analysis, and relationship to other somatomedins, Biochemistry 19, 790–797, 1980. 245. Bala, R.M. and Bhaumick, B., Puriication of a basic somatomedin, from human plasma Cohn fraction IV-1, with physicochemical and radioimmunoassay similarity to somatomedin-C and insulin-like growth factor, Can. J. Biochem. 57, 1289–1298, 1979. 246. Tadepalli, S.S., Bruley, D.F., Kang, D.A., and Drohan, W., Separation of protein C from fraction IV of the Cohn process using immobilized metal afinity chromatography, Adv. Exp. Med. Biol. 428, 639–644, 1997. 247. Wu, H. and Bruley, D.F., Homologous human blood protein separation using immobilized metal afinity chromatography: Protein C separation from prothrombin with application to the separation of factor IX and prothrombin, Biotechnol. Prog. 15, 928–931, 1999. 248. Van der Brande, J.L., Hoogerbrugge, C.M., Beyreuther, K., et al., Isolation and partial characterization of IGF-like peptides from Cohn fraction IV of human plasma, Acta Endocrinol. (Copenhagen) 122, 683–695, 1990. 249. Martin, J.L. and Baxter, R.C., Insulin-like growth factor-binding protein from human plasma. Puriication and characterization, J. Biol. Chem. 261, 8754–8760, 1986. 250. Blum, W.F., Jenne, E.W., Reppin, F., et al., Insulin-like growth factor I (IGF-I)-binding protein complex is a better mitogen than free IGF-I, Endocrinology 125, 766–772, 1989. 251. Miller, F. and Habicht, G.S., Serum-derived immunosuppressive substances. II. An evaluation of various sources for an endogenous regulator of lymphocyte activation, Int. Arch. Allergy Appl. Immunol. 55, 228–238, 1977. 252. Bednarik, T., Cajthamlova, H., Losticky, C., et al., Study on immunosuppressive activity of α-globulin fraction of human blood plasma, Czech. Med. 6, 107–115, 1983. 253. Gorin, E., Gonen, H., and Dickbuch, S., A serum protein inhibitor of acid lipase and its possible role in lipid accumulation in cultured ibroblasts, Biochem. J. 204, 221–227, 1982. 254. Bishop, D.F., Wampler, D.E., Sgouris, J.T., et al., Pilot scale puriication of α-galactosidase A from Cohn fraction IV-1 on human plasma, Biochim. Biophys. Acta 524, 109–120, 1978. 255. Sampaio, C.A. and Grisolia, D., Human plasma kallikrein. Preliminary studies on hydrolysis of proteins and peptides, Agents Actions 8, 125–131, 1978. 256. Jørgensen, C.S., Christiansen, M., Laursen, I., et al., Large-scale puriication and characterization of non-glycosylated Gc globulin (vitamin D-binding protein) from plasma fraction IV, Biotechnol. Appl. Biochem. 44, 35–44, 2006. 257. Christiansen, M., Jørgensen, C.S., Laursen, I., et al., Protein chemical characterization of Gc globulin (vitamin D-binding protein) isoforms: Gc-1f, Gc1a, and Gc-2, Biochim. Biophys. Acta 177, 481–492, 2007. 258. Waugh, W.H., Iron chelation by dibasic amino acid prevents glycoprotein insolubilities: A strategy to inhibit age-related macular degeneration?, J. Appl. Res. 4, 208–214, 2004. 259. Dalton, J. and Podmore, A., Enriched haptoglobin polymers for the treatment of disease, U.S. Patent Application, US20080293623 A1 20081127, 2008. 260. Scatchard, G., Strong, L.E., Hughes, W.L., Jr., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XXVI. The properties of solutions of human serum albumin of low salt content, J. Clin. Invest. 24, 671–679, 1945. 261. Cohn, E.J., Hughes, W.L., Jr., and Weare, J.H., Preparation and properties of serum and plasma proteins; crystallization of serum albumins from ethanol water mixtures, J. Am. Chem. Soc. 69, 1753–1761, 1947. 262. Blombäck, B. and Hanson, L.A. (eds.), Plasma Proteins, John Wiley and Sons, Chichester, 1979.

Plasma Fractionation and Plasma Protein Products

45

263. Putman, F.W., Perspectives, in The Plasma Proteins, 2nd edn., ed. F.W. Putman, Chapter 1, Academic Press, New York, 1975. 264. Dixon, M. and Webb, E.C., Enzymes, 2nd edn., Chapter 2, Academic Press, New York, 1964. 265. Ostlund-Lindqvist, A.M. and Boberg, J., Presence of apolipoprotein-CII in commercially available albumin fractions, Clin. Sci. 56, 99–100, 1979. 266. Berntorp, E., Factor VIII concentrates, in Hemophilia, eds. C.D. Forbes, L. Aledort, and R. Madhok, R., Chapter 17, Chapman & Hall, London, 1997. 267. Fricke, W.A. and Yu, M.Y., Characterization of von Willebrand factor in factor VIII concentrates, Am. J. Hematol. 31, 41–45, 1989. 268. Edsall, J.T., Ferry, R.M., and Armstrong, S.H., Jr., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XI. The proteins concerned in the blood coagulation mechanism, J. Clin. Invest. 23, 557–565, 1944. 269. Ferry, J.D. and Morrison, P.R., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XVI. Fibrin clots, ibrin ilms, and ibrinogen plastics, J. Clin. Invest. 23, 566–571, 1944. 270. Tanaka, K., Sawatani, E., Dias, G.A., et al., High quality of human immunoglobulin G puriied from Cohn fractions by liquid chromatography, Braz. J. Med. Biol. Res. 33, 27–30, 2000. 271. Wickerhauser, M. and Sgouris, J.T., Development of large-scale fractionation method. II. Isolation of a factor IX concentrate (prothrombin complex) for clinical use, Vox. Sang. 22, 137–160, 1972. 272. Andary, T.J., Berkebile, R.L., Thomas, W.R., and Tse, D.C., Therapeutic enzyme concompositions, U.S. Patent 4286056 A 19810825, 1981. 273. Mehl, J.W., O’Connell, W., and Degroot, J., Macroglobulin from human plasma which forms an enzymatically active compound with trypsin, Science 145, 821–822, 1964. 274. Tsuji, A., Oda, R., Sakiyama, K., et al., Lysosomal enzyme replacement using α2-macroglobulin as a transport vehicle, J. Biochem. 115, 937–944, 1994. 275. Peitsch, M.C., Kress, A., Lerch, P.G., et al., A puriication method for apolipoprotein A-I and A-II, Anal. Biochem. 178, 301–305, 1989. 276. Brinkman, N., Bigler, D., Bolii, F., and Foertsch, V., Methods for puriication of alpha-1antitrypsin and apolipoprotein A-I, International Patent, WO2009025754 A2 20090226, 2009. 277. Bednarik, T., Cajthamlova, H., Losticky, C. et al., Immunoelectrophoretic study of the pasteurized alpha-globulin of human plasma, Cesko-Slovenska Farmacie 31, 168–170, 1982. 278. Belew, M., Peterson, E.A., and Porath, J., A high-capacity hydrophobic adsorbent for human serum albumin, Analyt. Biochem. 151, 438–441, 1985. 279. Deutsch, H.F., Kasper, C.B., and Walsh, D.A., Rapid method for preparation of crystalline human ceruloplasmin from Cohn fraction IV-1, Arch. Biochem. Biophys. 99, 132– 135, 1962. 280. Glaser, C.B., Karic, L., and Fallat, R., Isolation and characterization of alpha-1-antitrypsin from the Cohn fraction IV-1 of human plasma, Prep. Biochem. 5, 333–348, 1975. 281. Chen, S.X., Hammond, D.J., Klos, A.M., et al., Chromatographic puriication of human α1-proteinase inhibitor from dissolved Cohn fraction IV-1 paste, J. Chromatogr. A 800, 207–218, 1998. 282. Lujan, H.D., Byrd, L.G., Mowatt, M.R., and Nash, T.E., Serum Cohn fraction IV-1 supports the growth of Giardia lamblia in vitro, Infect. Immun. 62, 4664–4666, 1994. 283. Bignold, L.P., Rogers, S.D., and Harkin, D.G., Effects of plasma proteins on the adhesion, spreading, polarization in suspension, random motility and chemotaxis of neutrophil leukocytes on polycarbonate (Nucleopore) iltration membranes, Eur. J. Cell. Biol. 53, 27–34, 1990.

46

Biotechnology of Plasma Proteins

284. Glasser, L., Fiederlein, R.L., and Huestis, D.W., Liquid preservation of human neutrophils stored in synthetic media at 22 degrees C: Controlled observations on storage variables, Blood 66, 267–272, 1985. 285. Houlgette, R., Wion, D., and Brachet, P., Serum contains a macromolecular effector promoting the synthesis of nerve growth factor (NGF) in L cells, Biochem. Biophys. Res. Commun. 150, 723–730, 1988. 286. Ng, P.K. and Dobkin, M.B., Cell-growth medium supplement, U.S. Patent 4452893 A 19840605, 1984. 287. Abdelnoor, A.M., Harvie, N.R., and Johnson, A.G., Neutralization of bacteria- and endotoxin-induced hypotension by lipoprotein-free human serum, Infect. Immun. 38, 157–161, 1982. 288. Miles, G.L., Taylaur, C.E., and Wilkinson, P.A., Comparison of methods for separating blood plasma lipoproteins, Nature 206, 191–192, 1965. 289. Kaplan, J. and Sunblad, L., Immunochemical determination of β-lipoproteins, Scand. J. Clin. Lab. Invest. 24, 61–68, 1969. 290. Blaton, V. and Peeters, H., Subunits of low- and high-density lipoproteins, Protides Biol. Fluids 16, 707–716, 1969. 291. Oesterreicher, S., Blum, W.F., Schmidt, B., et al., Interaction of insulin-like growth factor II (IGF-II) with multiple plasma proteins: High afinity binding of plasminogen to IGF-II and IGF-binding protein-C, J. Biol. Chem. 280, 9994–10000, 2005. 292. Liu, T.H. and Mertz, E.T., Studies on plasminogen. IX. Puriication of human plasminogen from Cohn fraction-3 by afinity chromatography, Can. J. Biochem. 49, 1055–1061, 1971. 293. Congote, L.F., Extraction of an erythrotropin-like factor from bovine serum albumin (Cohn fraction V), In Vitro Cell Dev. Biol. 23, 361–366, 1987. 294. Jenkins, N., Castro, P., Menon, S., et al., Effect of lipid supplements on the production and glycosylation of recombinant interferon-γ expressed in CHO cells, Cytotechnology 15, 209–215, 1994. 295. Schmid, K., Mao, S.K., Kimura, A., et al., Isolation and characterization of a serinethreonine-rich galactoglycoprotein from human plasma, J. Biol. Chem. 255, 3221–3226, 1980. 296. Kingdon, H.S., Lundblad, R.L., and Dingman, G., Factors affecting the evolution of factor XIa during blood coagulation, J. Lab. Clin. Med. 85, 826–831, 1975. 297. Schneider, D.J., Tracy, P.B., Mann, K.G., and Sobel, B.E., Differential effects of anticoagulants on the activation of platelets ex vivo, Circulation 96, 2877–2883, 1997. 298. Bernheim, B.M., Whole blood transfusion and citrated blood transfusion, JAMA 77, 275–279, 1921. 299. Mantovani, L.G., Monzini, M.S., Mannucci, P.M., et al., Differences between patients’, physicians’ and pharmacists’ preferences for treatment products in haemophilia: A discrete choice experiment, Haemophilia 11, 589–597, 2005. 300. Ehrnberg, H. and Sjoberg, N., Technological discontinuities, competition and irm performance, Technol. Analysis Strategic Manage., 7, 93–107, 1995. 301. Vamvakas, E.C., The relative safety of pooled whole-blood-derived platelets prepared by the buffy-coat method versus single-donor (apheresis) platelets, Clin. Lab. 56, 263– 279, 2010. 302. Shelet, S.G., Practical considerations for planning a therapeutic apheresis procedure, Am. J. Med. 123, 777–784, 2010.

3

Methods for the Analysis of Plasma and Plasma Protein Fractions

There are two different aspects of analytical biochemistry that can be applied to the characterization of plasma and plasma protein fractions. The irst is the type of characterization used in the biochemical and biophysical characterization of proteins as practiced in the academic departments of biochemistry and chemistry.* The second is the type of analytical science used for the assay of plasma proteins during the commercial puriication and product release. It has been acknowledged that these two areas overlap, but I would posit that assays are developed in basic science departments and then used for biopharmaceutical products. Mass spectrometry is an example that is commonly applied to the characterization of recombinant proteins but infrequently, if at all, in the plasma business.† Early methods for the characterization of blood plasma were irst based on electrophoretic methods1–3 as developed by Tiselius.4–8 The development of the electrophoretic analysis of protein is the subject of an excellent review9 by Righetti, which was published in 2005. Tiselius developed moving-boundary or free-boundary electrophoresis, in which colloids such as proteins migrate in free solution. In his original application, Tiselius used optical methods to measure the protein boundaries.10 This technique required substantial protein concentration but could provide information on macromolecular interactions without matrix interference.11 Optical methods are also used to measure the boundaries in analytical ultracentrifugation.12–14 A variation of free-boundary electrophoresis has been used to purify intravenous immunoglobulin (IVIG) from plasma.15 Free-boundary electrophoresis was initially replaced by zonal electrophoresis,16 irst on paper,17 then on starch,18 later on agarose,19 and today by capillary electrophoresis as described later in this book. Hjertén20 introduced a rotating column *



The question is whether an analytical result is relevant to the safety and eficacy of a product. While a inding may be of great scientiic interest, such a result may not be relevant to the critical product attributes. An example is provided by the carbohydrate analysis of human factor VIII (see Chapter 6), where there are differences in glycosylation patterns between the protein product in Chinese hamster ovary (CHO) cells and that produced in baby hamster kidney (BHK) cells, both of which are different from the native human protein. Furthermore, Schiestl and coworkers412 reported signiicant changes in the chemical properties of several approved protein drugs likely resulting from changes in the manufacturing process. Such changes were not associated with label changes and all products remained on the market, suggesting that the changes were considerably acceptable by the various regulatory agencies. While mass spectrometry is not, to the best of my knowledge, used in the production and the release of human serum albumin, this technique is used in the current studies of albumin, such as glycation.155

47

48

Biotechnology of Plasma Proteins

for electrophoresis for the reduction of zonal instability, which was later commercialized as the Rotofor by BioRad,21 and it has been in steady use in preparative isoelectric focusing.22–26 More advanced technologies combining electrophoresis, isoelectric focusing, chromatography, and mass spectrometry are being used for the proteomic analysis of blood plasma and serum; these technologies will be discussed later. Electrophoresis of intact serum is still used in clinical chemistry27 mostly for the detection of monoclonal gammopathies.28 Early work by Tiselius4,5 resulted in the veriication of the albumin and globulin components of blood serum and the separation of globulins into α-, β-, and γ-globulins.3 The terms albumin and globulin were introduced in the 1800s on the basis of the solubility differences between the two groups of proteins.29 The globulin fraction was further separated into various fractions such as pseudoglobulin and euglobulin, again by differing solubility characteristics.30 The work of later researchers demonstrated the heterogeneity of these fractions.31 The application of more advanced analytical technologies, as described later, demonstrated an even more complex picture of human blood plasma. Free-low electrophoresis is similar to free-boundary electrophoresis in the absence of a supporting matrix such as paper, starch, or polyacrylamide gel. Briely, free-low electrophoresis is a continuous system in which a sample is introduced into a buffer solution that lows between two plates in an electric ield, and proteins, subcellular organelles, and cells are separated on the basis of particle charge.32–35 This technology has been extensively used for subcellular organelles and cells.36–41 Free-low electrophoresis is of value for the prefractionation of cells, subcellular organelles, and polyprotein complexes prior to proteomic analysis.42,43 Free-low electrophoresis has also been applied to proteins.44–48 A commercial apparatus for analytical free-low electrophoresis is available from BD-diagnostics.49 A free-low system that is capable of therapeutic fractionation has been developed by Gradipore.50 It is possible to operate free-low electrophoresis as zone electrophoresis, isotachophoresis, and isoelectric focusing.51 Free-low electrophoresis appears to be more useful for sample preparation than for analysis. Given the absence of potential matrix effects, free-low electrophoresis might be useful for the study of protein–protein interactions and other biopolymer–ligand interactions. An example is provided by the study on the interactions of aptamers with protein targets (IgE, human immunodeiciency virus reverse transcriptase) using gradient micro free-low electrophoresis.52 Another example is the study of the binding of follicle-stimulating hormone (FSH) to solubilized plasma membrane receptor.53 Fieldlow fractionation, which has both analytical and preparative capabilities, is a more recent application of free-boundary electrophoresis.54–56 A number of new separation and analytical technologies were developed between 1945 and 1975, including practical ion-exchange chromatography for proteins, gel iltration, polyacrylamide gel electrophoresis (PAGE), scanning UV-VIS spectrophotometers, and analytical ultracentrifugation. However, compared to 2011, the laboratory research in protein separation and characterization was primitive and required substantial amounts of proteins. As a point of reference, consider the contents of a book on analytical techniques for plasma proteins57 that contained chapters on the measurement of protein concentration, electrophoresis, immunoprecipitation, immunoelectrophoresis, radioimmunoassay, analytical isoelectric focusing, and

Methods for the Analysis of Plasma and Plasma Protein Fractions

49

molecular weight determination.* These techniques are old, but they are still useful for the manufacture of plasma-derived therapeutic proteins. There may be more sophisticated instrumentation, but the biochemical and immunochemical principles underlying the assay remain the same. Another issue is the deinition of plasma protein. In 1975, Putnam58 discussed the question of what a plasma protein is. Among the criteria mentioned are primary function in the vascular space, synthesis in the reticuloendothelial systems or liver rather than endocrine tissue, actively secreted into blood rather than as a result of tissue damage, presence in plasma after the neonatal period, and an appreciable circulatory half-life (Chapter 1). Putnam also noted the dynamic range of plasma proteins. Thus, in 1975, Frank Putnam described 53 well-characterized proteins from plasma.58 Several years later, Heide and coworkers59 listed 100 well-characterized plasma proteins. About a decade later in 1984, Putnam60 listed a further increase in the number of plasma proteins but observed problems posed by the new functions ascribed to the old names, isoforms, and posttranslational modiications. More than three decades later, Anderson and coworkers61 compiled a list of 1,175 distinct gene products in the plasma proteome. This list was obtained by merging the data obtained by four different methods, but, as the authors note, it may not contain lowabundance proteins such as cytokines and protein/peptide factors such as S100B.62 It is estimated that there are some 21,000 gene products in the human proteome.63 Considering the dificulty involved in the validation of biomarkers,64 it is not surprising that there are only approximately 200 assays available for the discrete components in plasma or serum.65 It is granted that the issue of identiication as a plasma protein is of importance only in the analysis of intact plasma and/or serum. The primary goal of this book is to discuss the biotechnology of the plasma proteins with an emphasis on the protein therapeutic products that can be derived from human blood plasma. Notwithstanding the current interest in biomarkers, I am interested in these proteomic approaches only as those that directly address the blood-derived biopharmaceutical products.66–68 This will be discussed in greater detail later. That leaves, then, the consideration of various assays that are of value in the development and manufacture of plasma biopharmaceutical products. As an aside, the term biopharmaceutical has no regulatory or true scientiic meaning and is loosely considered to be a drug substance that is produced by biotechnology but has gained acceptance in the greater pharmaceutical community.69–72 The reader is directed to Rader’s analysis of biopharmaceutical73 for a more thorough consideration of this issue. The point is, at least according to this author, that the term biopharmaceutical is mostly useless as a descriptor in the current biotechnology world; thus, this work is concerned with the protein therapeutic products derived from plasma. The determination of the activity and the structure of a protein therapeutic product is a critical activity in the development, manufacture, and use of such a material. At the onset, the actual measurement is but one step in the analytical process. The reader is directed to an excellent chapter by Chris Burgess on analytical quality *

These researchers used a recombinant factor manufactured by Baxter Healthcare and obtained from a hemophilia treatment center. It is assumed that the inal drug product, Recombinate, was used in these studies instead of an active pharmaceutical ingredient (API).

50

Biotechnology of Plasma Proteins

management.74 The history of a sample used for the measurement of the activity is of particular importance for samples that may have been retained from production lots and for samples obtained from the recipients of products.75,76 The use of DNA proiling permits the unambiguous identiication of blood/plasma/tissue samples from human subjects.76,77 The measurement of the activity of a biological product is one of the several critical quality attributes that may be used to follow a manufacturing process as well as the activity of the API and inal drug product. The measurement of activity is also one of the several quality attributes that are used to evaluate the stability of the inal drug product. Before I leave activity in the discussion of activity measurement (activity measurement will be discussed in greater detail later with regard to speciic proteins), I would like to note that there is no real activity measurement for albumin, no real measurement for the immunomodulation activity of IVIG, and the measurement for factor VIII activity is problematic.78 Additional details on the development and validation of assays are described later. The chemical and physical characterizations are performed for several reasons, including (1) fuse in the manufacturing process to demonstrate that the process produces a reproducible product, (2) to gain regulatory approval for the marketing of the product, and (3) to provide information to the responsible health-care provider. Within the context of the current book, chemical characterization refers to the determination of the chemical structure of a protein, which would include primary structure (amino acid sequence), alignment of disulides, and determination of posttranslational modiications including glycosylation and glycation (if present). A iner analysis might include the determination of oxidation and the formation of β-aspartic acid. Physical characterization includes those techniques used to measure protein conformation.79 Publications should arise from such activities and, assuming that a researcher can sneak some from the legal department, such publications can provide support for the approval and reimbursement by third-party payers.80–85 The instrumentation available for the physical and chemical characterization of proteins in 2011 is impressive including mass spectrometry,86–95 which has largely supplanted the classical approaches to protein structure that involve the chemical and enzymatic fragmentation of a protein followed by the use of the Edman degradation to determine an amino acid sequence. A combination of mass spectrometry with chromatography or electrophoresis provides an elegant and sensitive approach to both the determination of structure and the establishment of purity for proteins. It is then somewhat unfortunate that most, if not all, plasma proteins exhibit some degree of heterogeneity. The heterogeneity may be minor as in the oxidation of the sulfhydryl group in albumin96 or from glycation.97 The glycosylation of proteins is a complex process resulting in O- and N-linked glycan chains. Thus, it is a bit surprising that there is remarkably little variation in N-glycan proiles in a normal population,98 and heterogeneity in the glycosylation of plasma proteins is observed mostly in certain disease states.99–102 Considering the great interest103 regarding the importance of glycosylation as a critical product attribute in the biosimilar area,104–106 it is surprising that, with the exception of the various blood coagulation factors of therapeutic importance, there is little work on the glycosylation of human plasma proteins in the peer-reviewed literature.107,108 Bruce Mackler notes that the concern in the biosimilar area is that glycosylation differences

Methods for the Analysis of Plasma and Plasma Protein Fractions

51

between the branded product and the biosimilar product may present a major hurdle to the approval of a biosimilar product.103 However, Mackler also notes that this is an opportunity for solid science to make the case for the importance of such differences. Speciically, it is important to understand that differences in glycosylation may or may not inluence product safety and eficacy. The structure and function of glycan chains on several of the blood coagulation factors, including factor VIII, factor IX, protein C, and factor VII/VIIa, have been the subject of investigation (see Chapters 6 and 8, for more details). The interest here is conceptually related to the biosimilar problem mentioned earlier in that there was great interest in understanding any differences between the protein isolated from plasma and that produced by recombinant DNA technology. In the case of factor VIII, the glycosylation pattern of the recombinant form produced in CHO cells was different from that observed with the material produced in BHK cells, both of which differed from native factor VIII.109,110 The differences in glycosylation do not appear to be related to any differences in recovery, circulatory half-life, or hemostatic effectiveness as judged by comparative clinical studies.111 Fay and coworkers observed that the removal of approximately 50% of the carbohydrate from plasma-derived factor VIII with a mixture of exoglycosidases and endoglycosidases had no effect on activity or circulatory half-life.112 In more recent studies, Kosloski and coworkers113 subjected recombinant factor VIIIc to a mixture of endoglycosidases114 and observed a decrease in its biological activity (one-stage partial thromboplastin time) but no change in immunogenicity. There are two studies, one somewhat older, that I have found quite useful in placing the glycosylation in perspective. The irst study, by Baynes and Wold115 published in 1976, examined the role of glycan in the circulatory half-life of bovine pancreatic ribonuclease (RNase) in normal and nephrectomized rats. RNase A contains no carbohydrate; RNase B possesses a single, simple N-linked oligosaccharide, while RNases C and D possess a single, complex N-linked oligosaccharide (biantennary [two sialic acids] in the case of C and tetraantennary [four sialic acids] in the case of D). All four RNase species are cleared quite rapidly in normal mice, consistent with the role of the kidney in the clearance of small proteins. RNase A is cleared quite slowly (9–10 h) in the nephrectomized rat, RNase B is cleared in 15 min, and RNases C and D are cleared in 11–17 h. The removal of the terminal mannose residues from RNase B extends the clearance to that observed in the case of RNase A. These researchers concluded that an increase in the complexity of glycosylation did not affect the circulatory half-life and that the exposure of mannose was the determining factor in clearance. The late Finn Wold was one of the premier protein chemists in the latter part of the twentieth century and a cofounder of The Protein Society, while John Baynes has been a leader in the characterization of glycoproteins. More recently,116 Skropeta presented an excellent review on the role of glycans in the biological activities of glycoproteins. First, glycosylation appears to have a major role in the secretion of proteins. The author is also familiar with a number of unpublished experiments on the importance of glycosylation in the expression of recombinant proteins from mammalian cell hosts, such as CHO cells or BHK cells. Skropeta also reviewed the diverse effects of glycoengineering on the biological activity of enzymes. This is a superb review article that also covers the effects of glycosylation on stability.

52

Biotechnology of Plasma Proteins

The study of blood plasma for the purpose of identifying as-yet undescribed proteins and the characterization of “new” and “old” proteins can be labeled plasma proteomics, where such a term would imply the study of all plasma proteins. A quick PubMed search yielded approximately 1,200 citations for the phrase “human blood plasma proteomics,” of which a vast majority of citations describe the search for a biomarker, while a much smaller number of citations address the characterization of plasma and/or serum.117–126 I have previously noted64 that the majority of studies on the identiication of biomarkers use blood plasma or serum as the source. We have previously commented127,128 on distinguishing between serum and plasma as a sample for clinical analysis and for the preparation of samples for proteomic analysis.129 A recent study by Siev and coworkers130 reports that serum and plasma can be used interchangeably for antibody detection assays for mycobacterial antigens but not for other antigens in which there are differences between plasma and serum samples. Serum or plasma can be used, depending on the biomarker, but the sample source and processing must be clearly stated. Proteomics can be described as the study of all of the proteins, and thus, plasma proteomics is the study of plasma proteins. This is admittedly redundant with the previous comment but is meant to reemphasize that proteomics is an area of study and not a technical approach. As noted by the author,129 protein chemistry has evolved into proteomics. Thus, for all practical purposes, there are no unique laboratory techniques in proteomics but rather the adaptation of classical solution protein chemistry to the study of small samples. The techniques in proteomics include chromatography, electrophoresis, mass spectrometry, and bioinformatics. There is no question that the combination of mass spectrometry and bioinformatics is the dominant factor in proteomics. Proteomics has been used in the identiication and characterization of “new” proteins in tissues in biological luids with the goal of identifying biomarkers that are useful in the diagnosis and prognosis of a disease with an emphasis on cancer. There are several studies that use proteomics for the characterization of products in transfusion medicine including therapeutic proteins.131–135 As stated earlier, proteomics is an area of study and not a technical approach. Two of these studies132,134 use a prefractionation step, followed by electrophoresis and/or chromatography, enzymatic fragmentation, and mass spectrometric analysis. Bioinformatic techniques are used to interpret the mass spectrometry results and allow the identiication of the proteins from a database.136,137 This is a classical approach to the identiication of protein biomarkers.64 Two of the other studies133,134 use the process of digestion of samples with proteases followed by the separation of peptides using high-performance liquid chromatography (HPLC) and analysis by mass spectrometry. One of the studies uses proteomic technology to study prothrombin complex concentrates.131 While this product seems to have been bypassed by a combination of technology and marketing, a prothrombin complex concentrate that contains factor VII is of considerable value.136–141 This study on the prothrombin complex concentrates131 provides information on the composition of this material, which is not available from the use of earlier technologies and will be most useful in establishing quality attributes for the processing of derivative products such as factor IX, antithrombin, or α1-antitrypsin. The proteomic technologies will be most useful in identifying the contaminants and/or impurities in

Methods for the Analysis of Plasma and Plasma Protein Fractions

53

plasma protein products, but of less value in ensuring the product identity when compared to other available technologies such as activity determination, immunological assays, spectroscopy, and analytical ultracentrifugation.79 Several of the techniques used in proteomics, such as mass spectrometry, HPLC, and electrophoresis, are of individual value in the characterization of plasma protein fractions. Mass spectrometry is the most important tool in modern protein chemistry, with approximately 60,000 citations on PubMed and a similar number on SciFinder. In the case of proteins, the direct applications of mass spectrometry include the assessment of chemical modiications, the determination of hydrogen/deuterium exchange, and the study of protein–protein interactions. There is a much larger application of mass spectrometry in the identiication of proteins as biomarkers.64 The current work is directed toward the description of the technology that has direct importance in the biotechnological development of plasma proteins and, as such, will focus on the application of mass spectrometry in the development of plasma protein preparations as biopharmaceuticals. The characterization of the posttranslational modiication of plasma proteins is of critical importance in the characterization of such proteins as biopharmaceutical products and for setting a standard as a “branded” product for the qualiication of recombinant products.142–153 The major posttranslational modiication (see above) is glycosylation in that most plasma proteins, with the exception of albumin, have signiicant glycosylation; albumin does undergo glycation.154–156 Other posttranslational modiications include γ-carboxylation of glutamic acid, O-sulfation of tyrosine, β-hydroxylation of aspartic acid, and transpeptidation. Mass spectrometry has been used to characterize the γ-carboxylation of proteins.157 Several plasma proteins of therapeutic importance contain γ-carboxyglutamic acid, but I could not ind any direct use of mass spectrometry for the characterization of this modiication in factor IX, factor VIIa, or activated protein C. Mass spectrometry is used for the characterization of the glycan moieties present in these proteins.158–162 There is also considerable literature on the use of mass spectrometry to study glycans in other proteins of interest.163 The problem of pathogens in the plasma supply for fractionation was discussed in Chapter 2. Mass spectrometry can be used to establish the presence of viral and bacterial pathogens164–168 and could be of value in establishing the presence of such materials in the active pharmaceutical intermediate. Mass spectrometry has been used to determine the presence of contaminants in the prothrombin complex concentrate131 and factor VIII preparations.169 Ahrends and coworkers170 used mass spectrometry to characterize the proteins in the Cohn Fraction IV-4. These researchers also compared displacement chromatography with gradient chromatography for the separation of tryptic peptides prior to mass spectrometry (electrospray ionization). Thiele and coworkers66 used proteomics to evaluate the potential changes in plasma due to viral inactivation with solvent/detergent or methylene blue/light. Some changes were observed at higher methylene blue concentrations. These researchers also used proteomic technologies to evaluate quality in red blood cell concentrates and platelet concentrates. It would appear that the greatest use of mass spectrometry is in the detection of contaminants and impurities in either an API or a inal drug product. A recent example is the application of mass spectrometry for the characterization of heparin.170–176 The author recognizes that heparin is not a plasma protein but its therapeutic activity is coupled to a plasma

54

Biotechnology of Plasma Proteins

protein, antithrombin (Chapter 8). In the case of heparin, given the small size and relative lack of complexity, the combination of nuclear magnetic resonance (NMR) and mass spectrometry establishes the correct structure(s) and the presence of impurities such as oversulfated chondroitin sulfate.177 Chromatography is a term used to describe the separation of materials on a matrix. The terms chromatogram and chromatography were introduced by Tsweet in 1906 for describing the separation of various colored compounds derived from plants; in Tsweet’s studies, a green material, chlorophyll, was separated from a yellow compound, a carotenoid.178 Over the ensuing 50 years, various developments took place,179–181 but it was not until the development of cellulosic ion-exchange matrices by Sober and Peterson182 and gel iltration by Jerker Porath and Per Flodin183 that chromatography was of practical value for protein puriication. It took some time though for these materials to become readily available from commercial sources; the author was still preparing carboxymethyl cellulose by the reaction of chloroacetic acid and cellulose in 1963 in a scene similar to the opening moment in Macbeth. A few years later, I was in the laboratories of Stanford Moore and William Stein at the Rockefeller University in New York where I could perform two amino acid analyses per day. I recall complaining to Professor Moore about the slow performance, and he pointed out that the current method was a lot better than the several weeks required for bacterial assay for amino acids, which was the state-of-the-art technique when he and Bill started their work in the late 1930s. Ion-exchange resins such as Dowex and Amberlite were introduced following World War II and found considerable use in the industrial sector for water puriication and chemical processing. Attempts to use these cross-linked polystyrene/polyacrylate resins for protein puriication were mostly unsuccessful because of the irreversible binding to the matrix (see below). HPLC is routinely used to evaluate the purity of proteins. Having said that, it must be recognized that there are many types of HPLC, including the ultrahighperformance liquid chromatography (uHPLC).184 HPLC is really a term used for the instrumentation rather than a speciic separation technology in which a solvent is pumped through a matrix consisting of very small particles (usually silica). That said, the term HPLC is most often used for the chromatography on a matrix, where hydrocarbon chains are bound to a matrix; this is called reverse-phase chromatography. Size-exclusion chromatography (SEC) can separate proteins (and other molecules) on the basis of hydrodynamic radius, while ion-exchange matrices separate proteins on the basis of charge. In the absence of a speciic molecular interaction as with afinity chromatography or immunoafinity chromatography, the separation of proteins is based on the use of size or charge. HPLC is a plumbing and detection system that can be used with a variety of chromatographic matrices such as ion-exchange, size-exclusion, reverse-phase, and afinity. The matrices must have the physical characteristics provided by particle size and stability to provide an acceptable low rate and resolution at high pressure. The generic advantages provided by the HPLC technology are analysis speed and sensitivity. HPLC and uHPLC are used primarily for the analysis and characterization of plasma protein therapeutics. Conventional chromatography uses matrices that are similar to those used for HPLC; the dimensions of the chromatographic columns used in the commercial

Methods for the Analysis of Plasma and Plasma Protein Fractions

55

biotechnology are quite different from those used for HPLC, frequently looking like relatives of R2D2 from Star Wars. They are operated at much lower pressures than HPLC or uHPLC systems. Chromatographic columns, with the exception of afinity matrices, can be operated in either a gradient mode or an isocratic mode.185 The isocratic mode is much easier to use in a manufacturing setting than a gradient system. Ion-exchange chromatography is used for both the puriication and the characterization of biopharmaceutical proteins.186–200 As noted in Chapter 2, chromatography is used only infrequently in plasma protein puriication for the manufacture of biopharmaceuticals. It is used more extensively for the puriication and characterization of therapeutic proteins produced by recombinant DNA technology. One problem is that most plasma proteins as well as recombinant proteins tend to exhibit some heterogeneity when subjected to chromatography on the current high-resolution ionexchange matrices. Johnson and coworkers200 observed heterogeneity in a monoclonal antibody where isoforms differed in that one heavy chain contained an amidated proline residue. The most common intrinsic heterogeneity in plasma proteins is a difference in glycosylation resulting in multiple glycoforms.201,202 Heterogeneity is also derived during processing, resulting in oxidation199 and succinimide formation.188,192,193 Wang and coworkers199 showed that the conformation inluenced the separation of an oxidized monoclonal antibody (t-butyl hydroperoxide) on cationexchange chromatography. While chromatography at pH 5.5 provided a minor resolution of the oxidized antibody from the native proteins, chromatography at pH 4.0 provided a signiicant resolution; the oxidized antibody is also separated from the native protein by afinity chromatography on a protein A column. It is suggested that a difference in the protein conformation is responsible for the resolution of the oxidized and native proteins. The conformational change has been suggested to be responsible for the change in chromatographic behavior of prothrombin in the presence and absence of calcium ions.203 Other studies on the inluence of conformation on the behavior of proteins on ion-exchange chromatography include the early studies on the effect of urea on the behavior of bovine albumin on anion-exchange chromatography.204 In these studies, Withka and coworkers showed that the loss of native bovine serum albumin structure in the presence of urea could be measured by the changes in behavior on a diethylaminoethanol (DEAE) matrix. These researchers also studied the effect of conformation on the behavior of proteins on SEC where a more rapidly migrating species was observed on denaturation with urea (4.8 M); oddly enough, there was little change in the amount of dimer. These researchers also studied the effect of urea denaturation on the cation-exchange (sulfonated matrix) chromatography of trypsin and lysozyme. Urea had a biphasic effect on the chromatography of trypsin with an increase in retention at low (0.5 M) urea concentrations while a decrease in retention at higher urea concentrations. There is a decrease in retention for lysozyme at all urea concentrations. Similar results for lysozyme were reported by Yamamoto and coworkers.205 Urea was not present in the chromatography solvent in these experiments; the samples were treated with 8.0 M urea. Cole had previously observed the necessity for the inclusion of urea in the chromatography of proteins on ion-exchange resins; in the absence of urea, proteins were irreversibly adsorbed onto ion-exchange resins.206 Voitl and coworkers207 observed that human serum albumin was resolved into two peaks on chromatography on a strong

56

Biotechnology of Plasma Proteins

cation-exchange matrix (sulfoethyl). Rechromatography of either the major or the minor peak resulted in the formation of two fractions, indicating an equilibrium process in the sample. Only a single peak was observed on a carboxylate matrix. Chromatography was performed at pH 5.0, below which a pH transition would be expected. Experiments with monoclonal antibodies208 do not suggest a conformational change with unmodiied proteins or in the absence of ligands. A further analysis by Voitl and coworkers209 supports the hypothesis that there are two conformations produced by an interaction with the strong cation-exchange matrix. Thus, in these studies, the matrix creates an apparent heterogeneity. Hou and coworkers210 showed that the protein conformation does inluence the behavior of a protein on ionexchange matrices, but such changes are complicated and dependent on the protein. Ideally, the interaction between a protein and a charged matrix would depend on the net particle charge on the protein. Surface charges are not distributed equally, and “patches” do exist where positively charged, negatively charged, or neutral/hydrophobic amino acids would be clustered together on the surface of a protein. In the case of thrombin, the anion-binding exosites211 are likely responsible for binding to the cation-exchange matrices.212,213 SEC refers to a technique that separates molecules on the basis of hydrodynamic volume (Stoke’s radius) using a cylindrical column with laminar luid low. There are two “types” of SEC: gel permeation chromatography is taken to mean the separation of organic compounds in apolar solvents,214 while gel iltration is taken to refer to the separation of materials in aqueous solvent systems.215–219 Gel iltration (most frequently referred to as SEC) is commonly used in the study of proteins and peptides, while there is little use of gel permeation chromatography in the protein chemistry laboratory. Gel permeation is used extensively in polymer chemistry. Gel iltration is commonly assumed to separate proteins on the basis of size and is used most frequently in the assessment of the aggregation of protein APIs in the inal drug product.220–225 It is recognized that analytical ultracentrifugation does provide the “gold standard” for assessing protein aggregation.226–229 It is noted that albumin (see Chapter 4) is provided as a solution that has undergone pasteurization, and visual inspection229 for particulate material is used in addition to gel iltration.205 Matrix effects cannot be ignored in SEC.231–236 Tarvers demonstrated an apparent decrease in the molecular weight of prothrombin in the presence of calcium ions as determined by gel iltration.237 Lundblad238 observed that the behavior of human prothrombin changed in the presence of calcium ions, suggesting the exposure of a hydrophobic patch that bound to the matrix and, thus, the decreased molecular weight of prothrombin observed by Tarvers in the presence of calcium ions. An interaction of aromatic compounds with dextran gels (Sephadex) has been known for some time.239 Martenson presented an excellent review on the use of gel iltration to study conformational changes.240 A conformational change that increases the axial ration of proteins will change the gel iltration behavior.241,242 It is strongly recommended that a change in behavior on gel iltration suggesting a change in size be validated by analytical ultracentrifugation. Hydrophobic interaction chromatography (HIC)243–246 is used for the puriication and characterization of proteins. It is useful because HIC, as with gel iltration, offers a method that is orthogonal to ion-exchange technique.247 There is great interest in

Methods for the Analysis of Plasma and Plasma Protein Fractions

57

the use of HIC in manufacturing biopharmaceuticals.248,249 I do not know of any direct application to plasma proteins. Extracorporeal HIC has been used in combination with IVIG in the treatment of autoimmune disease.250 HIC is used for the puriication and characterization of immunoglobulins251–253 and albumin.254–257 It has been suggested that proteins undergo conformational changes on binding to HIC matrices.258–262 The binding of the protein to the HIC matrix can result in denaturation, which, in turn, is dependent on mobile-phase modiiers.263 The effect of modiiers on the behavior of α-lactalbumin on HIC has been used as a model system. Wu published a well-cited paper in 1986264 on the effect of protein conformation on HIC with an emphasis on the mobile-phase composition. These researchers showed that α-lactoglobulin underwent a temperature-induced conformation change on an HIC column resulting in two peaks, the latter eluting and growing at the expense of the early peak; calcium ions stabilized α-lactoglobulin, while the presence of magnesium ions resulted in destabilization. A more recent work by Jones and Fernandez265 conirmed the presence of two peaks in the HIC of α-lactalbumin and showed by hydrogen-exchange that the second peak was mostly unfolded. There are a number of other studies on the inluence of protein conformation on binding to HIC matrices.266–273 One useful example is the increased binding of human prothrombin to an HIC matrix in the presence of calcium ions.238 This type of binding has been referred to as pseudoafinity chromatography by Yan.274 The metal ion–dependent binding of vitamin K–dependent proteins to antibodies is well known and was used by Smith275 to purify factor IX. The use of p-chlorobenzylamido-agarose to purify thrombin276 is an interesting example of what appears to be speciic hydrophobic afinity chromatography. Hydrophobic afinity chromatography has the potential to be used in biopharmaceutical manufacturing,277 while the speciicity of metal ion– induced changes in chromatographic behavior has the potential to be used as a surrogate measure of function for proteins, such as factor IX, where the function is dependent on calcium ions. Afinity chromatography, most notably immunoafinity chromatography, has been considerably used for the puriication of recombinant proteins. Afinity chromatography is based on a biologically signiicant interaction between the solute and the speciic binding ligand on the matrix.278 The classic model used a competitive inhibitor bound to a matrix such as the use of amidine derivatives in the afinity chromatography of trypsin.279 Immunoafinity chromatography is based on the interaction of antibodies and speciic epitopes on an antigen. Most often, an antibody is used to purify an antigen, but an antigen can also be used to purify an antibody. An example of the latter is the use of autoantibodies to identify speciic antibodies in polyclonal antibody preparations. Immunoafinity chromatography has been used to purify blood coagulation factor VIII from plasma for therapeutic use.280–284 A speciic immunoglobulin G (IgG) population responsible for the immunosuppressive effect of IVIG in autoimmune myasthenia gravis can be obtained from bulk IVIG by immunoafinity chromatography.285 The choice of an antibody for immunoafinity chromatography is not trivial. Early work with polyclonal antibodies required drastic conditions for elution because of the high afinity of polyclonal binding; the development of monoclonal antibodies, the use of batch binding for the selection of an antibody, and the conditions for application and elution made immunoafinity

58

Biotechnology of Plasma Proteins

chromatography a more reasonable choice for use with labile plasma proteins, such as factor VIII.286–289 While batch evaluation using enzyme-linked immunosorbent assay (ELISA) systems was (and is) useful for antibody selection and the optimization of binding and elution conditions,290–292 surface plasmon resonance technology may be more rapid.288,293 It is possible to achieve immunoafinity puriication using batch ELISA technology on polystyrene microplates.294,295 As a general consideration for immunoafinity chromatography, it is useful to have a speciic, “weak” antibody for immunoafinity chromatography such that drastic conditions are not required for the elution of the material of interest. There is a different critical attribute for an antibody used for capture in an ELISA assay where high afinity is a desirable quality. Thompson and coworkers296 report a method for the use of ELISA-elution technique to select polyol-sensitive monoclonal antibodies for use in immunoafinity chromatography. There are other examples where ELISA-elution technique has proved useful for antibody selection for immunoafinity chromatography.297–300 Antibodies are the most common biospeciic ligand used for afinity chromatography of plasma proteins. However, there are other speciic ligands that have proved useful. Heparin is likely the best example. Heparin binds to a substantial number of plasma proteins,301,302 but it binds more tightly to antithrombin. Antithrombin can be prepared from plasma by afinity chromatography on heparin–agarose.303–307 Afinity chromatography is rarely used for characterization. IgG can be puriied by afinity chromatography on protein A308,309 or protein G.310,311 Lectin afinity chromatography can be used to evaluate glycosylation quality.312 Lectins may also be used as probes in microarray technology for the characterization of glycosylation.313 Lectin afinity chromatography may also be used for the puriication of IgA and IgM immunoglobulins.314–316 Lectin binding has been used to monitor the in-process glycosylation during biopharmaceutical manufacturing.317 Mass spectrometry, however, remains the method of choice for the characterization of glycosylation.318 Electrophoresis is used extensively to demonstrate the purity of protein preparations. While electrophoresis may be performed in the absence of a supporting matrix, as discussed for free-boundary electrophoresis earlier, this technique is usually performed with a supporting matrix. Capillary electrophoresis is also an example of zone or free-boundary electrophoresis. Capillary electrophoresis offers the advantages of speed and sensitivity—attributes of importance when the sample volume is limited.319 While there is some suggestion that capillary electrophoresis suffers from the problems of sensitivity and reproducibility,320,321 this technique has been found useful for the characterization of biopharmaceutical proteins.322–327 As with the other separation technologies, capillary electrophoresis can be coupled with mass spectrometry for analysis.328 Polyacrylamide matrices are usually used for proteins; agarose matrices may be used for very high-molecular-weight proteins such as the von Willebrand factor.329,330 PAGE is used in the presence or absence of sodium dodecyl sulfate (SDS); when used in the presence of SDS, the technology is referred to as sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). In principle, the detergent denatures the protein and binds to the denatured protein to yield a negatively charged particle that will migrate in a cross-linked polyacrylamide gel with a velocity inversely proportional to the molecular weight. SDS-PAGE is frequently used to demonstrate protein purity,331 but can also be used to measure a functional attribute such

Methods for the Analysis of Plasma and Plasma Protein Fractions

59

as the cleavage of factor VIII by thrombin.332,333 SDS-PAGE can yield misleading results such as heterogeneity induced by sample processing.334 Proteins with extensive glycosylation stain poorly with Coomassie blue stains.335–337 There is a good recent article on staining for proteomic research that is quite useful338 as well as a review by Steinberg.339 This techniques is, as mentioned earlier, used primarily for the characterization of the purity of the inal product. Native gel electrophoresis is a method of electrophoresis in a polyacrylamide gel (or other matrix or under free-boundary conditions); native gel electrophoresis is most often used to characterize the interaction of proteins with other proteins or nucleic acids.340,341 Blue native electrophoresis is a technique that has been used to study membrane protein complexes.342–344 The use of blue native electrophoresis preserves the native state of the protein, permitting the use of conformationally sensitive antibodies345 and assays for enzymatic activity.346 Solid-phase-speciic binding assays are used extensively in the study of plasma proteins. The assay is based on the immobilization of a speciic binding agent to a matrix such as a microplate, capture of the analyte by the immobilized binding agent, and detection of the bound analyte most often by a monoclonal antibody coupled to a signal generation system such as peroxidase. Immunoassays are based on the “speciic” interaction between an antibody and an epitope on the analyte. The basic approach involves immobilizing an antibody in a microplate, adding the sample, and then using another antibody linked to a signal that can be measured and used as a measure of the amount of analyte captured by the antibody. This type of process can be used in other binding assays as described by Englebienne.347 Examples that substantiate the importance of the study of plasma proteins include the collagen binding of the von Willebrand factor,348,349 the use of immobilized ibrinogen to bind C1q and gC1q-R,350 and the use of immobilized glycoconjugates to measure glycan-binding proteins.351 The binding of proteins such as ibronectin352 and ibrinogen353 can result in the exposure of neoantigens. A pull-down assay354 is a variation of a solid-phase binding assay, which can be used for the study of protein–protein interactions.355,356 The most common binding assay used in plasma protein biotechnology is the immunoassay. The most common immunoassays are the ones that are based on solidphase interactions such as ELISA, but solution-phase assays such as nephelometry are still used in some cases.357–363 Immunoelectrophoresis is used for the diagnosis of von Willebrand’s disease.364 Rocket immunoelectrophoresis365 has been used to measure prothrombin366 and serpins367 in plasma. Many of these older methods may still be used in the quality control of some of the older plasma protein therapeutics such as albumin. It is dificult to meet the cost of developing and validating a new assay to replace an existing, approved assay; a new assay is worth developing only when the instrumentation and reagents for the approved assay are no longer available. On the other hand, a new assay, for example, the Coomassie blue dye–binding assay, frequently referred to as the Bradford assay, cannot be substituted for the biuret or Kjeldahl assay without a serious consideration of protein quality.368 Solidphase immunoassays may use a solid matrix such as a microplate for ELISA, microbeads as used in the Luminex systems for multiplex assays,369 or surface plasmon resonance systems.370 Surface plasmon resonance systems, as with nephelometry, are label-free assay systems.371 While it is a generalization, immunoassays may be said to measure the mass, while activity assays measure the function. An example is

60

Biotechnology of Plasma Proteins

blood coagulation where CRM+ mutants of factor IX have a defective protein that is immunologically similar to native factor IX.372–374 An ELISA tended to underestimate the potency of a Newcastle vaccine attenuated by chemical modiication (formaldehyde or β-propiolactone)375 but was still useful in deining its quality. A competitive ELISA technique was found to be useful in the evaluation of the potency of hepatitis B vaccines.376 Keller377 used ELISA to evaluate the quality of formaldehyde-modiied botulinum neurotoxin toxoid for vaccine purposes. These latter studies represented an attempt to reduce the use of animals in the testing of biologicals.378 Karnaukhova and colleagues379 developed an ELISA technique that is useful for the determination of active α1-antitrypsin in various matrices; proteolysis and aggregation (heat) resulted in the loss of activity but retention of immunological reactivity. In a conceptually related study, López-Expósito and coworkers380 showed that the proteolysis of ovalbumin reduced but did not eliminate the IgG or IgE reactivity. The production of allergoids from allergens involves the chemical modiication of lysine residues.381 Retention of antigenic reactivity upon chemical modiication, heating, and proteolysis was an integral metric in the early work on the epitope concept.382 The early work on the importance of native antigen structure in immunological reactivity was reviewed by Boyd in 1954.383 H. Gideon Wells, working at the University of Chicago in 1908, published work on the stability of egg albumin as an antigen (sensitizing substance) when subjected to chemical modiication or digestion with proteases. Using an anaphylaxis model, he showed that heating did not eliminate immunological reactivity (horse serum) and nor did iodination. The sensitizing antigen, egg albumin, was subjected to the action of a crude preparation of proteases (Pancreatin, a mixture of digestive enzymes including amylase, proteases, and lipases) for 10 days; antigenic activity was reduced and continuing digestion for 129 days resulted in only small additional decrease in antigenic activity. In a later publication,384 immunological reactivity was further reduced when the incubation of horse serum with Pancreatin385 had been continued for 314 days; biuret reactivity was absent. While the biuret reaction is reasonably speciic, it does lack sensitivity.368 The sensitivity of the biuret reaction depends on the peptide size,386 where the molar extinction coeficient for the biuret reaction with triglycine is 33.5, tetraglycine is 48, and insulin is 670. Noda demonstrated a linear relationship between the peptide size (the number of peptide bond linkages) and the extinction coeficient. Hortin and Meilinger also demonstrated the low reactivity of peptides with the biuret reagent.387 There was also a difference in the spectral characteristics of the reaction product of the biuret reagent with small peptides as compared to albumin. Hortin and Meilinger did report that asparagine and histidine demonstrated high reactivity with the biuret reagent. Taking these observations into account, while Wells did not observe biuret reactivity in his long-term digests, this does not exclude the presence of smaller peptides with low biuret reactivity, which retain their epitopic reactivity. Landsteiner388 showed that small peptides (Mr 600–1000, 8–12 amino acids) derived from the peptic hydrolysis of silk were competitive inhibitors of the precipitin reaction and therefore retained the immunological reactivity of the original epitope on the silk protein. Early researchers, including Wells384,385 and Wornall,389,390 observed that while the treatment of a protein under strongly alkaline conditions (pH > 11) reduced or eliminated antigenicity, the treatment with acid or less rigorous alkaline

Methods for the Analysis of Plasma and Plasma Protein Fractions

61

conditions (pH < 11) did not have the same effect. Nitration or iodination did not eliminate antigenicity and, in the case of nitration, created a new epitope(s). Nitration was accomplished either with nitric acid (producing xanthoproteins) or with tetranitromethane (producing nitrated proteins). Xanthoprotein is an older term used for proteins nitrated with nitric acid; nitrated proteins were used as substrates for proteases.391 Today, it is well known that the nitration of tyrosine residues in proteins creates a neoantigen that can be measured by speciic antibodies.392 Natural antibodies, mostly IgM, serve an important function in recognizing the oxidationspeciic epitopes.393–395 Aggregation is likely the most signiicant factor for product immunogenicity.396–399 Note that we are discussing immunogenicity, not antigenicity. The aggregation of a native protein, as discussed earlier, tends to eliminate the existing epitopes. A recent work400 on birch pollen allergens showed that aggregation reduced antigenicity but did not affect immunogenicity. Other recent observations relevant to the loss of antigenicity on aggregation include observations on the aggregation of recombinant hepatitis B surface antigen401 and virion protein 1 of equine rhinitis A virus.402 It is, however, important to note that aggregation does increase immunogenicity.403–406 This chapter has focused on what would be considered the classical methods for protein analysis. Methods directed at conformational analysis have largely been ignored, and the reader is referred to other works in this area.407–409 The author has a few suggestions based on his own personal experience with assays and assay development: 1. Do not develop a new assay unless it is absolutely necessary. Even if there is an obvious technical advantage in changing a method, the wise will evaluate the cost of change, which includes validation and training. 2. Do not depend on an assay for product release that uses the last instrument of its type from the sole manufacturer for which there are no more spare parts. 3. Make sure that the senior management clearly understands the cost of change, including validation as well as risk.410,411

REFERENCES 1. Stenhagen, E., Electrophoresis of human blood plasma: Electrophoretic properties of ibrinogen, Biochem. J. 32, 714–718, 1938. 2. Miller, G.L., Miller, E.E., and Eitelman, E.S., The pH-mobility relationships of components of human plasma, Arch. Biochem. 29, 413–419, 1950. 3. Hoch, H. and Chanutin, A., An electrophoretic study of human plasma stored at room temperature, J. Biol. Chem. 209, 661–669, 1954. 4. Tiselius, A., Electrophoresis of serum globulin—I, Biochem. J. 31, 313–317, 1937. 5. Tiselius, A., Electrophoresis of serum globulin: Electrophoretic analysis of normal and immune sera, BIochem. J. 31, 1464–1477, 1937. 6. Tiselius, A. and Kabat, E.A., Electrophoresis of immune serum, Science 87, 416–417, 1938. 7. Johnson, P. and Shooter, E.M., Some of the factors involved in the use of the Tiselius electrophoretic apparatus at 20°C, J. Colloid Sci. 2, 539–549, 1948.

62

Biotechnology of Plasma Proteins

8. Kyle, R.A. and Shampo, M.A., Arne Tiselius—Father of electrophoresis, Mayo Clin. Proc. 80, 302, 2005. 9. Righetti, P., Electrophoresis: The march of pennies, the march of dimes, J. Chromatogr. A 1079, 24–40, 2005. 10. Tiselius, A., A new apparatus for the electrophoretic analysis of colloidal mixtures, Trans. Faraday Soc. 33, 524–531, 1937. 11. Thompson, T.E. and McKernan, W.M., An electrophoretic investigation of interactions between bovine plasma albumin and charged dextran derivatives, Biochem. J. 81, 13–23, 1961. 12. Brown, P.H., Balbo, A., and Schuck, P., On the analysis of sedimentation velocity in the study of protein complexes, Eur. Biophys. J. 38, 1079–1099, 2009. 13. Planken, K.L. and Cölfen, H., Analytical ultracentrifugation of colloids, Nanoscale 2, 1849–1869, 2010. 14. Wandrey, C., Hasegawa, U., van der Vlies, A.J., et al., Analytical ultracentrifugation to support the development of biomaterials and biomedical devices, Methods 54, 92–100, 2011. 15. Li, G., Steward, R., Conlon, B., et al., Puriication of human immunoglobulin G: A new approach to plasma fractionation, Vox Sang. 83, 332–338, 2002. 16. Tiselius, A., Electrophoresis, past, present and future, in Protides of the Biological Fluids, 5th Colloquium, ed. H. Peters, Elsevier, Amsterdam, Netherlands, 1957. 17. Kunkel, H.G. and Tiselius, A., Electrophoresis of proteins on ilter paper, J. Gen. Physiol. 35, 88–118, 1951. 18. Kunkel, H.G. and Slater, R.J., Lipoprotein patterns of serum obtained by zone electrophoresis, J. Clin. Invest. 31, 677–684, 1952. 19. Laurell, C.B., Electrophoresis, speciic proteins assays, or both in measurement of plasma proteins?, Clin. Chem. 19, 99–102, 1973. 20. Hjertén, S., Free zone electrophoresis, in Protides of the Biological Fluids, 7th Colloquium, ed. H. Peters, Elsevier, Amsterdam, Netherlands, 1960. 21. http://www.bio-rad.com/LifeScience/pdf/Bulletin_3160.pdf. 22. Egen, N.B., Bliss, M., Mayersohn, M., et al., Isolation of monoclonal antibodies to phencyclidine from ascites luid by preparative isoelectric focusing in the Rotofor, Anal. Biochem. 172, 488–494, 1988. 23. Shimazaki, K., Kawaguchi, A., Sato, T., et al., Analysis of human and bovine milk lactoferrins by Rotofor and chromatofocusing, Int. J. Biochem. 25, 1653–1658, 1993. 24. Goldfarb, M.F., Use of Rotofor in two-dimensional electrophoretic analysis: Identiication of the 100 kDa monoclonal IgG heavy chain in myeloma serum, Electrophoresis 14, 1379–1381, 1993. 25. Ayala, A., Parrado, J., and Machado, A., Use of Rotofor preparative isoelectrofocusing cell in protein puriication procedure, Appl. Biochem. Biotechnol. 69, 11–16, 1998. 26. Ullrich, R., Liers, C., Schimpke, S., and Hofrichter, M., Puriication of homogeneous forms of fungal peroxygenase, Biotechnol. J. 4, 1619–1626, 2009. 27. Vavricka, S.R., Burri, E., Beglinger, C., et al., Serum protein electrophoresis: An underused but very useful test, Digestion 79, 203–210, 2009. 28. Kyle, R.A. and Rajkumar, S.V., Monoclonal gammopathy of undetermined signiicance and smoldering multiple myeloma, Curr. Hematol. Malig. Rep. 5, 62–69, 2010. 29. Putnam, F.W., Introduction, in The Plasma Proteins, ed. F.W. Putnam, Chapter 1, pp. 1–7, Academic Press, New York, 1960. 30. Howe, P.E., The function of the plasma proteins, Physiol. Rev. 5, 439–476, 1925. 31. Svensson, H., Fractionation of serum with ammonium sulfate and water dialysis, studied by electrophoresis, J. Biol. Chem. 139, 805–825, 1941. 32. Hannig, K., Eine neuentwicklung der trägerfreien kontinuierlichen elektrophorese. Zur trennung hochmolekularer und grobdisperser teilchen, Hoppe Seylers Z. Physiol. Chem. 388, 211–227, 1964.

Methods for the Analysis of Plasma and Plasma Protein Fractions

63

33. Streeter, D.G. and Gordon, M.P., Separation of U-1 and U-2 strains of tobacco mosaic virus by continuous free-low electrophoresis, Phytopathology 56, 419–421, 1966. 34. Sulkowski, E. and Laskowski Sr, M., The separation of nucleotides and their derivatives by continuous free-low electrophoresis, Anal. Biochem. 20, 94–101, 1967. 35. Krivánková, L. and Bocek, P., Continuous free-low electrophoresis, Electrophoresis 19, 1064–1074, 1998. 36. Zeiller, K., Hannig, K., and Pascher, G., Free-low electrophoretic separation of lymphocytes. Separation of graft versus host reactive lymphocytes of rat spleens, Hoppe Seylers Z. Physiol. Chem. 352, 1168–1170, 1971. 37. Bauer, J., Electrophoretic separation of cells, J. Chromatogr. 418, 359–383, 1987. 38. Stahn, R., Maier, K.P., and Hannig, K., A new method for the preparation of rat liver lysosomes. Separation of cell organelles of rat liver by carrier-free continuous electrophoresis, J. Cell Biol. 46, 576–591, 1970. 39. Harms, E., Kern, H., and Schneider, J.A., Human lysosomes can be puriied from diploid skin ibroblasts by free-low electrophoresis, Proc. Natl. Acad. Sci. USA 77, 6139–6143, 1980. 40. Schmid, S.L., Toward a biochemical deinition of the endosomal compartment. Studies using free low electrophoresis, Subcell. Biochem. 19, 1–28, 1993. 41. Miller, A.L., Kress, B.C., Stein, R., et al., Properties of N-acetyl-β-D-hexosaminidase from isolated and normal lysosomes, J. Biol. Chem. 256, 9352–9362, 1981. 42. Righetti, P.G., Castagna, A., Herbert, B., et al., Prefractionation techniques in proteome analysis, Proteomics 3, 1397–1407, 2003. 43. Islinger, M., Eckerskorn, C., and Völkl, A., Free-low electrophoresis in the proteomic era: A technique in lux, Electrophoresis 31, 1754–1763, 2010. 44. Pusztal, A. and Walt, W.B., Free-low electrophoresis of proteins in phenol-containing solvents at various pH values, Biochim. Biophys. Acta 251, 158–163, 1971. 45. Shukun, S.A., Gavryushkin, A.V., Brezgunov, A.V., and Zav’yalov, V.P., Protein separation in pH gradients using free-low electrophoretic apparatus. II: The pH gradients formed by the concentration gradient of boric acid in solutions of borax and mannitol, Electrophoresis 6, 75–77, 1985. 46. Kaufmann, M., Unstable proteins: How to subject them to chromatographic separations for puriication procedures, J. Chromatogr. B Biomed. Sci. Appl. 699, 347–369, 1997. 47. Kobayashi, H., Shimamura, K., Akaida, T., et al., Free-low electrophoresis in a microfabricated chamber with a micromodule fraction separator. Continuous separation of proteins, J. Chromatogr. A 990, 169–178, 2003. 48. Nissum, M. and Foucher, A.L., Analysis of human plasma proteins: A focus on sample collection and separation using free-low electrophoresis, Expert Rev. Proteomics 5, 571–587, 2008. 49. http://www.bd.com/proteomics/products/ffe/faqs.asp. 50. http://www.gradipore.com/Technology/. 51. Wagner, H., Free-low electrophoresis, Nature 341, 699–700, 1989. 52. Turgeon, R.T., Fonslow, B.R., Jing, M., and Bowser, M.T., Measuring aptamer equilibria using gradient micro free low electrophoresis, Analyt. Chem. 82, 3636–3641, 2010. 53. Bluestein, B.I. and Vaitukaitis, J.L., The effect of ionic environment in speciic FSH binding to plasma membrane receptor, J. Recept. Res. 2, 245–266, 1981. 54. Roda, B., Zattoni, A., Reschiglian, P., et al., Field-low fractionation in bioanalysis: A review of recent trends, Anal. Chim. Acta 635, 132–143, 2009. 55. Reschiglian, P. and Moon, M.H., Flow ield-low fractionation: A pre-analytical method for proteomics, J. Proteomics 71, 265–276, 2008. 56. Yohannes, G., Jussila, M., Hartonen, K., and Riekkola, M.L., Asymmetrical low ieldlow fractionation technique for separation and characterization of biopolymers and bioparticles, J. Chromatogr. A 1218, 4104–4116, 2011.

64

Biotechnology of Plasma Proteins

57. Allen, P.C., Hill, E.A., and Stokes, A.M., Plasma Proteins Analytical and Preparative Techniques, Blackwell, Oxford, UK, 1977. 58. Putnam, F.W., Perspectives-past, present, future, in The Plasma Proteins, 2nd edn., ed. F.W. Putnam, Vol. 1, Chapter 1, pp. 2–55, Academic Press, New York, USA, 1975. 59. Heide, K., Haupt, H., and Schwick, H.G., Plasma protein fractionation, in The Plasma Proteins, 2nd edn., ed. F.W. Putnam, Vol. III, Chapter 8, pp. 546–597, Academic Press, New York, 1977. 60. Putnam, F.W., Progress in plasma proteins, in The Plasma Proteins, 2nd edn., ed. F.W. Putnam, Vol. IV, Chapter 1, pp. 2–41, Academic Press, New York, 1984. 61. Anderson, N.L., Polanski, M., Pieper, R., et al., The human plasma proteome A nonredundant list developed by combination of four separate sources, Mol. Cell. Proteomics 3, 311–326, 2004. 62. Donato, R., Sorci, G., Riuzzi, F., et al., S100B’s double life: Intracellular regulator and extracellular signal, Biochim. Biophys. Acta 1793, 1008–1022, 2009. 63. Anderson, N.L., Anderson, N.G., Pearson, T.W., et al., A human proteome detection and quantitation project, Mol. Cell. Proteomics 8, 883–886, 2009. 64. Lundblad, R.L., Development and Applications of Biomarkers, CRC Press, Boca Raton, FL, 2011. 65. Anderson, N.L., The clinical plasma proteome: A survey of clinical assays for proteins in plasma and serum, Clin. Chem. 56, 177–185, 2010. 66. Thiele, T., Steil, L., Völker, U., and Greinacher, A., Proteomics of blood-based therapeutics: A promising tool for quality assurance in transfusion medicine, BioDrugs 21, 179–183, 2007. 67. D’Alessandro, A. and Zolla, L., Proteomics for quality-control processes in transfusion medicine, Anal. Bioanal. Chem. 398, 111–124, 2010. 68. Gaso-Sokac, D. and Josic, D., The role of proteomics in plasma fractionation and quality control of plasma-derived therapeutic proteins, Blood Transfus. (Suppl. 3), s86–s91, 2010. 69. Chuong, M.C., Palugan, L., Su, T.M., et al., Formulation of controlled-release capsules of biopharmaceutical classiication system I drugs using niacin as a model, AAPS Pharm. Sci. Tech. 11, 1650–1661, 2010. 70. Walsh, G., Biopharmaceutical benchmarks, Nat. Biotechnol. 28, 917–928, 2010. 71. Bhambure, R., Kumar, K., and Rathore, A.S., High-throughput process development for biopharmaceutical drug substance, Trends Biotechnol. 29, 127–135, 2011. 72. Neeraj, A., Chandrasekar, M.J., Sara, U.V., et al., Poly(HEMA-Zidovudine) conjugate: A macromolecular pro-drug for improvement in the biopharmaceutical properties of the drug, Drug Delivery 18, 272–280, 2011. 73. Rader, R.A., (Re)deining biopharmaceutical, Nat. Biotechnol. 26, 743–751, 2008. 74. Burgess, C., Analytical quality management, in Analytical Chemistry: A Modern Approach to Analytical Science, 2nd edn., eds. R. Kellner, J.-M. Mermet, M. Otto, M. Valcarcel, and H.M. Widmer, Chapter 6, pp. 69–88, Wiley-VCH, Weinheim, Germany, 2004. 75. Ransom, C., Bioequivalence/bioavailability retention samples, Qual. Assur. 2, 42–43, 1993. 76. Glock, B., Reisacher, R.B., Schöck, M.A., et al., DNA proiling: A valuable tool for quality control of sample logistics including occurrences of suspected sample confusion in a blood donation centre, Vox Sang. 82, 137–140, 2002. 77. Cardosa, S., Valverde, L., Odriozola, A., et al., Quality standards in Biobanking: Authentication by genetic proiling of blood spots from donor’s original sample, Eur. J. Hum. Genet. 18, 848–851, 2010. 78. Lundblad, R.L., Kingdon, H.S., Mann, K.G., and White, G.C., Issues with the assay of factor VIII activity in plasma and factor VIII concentrates, Thromb. Haemost. 84, 942–948, 2000. 79. Lundblad, R.L., Approaches to the Conformational Analysis of Biopharmaceuticals, CRC Press, Boca Raton, FL, 2010.

Methods for the Analysis of Plasma and Plasma Protein Fractions

65

80. Ligthart, S., Vlemmix, F., Dendukuri, N., and Brophy, J.M., The cost-effectiveness of drug-eluting stents: A systematic review, Can. Med. Assoc. J. 176, 199–205, 2007. 81. van Luijn, J.C., Stolk, P., Gribnau, F.W., and Leufkens, H.G., Gap in publication of comparative information on new medicines, Br. J. Clin. Pharmacol. 65, 716–722, 2008. 82. Rogowski, W.H., Hartz, S.C., and John, J.H., Clearing up the hazy road from bench to bedside: A framework for integrating the fourth hurdle into translational medicine, BMC Health Serv. Res. 8, 194, 2008. 83. Deverka, P.A., Pharmacogenomics, evidence, and the role of payers, Public Health Genomics 12, 149–157, 2009. 84. Cohen, J., Wilson, A., and Faden, L., Off-label use reimbursement, Food Drug Law J. 64, 391–403, 2009. 85. Luce, B.R. and Brown, R.E., The use of technology assessment by hospitals, health maintenance organizations, and third-party in these United States, Int. J. Technol. Assess. Health Care 11, 79–92, 1995. 86. Ruotolo, B.T. and Robinson, C.V., Aspects of native proteins are retained in vacuum, Curr. Opin. Chem. Biol. 10, 402–408, 2006. 87. Srebalus Barnes, C.A. and Lim, A., Applications of mass spectrometry for the structural characterization of recombinant protein pharmaceuticals, Mass Spectrom. Rev. 26, 370–388, 2007. 88. Brueker, K., Jin, M., Han, X., et al., Top-down identiication and characterization of biomolecules by mass spectrometry, J. Am. Soc. Mass Spectrom. 19, 1045–1053, 2008. 89. Zhang, X., Pan, H., and Chen, X., Mass spectrometry for structural characterization of therapeutic antibodies, Mass Spectrom. Rev. 28, 147–176, 2009. 90. Priego Capote, F. and Sanchez, J.C., Strategies for proteomic analysis of nonenzymatically glycated proteins, Mass Spectrom. Rev. 28, 135–146, 2009. 91. Mendoza, V.L. and Vachet, R.W., Probing protein structure by amino acid-speciic covalent labeling and mass spectrometry, Mass Spectrom. Rev. 28, 785–815, 2009. 92. Grandori, R., Santambrogio, C., Brocca, S., et al., Electrospray-ionization mass spectrometry as a tool for fast screening of protein structural properties, Biotechnol. J. 4, 73–87, 2009. 93. Baczek, T. and Kaliszan, R., Prediction of peptides’ retential times in reversed-phase liquid chromatography as a new supportive tool to improve protein identiication in proteomics, Proteomics 9, 835–847, 2009. 94. Becker, J.S. and Jakubowski, N., The synergy of elemental and biomolecular mass spectrometry: New analytical strategies in life sciences, Chem. Soc. Rev. 38, 1969–1983, 2009. 95. Mariño, K., Bones, J., Kattla, J.J., and Rudd, P.M., A systematic approach to protein glycosylation analysis: A path through the maze, Nat. Chem. Biol. 6, 713–723, 2010. 96. Oettl, K. and Marsche, G., Redox state of human serum albumin in terms of cysteine-34 in health and disease, Meth. Enzymol. 474, 181–195, 2010. 97. Bunk, D.M., Characterization of the glycation of albumin in freeze-dried and frozen human serum, Anal. Chem. 69, 2457–2463, 1997. 98. Gornik, O., Wagner, J., Pucić, M., et al., Stability of N-glycan proiles in human plasma, Glycobiology 19, 1547–1553, 2009. 99. Smith, K.D., Pollacchi, A., Field, M., and Watson, J., The heterogeneity of the glycosylation of alpha-1-acid glycoprotein between the sera and synovial luid in rheumatoid arthritis, Biomed. Chromatogr. 6, 261–266, 2002. 100. Valmu, L., Kallkinen, N., Husa, A., and Rye, P.D., Differential susceptibility of transferrin glycoforms to chymotrypsin: A proteomics approach to the detection of carbohydratedeicient transferrin, Biochemistry 44, 16007–16013, 2005. 101. Raghav, S.K., Gupta, B., Agrawal, C., et al., Altered expression and glycosylation of plasma proteins in rheumatoid arthritis, Glycoconj. J. 23, 167–173, 2006.

66

Biotechnology of Plasma Proteins

102. Harazono, A., Kawasaki, N., Hoh, S., et al., Simultaneous glycosylation analysis of human serum glycoproteins by high-performance liquid chromatography/tandem mass spectrometry, J. Chromatogr. B 869, 20–30, 2008. 103. Mackler, B., Biosimilars and follow-on branded biologics, Genet. Eng. Biotechnol. News 29(4), 2009. 104. Kawasaki, N., Itoh, S., Hashii, N., et al., The signiicance of glycosylation analysis in development of biopharmaceuticals, Biol. Pharm. Bull. 32, 796–800, 2009. 105. Jiang, H., Wu, S.L., Karger, B.L., and Hancock, W.S., Characterization of the glycosylation occupancy and the active site in the follow-on protein therapeutic: TNK-tissue plasminogen activator, Anal. Chem. 82, 6154–6162, 2010. 106. Xie, H., Chakraborty, A., Ahn, J., et al., Rapid comparison of a candidate biosimilar to an innovator monoclonal antibody with advanced liquid chromatography and mass spectrometry technologies, MAbs 2, 379–394, 2010. 107. Vang, Y., Wu, Sh-i, and Hancock, W.S., Approaches to the study of N-linked glycoproteins in human plasma using lectin afinity chromatography and nano-HPLC coupled to electrospray linear ion-trap-Fourier transform mass spectrometry, Glyobiology 16, 514–523, 2006. 108. Haegglund, P., Matheson, R., Elortza, F., et al., An enzymatic deglycosylation scheme enabling identiication of core fucosylated N-glycans and O-glycosylation site mapping of human plasma proteins, J. Proteome Res. 6, 3021–3031, 2007. 109. Kumar, H.P., Hague, C., Haley, T., et al., Elucidation of N-linked oligosaccharide structures of recombinant human factor VIII using luorophore-assisted carbohydrate electrophoresis, Biotechnol. Appl. Biochem. 24, 207–216, 1996. 110. Lundblad, R.L. and Bradshaw, R.A., Addressing Product Improvement Using Chemical Modiication in Biopharmaceutical Manufacture: A case study in blood coagulation factor VIII, Bioprocess Int. 4, 1–8, September, 2006. 111. Musso, R., Eficacy and safety of recombinant factor VIII products in patients with hemophilia A, Drugs Today (Barc) 44, 735–750, 2008. 112. Fay, P.J., Chavin, S.I., Malone, J.E., et al., The effect of carbohydrate depletion on procoagulant activity and in vivo survival of highly puriied human factor VIII, Biochim. Biophys. Acta 800, 152–158, 1984. 113. Kosloski, M.P., Miclea, R.D., and Balu-Iyer, S.V., Role of glycosylation in conformational stability, activity, macromolecular interaction and immunogenicity of recombinant factor VIII, AAPS J. 11, 424–431, 2009. 114. Maley, F., Trimble, R.B., Tarentino, A.L., and Plummer, T.H., Jr., Characterization of glycoproteins and their associated oligosaccharides through the use of endoglycosidases, Anal. Biochem. 180, 195–204, 1989. 115. Baynes, J.W. and Wold, F., Effect of glycosylation on the in vivo circulating half-life of ribonuclease, J. Biol. Chem. 251, 6016–6024, 1976. 116. Skropeta, D., The effect of individual N-glycans on enzyme activity, Bioorg. Med. Chem. 17, 2645–2653, 2009. 117. van Eijk, H.M., and Deutz, N.E., Plasma protein synthesis measurements using a proteomics strategy, J. Nutr. 133(6 Suppl. 1), 2084S–2089S, 2003. 118. Schmidt, A. and Aebersold, R., High-accuracy proteome maps of human body luids, Genome Biol. 7, 242, 2006. 119. Herosimczyk, A., Dejeans, N., Sayd, T., et al., Plasma proteome analysis: 2D gels and chips, J. Physiol. Pharmacol. 57(Suppl. 5), 81–93, 2006. 120. Issaq, H.J., Xiao, Z., and Veenstra, T.D., Serum and plasma proteomics, Chem. Rev. 107, 3601–3620, 2007. 121. Garbett, N.C., Mekmaysy, C.S., Helm, C.W., et al., Differential scanning calorimetry of blood plasma for clinical diagnosis and monitoring, Exp. Mol. Pathol. 86(3), 186– 191, 2009.

Methods for the Analysis of Plasma and Plasma Protein Fractions

67

122. Mauri, P. and Scigelova, M., Multidimensional protein identiication technology for clinical proteomic analysis, Clin. Chem. Lab. Med. 47, 636–646, 2009. 123. Apweiler, R., Aslanidis, C., Deufel, T., et al., Approaching clinical proteomics: Current state and future ields application in luid proteomics, Clin. Chem. Lab. Med. 47, 724–744, 2009. 124. Hawridge, A.M. and Muddiman, D.C., Mass spectrometry-based biomarker discovery: Toward a global proteome index of individuality, Annu. Rev. Anal. Chem. 2, 265–277, 2009. 125. Liumbruno, C., D’Alessandro, A., Grazzini, G., and Zolla, L., Blood-related proteomics, J. Proteomics 73, 483–507, 2010. 126. D’Alessandro, A. and Zolla, L., Proteomics for quality-control processes in transfusion medicine, Anal. Bioanal. Chem. 398, 111–124, 2010. 127. Sapan, C.V. and Lundblad, R.L., Considerations regarding the use of blood samples in the proteomic identiication of biomarkers for cancer diagnosis, Cancer Genomics Proteomics, 33, 227–231, 2006. 128. Burgess, C. and Lundblad, R.L., Plasma or serum in proteomic analysis, Clin. Lab. Int. 31(1), 10–11, 2007. 129. Lundblad, R.L., The Evolution of Protein Chemistry to Proteomics: Basic Science to Clinical Application, CRC Press, Boca Raton, FL, 2005. 130. Siev, M., Yu, X., Prados-Rosales, R., et al., Correlation between serum and plasma antibody titers mycobacterial antigens, Clin. Vaccine Immunol. 18, 173–175, 2011. 131. Brigulla, M., Thiele, T., Scharf, C., et al., Proteomics as a tool for assessment of therapeutics in transfusion medicine: Evaluation of prothrombin complex concentrates, Transfusion 46, 377–385, 2006. 132. Yang, X., Clifton, J., Huang, F., et al., Proteomic analysis for process development and control of therapeutic protein separation from human plasma, Electrophoresis 30, 1185– 1193, 2009. 133. Gaso-Sokac, D. and Josic, D., The role of proteomics in plasma fractionation and quality control of plasma-derived therapeutic proteins, Blood Transfusion 8(Suppl. 3), S86– S89, 2010. 134. Clifton, J., Huang, F., Gaso-Sokac, D., et al., Use of proteomics for validation of the isolation process of clotting factor IX from human plasma, J. Proteomics 73, 678–688, 2010. 135. D’Alessandro, A. and Zolla, L., Proteomics for quality-control processes in transfusion medicine, Anal. Bioanal. Chem. 398, 111–124, 2010. 136. Pusch, W., Flocco, M.T., Leung, S.M., et al., Mass spectrometry-based clinical proteomics, Pharmacogenomics 4, 463–476, 2003. 137. Hernandez, P., Müller, M., and Appel, R.D., Automatic protein identiication by tandem mass spectrometry: Issues and strategies, Mass Spectrom. Rev. 25, 234–254, 2006. 138. Demeyere, R., Gillardin, S., Arnout, J., and Strengers, P.F., Comparison of fresh frozen plasma and prothrombin complex concentrate for the reversal of oral anticoagulants in patients undergoing cardiopulmonary bypass surgery: A randomized study, Vox Sang. 99, 251–260, 2010. 139. Morimoto, Y., Niwa, H., and Nakatani, T., On the use of prothrombin complex concentrate in patients with coagulopathy requiring tooth extraction, Oral Surg. Oral Med. Oral Pathol. 111, e7–e10, 2010. 140. Illanes, S., Zhou, W., Schwarting, S., et al., Comparative effectiveness of hemostatic therapy in experimental warfarin-associated intracerebral hemorrhage, Stroke 42, 191–195, 2011. 141. Makris, M. and Van Veen, J.J., Three or four factor prothrombin complex concentrate for emergency anticoagulation reversal?, Blood Transfusion 9, 117–119, 2011. 142. Bond, M., Jankowski, H., Patal, S., et al., Biochemical characterization of recombinant factor IX, Semin. Hematol. 35(Suppl. 2), 11–17, 1998.

68

Biotechnology of Plasma Proteins

143. Barrowcliffe, T.W., Insights from factor IX activation studies with chromogenic assays: Implications of disparate product results, Haemophilia 16(Suppl. 6), 9–12, 2010. 144. Thim, L., Bjoern, S., Christensen, M., et al., Amino acid sequence and posttranslational modiications of human factor VIIa from plasma and transfected baby hamster kidney cells, Biochemistry 27, 7785–7793, 1988. 145. Torio, A., Matino, D., D’Amico, R., and Makris, M., Recombinant factor VIIa concentrate versus plasma derived concentrates for the treatment of acute bleeding episodes in people with haemophilia and inhibitors, Cochrane Database Syst. Rev. 4(8), CD004449, 2010. 146. Eaton, D.L., Hass, P.E., Ribble, L., et al., Characterization of recombinant human factor VIII, J. Biol. Chem. 262, 3285–3290, 1987. 147. Bray, G.L., Recent advances in the preparation of plasma-derived and recombinant coagulation factor VIII, J. Pediatr. 117, 503–507, 1990. 148. Kemball-Cook, G., Tubbs, J.E., Dawson, N.J., and Barrowcliffe, T.W., The behaviour of different factor VIII concentrates in a chromogenic factor X-activating system, Br. J. Haematol. 84, 273–278, 1993. 149. Hironaka, T., Furukawa, K., Esmon, P.C., et al., Comparative study of the sugar chains of factor VIII puriied from human plasma and from the culture media of recombinant baby hamster kidney cells, J. Biol. Chem. 267, 8012–8020, 1992. 150. Bihoreau, N., Pin, S., de Kersabiec, A.M., et al., Copper-atom identiication in the active and inactive forms of plasma-derived FVIII and recombinant FVIII-delta II, Eur. J. Biochem. 222, 41–48, 1994. 151. Hironaka, T., Furukawa, K., Esmon, P.C., et al., Structural study of the sugar chains of porcine factor VIII–tissue- and species-speciic glycosylation of factor VIII, Arch. Biochem. Biophys. 307, 316–330, 1993. 152. Kumar, H.P., Hague, C., Haley, T., et al., Elucidation of N-linked oligosaccharide structures of recombinant human factor VIII using luorophore-assisted carbohydrate electrophoresis, Biotechnol. Appl. Biochem. 24, 207–216, 1996. 153. Bjoern, S., Foster, D.C., Thim, L., et al., Human plasma and recombinant factor VII. Characterization of O-glycosylations at serine residues 52 and 60 and effects of sitedirected mutagenesis of serine 52 to alanine, J. Biol. Chem. 266, 11051–11057, 1991. 154. Stefanowicz, P., Kijewska, M., Kluczyk, A., and Szewczuk, Z., Detection of glycation sites in proteins by high-resolution mass spectrometry combined with isotopic labeling, Anal. Biochem. 400, 237–243, 2010. 155. Barnaby, O.S., Was, C., Cerny, R.L., et al., Quantitative analysis of glycation sites on human serum albumin using 16O/18O-labeling and matrix-assisted laser desorption/ ionization time-of-light mass spectrometry, Clin. Chim. Acta 411, 1102–1110, 2010. 156. Frolov, A. and Hoffman, R., Identiication and relative quantiication of speciic glycation sites in human serum albumin, Anal. Bioanal. Chem. 397, 2349–2356, 2010. 157. Rüggeberg, S., Horn, P., Li, X., et al., Detection of a γ-carboxy-glutamate as novel posttranslational modiication of human transthyretin, Protein Pept. Lett. 15, 43–46, 2008. 158. Harris, R.J., Ling, V.T., and Spellman, M.W., O-linked fucose is present in the irst epidermal growth factor domain of factor XII but not protein C, J. Biol. Chem. 267, 5102–5107, 1998. 159. Rouse, J.C., McClellan, J.E., Patel, H.K., et al., Top-down characterization of protein pharmaceuticals by liquid chromatography/mass spectrometry: Application to recombinant factor IX comparability—A case study, Meth. Mol. Biol. 308, 435–460, 2005. 160. Femaille, F., Grosseil, C., Ramon, C., et al., Mass spectrometric characterization of N- and O-glycans of plasma-derived coagulation factor VII, Glycoconj. J. 25, 827–842, 2008. 161. Gil, G.C., Velander, W.H., and Van Cott, K.E., N-Glycosylation microheterogeneity and site occupancy of an Asn-X-Cys sequon in plasma-derived and recombinant protein C, Proteomics 9, 2555–2567, 2009.

Methods for the Analysis of Plasma and Plasma Protein Fractions

69

162. Müller, R., Marchetti, M., Kratzmeier, M., et al., Comparison of planar SDS-PAGE, CGE-on-a-chip, and MALDI-TOF mass spectrometry for analysis of the enzymatic de-N-glycosylation of antithrombin and coagulation factor IX with PNGase F, Anal. Bioanal. Chem. 389, 1859–1868, 2007. 163. Belgacem, O., Buchacher, A., Pock, K., et al., Molecular mass determination of plasmaderived glycoproteins by ultraviolet matrix-assisted laser desorption/ionization time-oflight mass spectrometry with internal calibration, J. Mass Spectrom. 37, 1118–1130, 2002. 164. Deyde, V.M., Sampath, R., and Gubareva, L.V., RT-PCR/electrospray ionization mass spectrometry approach in detection and characterization of inluenza viruses, Expert Rev. Mol. Diagn. 11, 41–52, 2011. 165. Ye, Y., Mar, E.C., Tong, S., et al., Application of proteomics methods for pathogen discovery, J. Virol. Meth. 163, 87–95, 2010. 166. Schwahn, A.B., Wong, J.W., and Downard, K.M., Typing of human and animal strains of inluenza virus with conserved signature peptides of matrix M1 protein by high resolution mass spectrometry, J. Virol. Meth. 165, 178–185, 2010. 167. Grant-Klein, R.J., Baldwin, C.D., Turell, M.J., et al., Rapid identiication of vectorborne laviviruses by mass spectrometry, Mol. Cell. Probes 24, 219–228, 2010. 168. Chen, K.F., Rothman, R.E., Ramachandran, P., et al., Rapid identiication of virus from nasal pharyngeal aspirates in acute viral respiratory infections by RT-PCR and electrospray ionization mass spectrometry, J. Virol. Meth. 173, 60–66, 2011. 169. D’Amici, G.M., Tiperio, A.M., Gevi, F., et al., Recombinant clotting factor VIII concentrate: Heterogeneity and high-purity evaluation, Electrophoresis 31, 2730–2739, 2010. 170. Ahrends, R., Lichtner, B., Bertsch, A., et al., Application of displacement chromatography for the proteome analysis of a human plasma protein fraction, J. Chromatogr. A 1217, 3321–3329, 2010. 171. Pan, J., Qian, Y., Zhou, X., et al., Oversulfated chondroitin sulfate is not the sole contaminant in heparin, Nat. Biotechnol. 28, 203–207, 2010. 172. Pan, J., Qian, Y., Zhou, X., et al., Identiication of chemical sulfated/desulfated glycosaminoglycans in contaminated heparins and development of a simple assay for the detection of most contaminants in heparin, Glycobiol. Insights 2010, 1–12, 2010. 173. Brustkern, A.M, Buhse, L.F., Nasr, M., et al., Characterization of currently marketed heparin products: Reversed-phase ion-pairing liquid chromatography mass spectrometry of heparin digests, Anal. Chem. 82, 9865–9870, 2010. 174. Tran, V.M., Nguyen, T.K., Raman, K., and Kuberan, B., Applications of isotopes in advancing structural and functional heparanomics, Anal. Bioanal. Chem. 399, 559–570, 2011. 175. Lee, S.E., Chess, E.K., Rabinow, B., et al., NMR of heparin API: Investigation of unidentiied signals in the USP-speciied range of 2.12–3.00 ppm, Anal. Bioanal. Chem. 399, 651–662, 2011. 176. Yang, B., Solakylidirim, K., Chang, Y., and Linhardt, R.J., Hyphenated techniques for the analysis of heparin and heparan sulfate, Anal. Bioanal. Chem. 399, 541–557, 2011. 177. McKee, J., Bairstow, S., Szabo, C., et al., Structure elucidation and biological activity of the oversulfated chondroitin sulfate contaminant in Baxter heparin, J. Clin. Pharmacol. 50, 1159–1170, 2010. 178. Zechmeister, L., History, scope, and methods of chromatography, Ann. N.Y. Acad. Sci. 49, 145–160, 1948. 179. Weil, H. and Williams, T.I., Early history of chromatography, Nature 167, 906–907, 1951. 180. Synge, R.L., Tsvet, Willstatter, and the use of adsorption for puriication of proteins, Arch. Biochem. Biophys. (Suppl. 1), 1–6, 1962. 181. Stewart, G.A., Historical review of the analytical control of insulin, Analyst 99, 913– 928, 1974.

70

Biotechnology of Plasma Proteins

182. Sober, H.A. and Peterson, E.A., Protein chromatography on ion exchange cellulose, Fed. Proc. 17, 1116–1126, 1958. 183. Porath, J. and Flodin, P., Gel iltration: A method for desalting and group separation, Nature 183, 1657–1659, 1959. 184. Kay, R.G., Barton, C., Velloso, C.P., et al., High-performance ultra-high-performance liquid chromatography/tandem mass spectrometry quantitation of insulin-like growth factor-I and leucine-rich α-2-glycoprotein in serum as biomarkers of recombinant human growth hormone administration, Rapid Commun. Mass Spectrom. 23, 3173–3182, 2009. 185. Yao, K. and Hjertén, S., Gradient and isocratic high-performance liquid chromatography of proteins on a new agarose-based anion exchanger, J. Chromatogr. 385, 87–98, 1987. 186. Patst, T.M., Buckley, J.J., Ramasubramanyan, N., and Hunter, A.K., Comparison of strong anion-exchangers for the puriication of a PEGylated protein, J. Chromatogr. A 1147, 172–182, 2007. 187. Kumpalume, P., Podmore, A., LePage, C., and Dalton, J., New process for the manufacture of α1-antitrypsin, J. Chromatogr. A 1148, 31–37, 2007. 188. Chu, G.C., Chelius, D., Xiao, G., et al., Accumulation of succinimide in a recombinant monoclonal antibody in mildly acidic buffers under elevated temperatures, Pharm. Res. 24, 1145–1156, 2007. 189. Dave, N., Hazra, P., Khedkar, A., et al., Process and puriication for manufacture of a modiied insulin intended for oral delivery, J. Chromatogr. A 1177, 282–286, 2008. 190. Kelley, B.D., Switzer, M., Bastek, P., et al., High-throughput screening of chromatographic separations: IV. Ion-exchange, Biotechnol. Bioeng. 100, 950–963, 2008. 191. Azevedo, A.M., Rosa, P.A., Ferreira, I.F., et al., Downstream processing of human antibodies integrating an extraction capture step and cation exchange chromatography, J. Chromatogr. B 9, 50–58, 2009. 192. Yan, B., Steen, S., Hambly, D., et al., Succinimide formation at Asn 55 in the complementarity determining region of a recombinant monoclonal antibody IgG1 heavy chain, J. Pharm. Sci. 98, 3509–3521, 2009. 193. Vlasak, J., Bussat, M.C., Wang, S., et al., Identiication and characterization of asparagine deamidation in the light chain CDR1 of a humanized IgG1 antibody, Anal. Biochem. 392, 145–154, 2009. 194. Pristatsky, P., Cohen, S.L., Krantz, D., et al., Evidence of trisulide bonds in a recombinant variant of a human IgG2 monoclonal antibody, Anal. Chem. 81, 6148–6155, 2009. 195. Schmoeger, E., Berger, E., Treilov, A., et al., Matrix-assisted refolding of autoprotease fusion proteins on an ion exchange column, J. Chromatogr. A 1216, 8460–8469, 2009. 196. Chen, J. Tetrault, J., Zhang, Y., et al., The distinctive separation attributes of mixedmode resins and their application in monoclonal antibody downstream puriication processes, J. Chromatogr. A 1217, 216–224, 2010. 197. Thim, L., Vandahl, B., Karlsson, J., et al., Puriication and characterization of a new recombinant factor VIII (N8), Haemophilia 16, 349–359, 2010. 198. Fweja, L.W., Lewis, M.J., and Grandison, A.S., Isolation of lactoperoxidase using different cation exchange resins by batch and column procedures, J. Dairy Res. 77, 357–367, 2010. 199. Wang, S., Ionescu, R., Peekhaus, N., et al., Separation of post-translational modiications in monoclonal antibodies by exploiting subtle conformational changes under mildly acidic conditions, J. Chromatogr. A 1217, 6496–6502, 2010. 200. Johnson, K.A., Paisley-Flango, K., Tangarone, B.S., et al., Cation exchange-HPLC and mass spectrometry reveal C-terminal amidation of an IgG1 heavy chain, Anal. Biochem. 360, 75–83, 2007. 201. Kakehi, K., Kinoshita, M., Kawakami, D., et al., Capillary electrophoresis of sialic acidcontaining glycoprotein: Effect of the heterogeneity of carbohydrate chains on glycoform separation using an α1-acid glycoprotein as a model, Anal. Chem. 73, 2640–2647, 2001.

Methods for the Analysis of Plasma and Plasma Protein Fractions

71

202. Liu, C., Dong, S., Xu, X.U., et al., Assessment of the quality and structural integrity of a complex glycoprotein mixture following extraction from the formulated biopharmaceutical drug product, J. Pharm. Biomed. Anal. 54, 27–36, 2011. 203. Lundblad, R.L., A hydrophobic site in human prothrombin present in a calciumstabilized conformer, Biochem. Biophys. Res. Commun. 157, 295–300, 1988. 204. Withka, J., Moncuse, P., Baziotis, A., and Maskiewicz, R., Use of high-performance size-exclusion chromatography, ion-exchange and hydrophobic interaction chromatography for measurement of protein conformational change and stability, J. Chromatogr. 398, 175–202, 1987. 205. Yamamoto, S., Fuji, S., Yoshimoto, N., and Akbarzadehlaleh, P., Effect of protein conformational changes on separation performance in electrostatic interaction chromatography: Unfolded proteins and PEGylated proteins, J. Biotechnol. 132, 196–201, 2007. 206. Cole, R.D., The chromatography of insulin in urea-containing buffer, J. Biol. Chem. 235, 2294–2299, 1960. 207. Voitl, A., Butté, A., and Morbidelli, M., Behavior of human serum albumin on strong cation exchange resins. I: Experimental analysis, J. Chromatogr. A 1217, 5484–5491, 2010. 208. Saito, K., Hamano, K., Nakagawa, M., et al., Conformational analysis of human serum albumin and its nonenzymatic glycation products using monoclonal antibodies, J. Biochem. 149, 569–580, 2011. 209. Voitl, A., Butté, A., and Morbidelli, M., Behavior of human serum albumin on strong cation exchange resins. II: Model analysis, J. Chromatogr. A 1217, 5492–5500, 2010. 210. Hou, T., Hansen, T.B., Staby, A., and Cramer, S.M., Effects of urea induced protein conformational changes on ion exchange chromatographic behavior, J. Chromatogr. A 1217, changes on ion exchange resins, J. Chromatogr. A 1217, 7393–9400, 2010. 211. Fenton II, J.W., Olson, T.A., Zabinski, M.P., and Wilner, G.D., Anion-binding exosite of human α-thrombin and ibrin(ogen) recognition, Biochemistry 27, 7106–7112, 1988. 212. Lundblad, R.L., A rapid method for the puriication of bovine thrombin and the inhibition of the puriied enzyme with phenylmethylsulfonyl luoride, Biochemistry 10, 2501– 2506, 1971. 213. Stubbs, M.T. and Bode, W., A player of many parts: The spotlight falls on thrombin’s structure, Thromb. Res. 69, 1–58, 1993. 214. Sharma, J., Principles of gel permeation chromatography, J. AOAC Int. 91, 113A–118A, 2008. 215. Wood, G.C. and Cooper, P.F., The application of gel iltration to the study of proteinbinding of small molecules, Chromatogr. Rev. 1, 88–107, 1970. 216. Andrews, P., Estimation of molecular size and molecular weights of biological compounds by gel iltration, Meth. Biochem. Anal. 18, 1–53, 1970. 217. Williams, K.W., Solute-gel interactions in gel iltration, Lab. Pract. 21, 667–670, 1972. 218. Winzor, D.J., From gel iltration to biosensor technology: The development of chromatography for the characterization of protein interactions, J. Mol. Recognit. 13, 279–298, 2000. 219. Porath, J., From gel iltration to adsorptive size exclusion, J. Protein Chem. 16, 463– 468, 1997. 220. Drohan, W.N., Miekka, S.I., Griko, Y.V., et al., Gamma irradiation of intravenous immunoglobulin, Dev. Biol. 118, 133–138, 2004. 221. Iwase, H., Tanaka, A., Hiki, Y., et al., Aggregated human serum immunoglobulin A1 induced by neuraminidase treatment had a lower number O-linked sugar chains on the hinge portion, J. Chromatogr. B 724, 1–7, 1999. 222. Dave, N., Hazra, P., Khedkar, A., et al., Process and puriication for manufacture of a modiied insulin intended for oral delivery, J. Chromatogr. A 1177, 282–286, 2008.

72

Biotechnology of Plasma Proteins

223. Kunitani, M., Wolfe, S., Rana, S., et al., Classical light scattering quantitation of protein aggregates: Off-line spectroscopy versus HPLC detection, J. Pharm. Biomed. Anal. 16, 573–586, 1997. 224. Bolli, R., Woodtli, K., Bärtschi, M., et al., L-Proline reduces IgG dimer content and enhances the stability of intravenous immunoglobulin (IVIG) solutions, Biologicals 38, 150–157, 2010. 225. Nezlin, R., Interactions between immunoglobulin G molecules, Immunol. Lett. 132, 1–5, 2010. 226. Philo, J.S., Is any measurement optimal for all aggregate sizes and types?, AAPS J. 8, E564–E571, 2006. 227. Franey, H., Brych, S.R., Kolvenbach, C.G., et al., Increased aggregation propensity of IgG2 subclass over IgG1: Role of conformational changes and covalent character in isolated aggregates, Protein Sci. 19, 1601–1615, 2010. 228. Gabrielson, J.P. and Arthur, K.K., Measuring low levels of protein aggregation by sedimentation velocity, Methods 54, 83–91, 2011. 229. http://www.nibsc.ac.uk/science/biotherapeutics/parenterals/albumin/control__standardisation.aspx. 230. Campbell, C., Shaw, R., Garinkle, B., and Gray, A., Gel permeation chromatography as a stability-indicating assay for human serum albumin, Dev. Biol. Standard. 44, 95–98, 1979. 231. Mitch, W.E. Jr. and Levy, C.C., Anomalous behavior of ribonuclease A on Sephadex G-100, Biochim. Biophys. Acta 251, 388–392, 1971. 232. Creamer, L.K. and Richardson, T., Anomalous behavior of bovine alpha s1- and betacaseins on gel electrophoresis in sodium dodecyl sulfate buffers, Arch. Biochem. Biophys. 234, 476–486, 1984. 233. Rinderle, S.J., Goldstein, I.J., and Remsen, E.E., Physicochemical properties of amaranthin, the lectin from Amaranthus caudatus seeds, Biochemistry 29, 10555–10561, 1990. 234. Francis, B., Schmidt, J., Yang, Y., et al., Anions and the anomalous gel iltration behavior of notexin and scutoxin, Toxicon 33, 779–789, 1995. 235. Iakooucheva, L.M., Kimzey, A.L., Masselon, C.D., et al., Aberrant mobility phenomena of the DNA repair protein XPA, Protein Sci. 10, 1353–1362, 2001. 236. Corradini, D., Effects of electrostatic and hydrophobic interactions on the chromatographic behavior of biopolymers in HPLC columns, Macromol. Symp. 110, 57–63, 1996. 237. Tarvers, R.C., Calcium-dependent changes in properties of human prothrombin: A study using high-performance size-exclusion chromatography and gel-permeation chromatography, Arch. Biochem. Biophys. 241, 639–648, 1985. 238. Lundblad, R.L., A hydrophobic site in human prothrombin present in a calciumstabilized conformer, Biochem. Biophys. Res. Commun. 157, 295–300, 1988. 239. Determann, H. and Walter, I., Source of aromatic afinity to “Sephadex” dextran gels, Nature 219, 604–605, 1968. 240. Martenson, R.E., The use of gel iltration to follow conformational changes in proteins. Conformational lexibility of bovine myelin basic protein, J. Biol. Chem. 253, 8887– 8893, 1978. 241. Lizárraga, B., Sánchez-Romero, D., Gil, A., and Melgar, E., The role of Ca2+ on pHinduced hydrodynamic changes of bovine pancreatic deoxyribonuclease A, J. Biol. Chem. 253, 3191–3195, 1978. 242. Kuroda, M., Kohira, Y., and Sasaki, M., Conformational change of skeletal muscle α-actinin induced by salt, Biochim. Biophys. Acta 1205, 97–104, 1994. 243. Fornstedt, T., Characterization of adsorption processes in analytical liquid–solid chromatography, J. Chromatogr. A 1217, 792–812, 2010.

Methods for the Analysis of Plasma and Plasma Protein Fractions

73

244. Girard, M., Mousseau, N., Bietiol, H., and Whitehouse, L.W., Selected examples of physicochemical methods for the analysis of biopharmaceuticals on the Canadian market, Pharm. Sci. 3, 9–14, 1997. 245. Mant, C.T., Kovacs, J.M., Kim, H.M., et al., Intrinsic amino acid side-chain hydrophilicity/ hydrophobicity coeficients determined by reversed-phase high-performance liquid chromatography of model peptides: Comparison with other hydrophilicity/hydrophobicity scales, Biopolymers 92, 573–595, 2009. 246. McCue, J.T., Theory and use of hydrophobic interaction chromatography in protein puriication applications, Methods Enzymol. 463, 405–414, 2009. 247. Rathore, A.S., Parr, L., Dermawan, S., et al., Large scale demonstration of a process analytical technology applications in bioprocessing: Use of on-line high performance chromatography for making real-time pooling decisions for process chromatography, Biotechnol. Prog. 26, 446–457, 2010. 248. Hunter, A.K., Wang, X., Suda, E.J., et al., Separation of product associated E. coli host cell proteins OppA and DppA from recombinant apolipoprotein A-Imilano in an industrial HIC unit operation, Biotechnol. Prog. 25, 446–453, 2009. 249. Jiang, C., Flansburg, L., Ghose, S., et al., Deining process design space for a hydrophobic interaction chromatography (HIC) puriication step: Application of quality by design (QbD) principles, Biotechnol. Bioeng. 107, 985–997, 2010. 250. Borberg, H., Jimenez, C., Belàk, M., et al., Treatment of autoimmune disease by immunomodulation through extracorporeal elimination and intravenous immunoglobulin, Transfus. Sci. 15, 409–418, 1994. 251. Hassl, A. and Aspöck, H., Puriication of egg yolk immunoglobulins. A two-step procedure using hydrophobic interaction chromatography and gel iltration, J. Immunol. Meth. 110, 225–228, 1988. 252. Valliere-Douglass, J., Jones, L., Shpekor, D., et al., Separation and characterization of an IgG2 antibody containing a cyclic imide in CDR1 of light chain by hydrophobic interaction chromatography and a cyclic imide in CDR1 of light chain by hydrophobic interaction chromatography and mass spectrometry, Anal. Chem. 80, 3168–3174, 2008. 253. Lu, Y., Williamson, B., and Gillespie, R., Recent advancement in application of hydrophobic interaction chromatography for aggregate removal in industrial puriication process, Curr. Pharm. Biotechnol. 10, 427–433, 2009. 254. Suzuki, T., Muroi, N., and Tomono, T., Interactions of human serum albumin with a modiied poly(vinyl alcohol) gel packing for high-performance liquid chromatography, J. Biomater. Sci. Polym. Ed. 1, 3–16, 1989. 255. Bjerrum, O.J., Bjerrum, M.J., and Heegaard, N.H., Electrophoretic and chromatographic differentiation of two forms of albumin in equilibrium at neutral pH: New screening techniques for determination of ligand binding to albumin, Electrophoresis 16, 1401–1407, 1995. 256. Lund, M., Bjeerum, O.J., and Bjerrum, M.J., Structural heterogeneity of the binding site of HSA for phenyl-groups and medium-chain fatty acids. Demonstration of equilibrium between different binding conformations, Eur. J. Biochem. 260, 470–476, 1999. 257. Ghosh, R. and Wang, L., Puriication of humanized monoclonal antibody by hydrophobic interaction membrane chromatography, J. Chromatogr. A 1107, 104–109, 2006. 258. Deitscher, R.W., Xiao, Y., O’Connell, J.P., et al., Protein instability during HIC: Evidence of unfolding reversibility, and apparent adsorption strength of disulide bondreduced α-lactalbumin variants, Biotechnol. Bioeng. 102, 1416–1427, 2009. 259. Zhang, L., Lu, D., and Liu, Z., Dynamic control of protein conformation transition in chromatographic separation based on hydrophobic interactions: Molecular dynamic simulation, J. Chromatogr. A 1216, 2483–2490, 2009. 260. Ueberbacher, R., Rodeler, A., Hahn, R., and Jungbauer, A., Hydrophobic interaction chromatography of proteins: Thermodynamic analysis of conformational changes, J. Chromatogr. A 1217, 184–190, 2010.

74

Biotechnology of Plasma Proteins

261. Gospodarek, A.M., Smatlak, M.E., O’Connell, J.P., and Fernandez, E.J., Protein stability and structure in HIC: Hydrogen exchange experiments and COREX calculations, Langmuir 27, 286–295, 2011. 262. Jungbauer, A., Machold, C., Hahn, R., et al., Hydrophobic interaction chromatography of proteins. III. Unfolding of proteins upon adsorption, J. Chromatogr. 1079, 221–228, 2005. 263. Xiao, Y., Freed, A.S., Jones, T.T., et al., Protein instability during HIC: Describing the effects of mobile phase conditions on instability and chromatographic retention, Biotechnol. Bioeng. 93, 1177–1189, 2006. 264. Wu, S.L., Figueroa, A., and Karger, B.L., Protein conformational effects in hydrophobic interaction chromatography: Retention characterization and the role of mobile phase additives and stationary phase hydrophobicity, J. Chromatogr. 371, 3–27, 1986. 265. Jones, T.T. and Fernandez, E.J., α-Lactalbumin tertiary structure changes on hydrophobic interaction chromatography surfaces, J. Colloid Interface Sci. 259, 27–35, 2003. 266. Purcell, A.W., Aguilar, M.I., and Hearn, M.T., Probing the binding behavior and conformational states of globular proteins in reversed-phase high-performance liquid chromatography, Anal. Chem. 71, 2440–2451, 1999. 267. Turner, N.A., Needs, E.C., Kahn, J.A., and Vulfson, E.N., Analysis of conformational states of Candida rugosa lipase in solution: Implications for mechanism of interfacial activation and separation of open and closed forms, Biotechnol. Bioeng. 72, 108–118, 2001. 268. Lin, F.Y., Chen, W.Y., and Hearn, M.T., Microcalorimetric studies on the interaction mechanism between proteins and hydrophobic solid surfaces in hydrophobic interaction chromatography: Effects of salts, hydrophobicity of the sorbent, and structure of the protein, Anal. Chem. 73, 3875–3883, 2001. 269. To, B.C. and Lenhoff, A.M., Hydrophobic interaction chromatography of proteins. I: The effects of protein and adsorbent properties on retention and recovery, J. Chromatogr. A 1141, 191–205, 2007. 270. Wakankar, A.A., Liu, J., Vandervelde, D., et al., The effect of cosolutes on the isomerization of aspartic acid residues and conformational stability in a monoclonal antibody, J. Pharm. Sci. 96, 1708–1718, 2007. 271. Xiao, Y., Rathore, A., O’Connell, J.P., and Fernandez, E.J., Generalizing a two-conformation model for describing salt and temperature effects on protein retention and stability in hydrophobic interaction chromatography, J. Chromatogr. A 1157, 197–206, 2007. 272. Ueberbacher, R., Rodler, A., Hahn, R., and Jungbauer, A., Hydrophobic interaction chromatography of proteins: Thermodynamic analysis of conformational changes, J. Chromatogr. A 1217, 184–190, 2010. 273. Gospodarek, A.M., Smatlak, M.E., O’Connell, J.P., and Fernandez, E.J., Protein stability and structure in HIC: Hydrogen exchange experiments and COREX calculations, Langmuir 27, 286–295, 2011. 274. Yan, S.B., Review of conformation-speciic afinity puriication methods for plasma vitamin K-dependent proteins, J. Mol. Recognit. 9, 211–218, 1996. 275. Smith, K.J., Immunoafinity puriication of factor IX from commercial concentrates and infusion studies in animals, Blood 72, 1269–1277, 1988. 276. Lundblad, R.L., Tsai, J., Wu, H.F., et al., Hydrophobic afinity chromatography of human thrombin, Arch. Biochem. Biophys. 302, 109–112, 1993. 277. Hoffer, L., Schwinn, H., and Josić, D., Production of highly puriied clotting factor IX by a combination of different chromatographic methods, J. Chromatogr. A 844, 119– 128, 1999. 278. Cuatrecasas, P., Wilchek, M., and Aninsen, C.B., Selective enzyme puriication by afinity chromatography, Proc. Natl. Acad. Sci. USA 61, 636–643, 1968.

Methods for the Analysis of Plasma and Plasma Protein Fractions

75

279. Jameson, G.W. and Elmore, D.R., Afinity chromatography of bovine trypsin: A rapid separation of bovine α- and β-trypsin, Biochem. J. 141, 555–565, 1974. 280. Wofsy, L. and Burr, B., The use of afinity chromatography for the speciic puriication of antibodies and antigens, J. Immunol. 103, 380–382, 1969. 281. Hornsey, V.X., Grifin, B.C., Pepper, D.C., et al., Immunoafinity puriication of factor VIII complex, Thromb. Haemost. 57, 102–105, 1987. 282. Croissant, M.P., van de Pol, H., Lee, H.H., and Allain, J.P., Characterization of four monoclonal antibodies to factor VIII coagulant protein and their use in immunopuriication of factor VIII, Thromb. Haemost. 56, 271–276, 1986. 283. Brettler, D.B., Forsberg, A.D., Levine, P.H., et al., Factor VIII; C concentrate puriied from plasma using monoclonal antibodies: Human studies, Blood 73, 1859–1863, 1989. 284. Grifith, M., Ultrapure plasma factor VIII produced by anti-F VIII c immunoafinity chromatography and solvent/detergent viral inactivation. Characterization of the Method M process and Hemoil M antihemophilic factor (human), Ann. Hematol. 63, 131–137, 1991. 285. Fuchs, S., Feferman, T., Meidler, R., et al., A disease-speciic fraction isolated from IVIG is essential for the immunosuppressive effect of IVIG in experimental autoimmune myasthenia gravis, J. Neuroimmunol. 194, 89–96. 286. Neuwelt, E.A., Frank, J.J., and Levy, C.C., Puriication of human spleen ribonuclease by immunoabsorption. Similarity of the enzyme with human liver ribonuclease, J. Biol. Chem. 251, 5752–5758, 1978. 287. Firer, M.A., Eficient elution of functional proteins in afinity chromatography, J. Biochem. Biophys. Meth. 49, 433–442, 2001. 288. Oh, H.K., Lee, J.M., Byuna, T.H., et al., Puriication of recombinant human B-domaindeleted factor VIII using anti-factor VIII monoclonal antibody selected by the surface plasmon resonance biosensor, Biotechnol. Prog. 17, 1119–1127, 2001. 289. Thompson, N.E., Foley, K.M., Stalder, E.S., and Burgess, R.R., Identiication, production, and use of polyol-responsive monoclonal antibodies for immunoafinity chromatography, Meth. Enzymol. 463, 475–494, 2009. 290. Bonde, M., Froskier, H., and Pepper, D.S., Selection of monoclonal antibodies for immunoafinity chromatography: Model studies with antibodies against soybean trypsin inhibitor, J. Biochem. Biophys. Meth. 23, 73–82, 1991. 291. Kummer, A. and Li-Chan, E.C.Y., Application of an ELISA-elution assay as a screening tool for dissociation of yolk antibody–antigen complexes, J. Immunol. Meth. 211, 125–137, 1998. 292. Singh, K.V., Kaur, J., Raje, M., et al., ELISA-based approach to optimize elution conditions for obtaining hapten-speciic antibodies, Anal. Bioanal. Chem. 377, 220–224, 2003. 293. Regault, V., Arvieux, J., Vallar, L, and Lecompte, T., Immunopuriication of human β2-glycoprotein I with a monoclonal antibody selected for its binding kinetics using a surface plasmon resonance biosensor, J. Immunol. Meth. 21, 191–197, 1998. 294. Bayry, J., Prabhudas, K., Bist, P., et al., Immunopuriication of foot and mouth disease virus type speciic antibodies using recombinant protein adsorbed to polystyrene wells, J. Virol. Meth. 81, 21–30, 1999. 295. Mallorquí, J., Llop, E., de Bolòs, C., et al., Puriication of erythropoietin from human plasma samples using an immunoafinity well plate, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 878, 2117–2122, 2010. 296. Thompson, N.E., Foley, K.M, Stalder, E.S., and Burgess, R.R., Identiication. production, and use of polyol-sensitive monoclonal antibodies for immunoafinity chromatography, Meth. Enzymol. 463, 475–494, 2009. 297. Lamberski, J.A., Thompson, N.E., and Burgess, R.R., Expression and puriication of a single-chain variable fragment antibody derived from a polyol-responsive monoclonal antibody, Protein Expr. Purif. 47, 82–92, 2006.

76

Biotechnology of Plasma Proteins

298. Ibarra, N., Caballero, A., González, E., and Valdés, R., Comparison of different elution conditions for the immunopuriication of recombinant hepatitis B surface antigen, J. Chromatogr. B Biomed. Sci. Appl. 735, 271–277, 1999. 299. Martins, S., Lourenco, S., Karmali, A., and Serralheiro, M.L., Monoclonal antibodies recognize conformational epitopes on wild-type and recombinant mutant amidases from Pseudomonas aeruginosa, Mol. Biotechnol. 37, 136–145, 2007. 300. Duellman, S.J. and Burgess, R.R., Antigen-binding properties of monoclonal antibodies reactive with EBNA1 and use in immunoafinity chromatography, PLoS One 4(2), e4614, 2009. 301. Longas, M.O., Ferguson, W.S., and Finlay, T.H., Studies on the interaction of heparin with thrombin, antithrombin, and other plasma proteins, Arch. Biochem. Biophys. 200, 595–602, 1980. 302. Saito, A. and Munakata, H., Analysis of plasma proteins that bind to glycosaminoglycans, Biochim. Biophys. Acta 1770, 241–248, 2007. 303. Hoffman, D.L., Puriication and large-scale preparation of antithrombin III, Am. J. Med. 87, 23S–26S, 1989. 304. Lebing, W.R., Hammond, D.J., Wydick III, J.E., and Baumbach, G.A., A highly puriied antithrombin III concentrate prepared from human plasma fraction IV-1 by afinity chromatography, Vox Sang. 67, 117–124, 1994. 305. Heger, A., Grunert, T., Schulz, P., et al., Separation of active and inactive forms of human antithrombin by heparin afinity chromatography, Thromb. Res. 106, 157–164, 2002. 306. Kleinova, M., Buchacher, A., Heger, A., et al., Exact molecular mass determination of various forms of native and de-N-glycosylated human plasma-derived antithrombin by means of electrospray ionization ion trap mass spectrometry, J. Mass Spectrom. 39, 1429–1436, 2004. 307. Demelbauer, U.M., Plematl, A., Josic, D., et al., On the variation of glycosylation in human plasma derived antithrombin, J. Chromatogr. A 1080, 15–21, 2005. 308. Kabir, S., Immunoglobulin puriication by afinity chromatography using protein A mimetic ligands prepared by combinatorial chemical synthesis, Immunol. Invest. 31, 263–278, 2002. 309. Skukla, A.A., Hubbard, B., Tressel, T., et al., Downstream processing of monoclonal antibodies—Application of platform approaches, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 848, 28–39, 2007. 310. Dancette, O.P., Taboreau, J.L., Tournier, E., et al., Puriication of immunoglobulins G by protein A/G afinity membrane chromatography, J. Chromatogr. B Biomed. Sci. Appl. 723, 61–68, 199. 311. Watanabe, H., Matsumaru, H., Ooishi, A., et al., Optimizing pH response of afinity between protein G and IgG Fc: How electrostatic modulations affect protein–protein interactions, J. Biol. Chem. 284, 12373–12383, 2009. 312. Wang, Y., Wu, S.-L., and Hancock, W.S., Approaches to the study of N-linked glycoproteins in human plasma using lectin afinity chromatography and nano-HPLC coupled to electrospray linear-ion-trap Fourier transform mass spectrometry, Glycobiology 16, 514–523, 2006. 313. Patwa, T., Li, C., Simeone, D.M., and Lubman, D.M., Glycoprotein analysis using protein microarrays and mass spectrometry, Mass Spectrom. Rev. 29, 830–844, 2010. 314. Fassina, G., Ruvo, M., Palombo, G., et al., Novel ligands for the afinity chromatographic puriication of antibodies, J. Biochem. Biophys. Meth. 49, 481–490, 2001. 315. Arnold, J.N., Wormald, M.R., Suter, D.M., et al., Human serum IgM glycosylation: Identiication of glycoforms that can bind to mannan-binding lectin, J. Biol. Chem. 280, 29080–29087, 2005. 316. Zhang, J.J., Xu, L.X., Zhang, Y., and Zhao, M.H., Binding capacity of in vitro deglycosylated IgA1 to human mesangial cells, Clin. Immunol. 119, 103–109, 2006.

Methods for the Analysis of Plasma and Plasma Protein Fractions

77

317. Xu, W., Chen, J., Yamasaki, G., et al., Lectin binding assays for in-process monitoring of sialylation in protein production, Mol. Biotechnol. 45, 248–256, 2010. 318. Jiang, H., Wu, S.L., Karger, B.L., and Hancock, W.S., Characterization of the glycosylation occupancy and the active site in the follow-on protein therapeutic: TNK-tissue plasminogen activator, Anal. Chem. 82, 6154–6162, 2010. 319. Krull, I.S. and Mazzeo, J.R., Capillary electrophoresis: The promise and the practice, Nature 357, 92–94, 1992. 320. Tripodi, V., Flor, S., Dobrecky, C., et al., Novel and highly sensitive mixed-polymeric electrokinetic chromatography system for determination of contaminants and impurities of heparin samples, Electrophoresis 31, 3606–3612, 2010. 321. Liu, Y., Fu, X., Bai, Y., et al., Improvement of reproducibility and sensitivity of CE analysis by using the capillary coated dynamically with carboxymethyl chitosan, Anal. Bioanal. Chem. 399, 2821–2929, 2011. 322. Benavente, F., Gimémez, E., Olivieri, A.C., et al., Estimation of the composition of recombinant erythropoietin mixtures using capillary electrophoresis and multivariate calibration methods, Electrophoresis 27, 4008–4015, 2006. 323. Apostol, I., Miller, K.J., Ratto, J., and Kelner, D.N., Comparison of different approaches for evaluation of the detection and quantitation limits of a purity method: A case study using a capillary isoelectrofocusing method for a monoclonal antibody, Anal. Biochem. 385, 101–106, 2009. 324. Jaworska, M., Cygan, P., Wilk, M., and Anuszewska, E., Capillary electrophoresis with indirect UV detection for the determination of stabilizer and citrates present in human albumin solutions, J. Pharm. Biomed. Anal. 50, 90–95, 2009. 325. Dalmora, S.L., D’Avila, F.B., da Silva, L.M., et al., Development and validation of a capillary zone electrophoresis method for assessment of recombinant human granulocyte colony-stimulating factor in pharmaceutical formulations and its correlation with liquid chromatography methods and bioassay, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 877, 2471–2476, 2009. 326. Lacher, N.A., Roberts, R.K., He, Y., et al., Development, validation, and implementation of capillary gel electrophoresis as a replacement for SDS-PAGE for purity analysis of IgG2 mAbs, J. Sep. Sci. 33, 218–227, 2010. 327. Alahmad, Y., Taverna, M., Mobdi, H., et al., A validated capillary electrophoresis method to check for batch-to-batch consistency during recombinant human glycosylated interleukin-7 production campaigns, J. Pharm. Biomed. Anal. 51, 882–888, 2010. 328. Kuzyk, M.A., Smith, D., Yang, J., et al., Multiple reaction monitoring-based, multiplexed, absolute quantitation of 45 proteins in human plasma, Mol. Cell. Proteomics 8, 1860–1877, 2009. 329. Jin, Y. and Manabe, T., Analysis of PEG-fractionated high-molecular-mass proteins in human plasma by non-denaturing micro 2-DE and MALDI-MS PMF, Electrophoresis 20, 3613–3621, 2009. 330. Ledford-Kraemer, M.R., Analysis of von Willebrand factor structure by multimer analysis, Am. J. Haematol. 85, 510–514, 2010. 331. Rhodes, D.G. and Loue, T.M., Determination of protein purity, Meth. Enzymol. 463, 9–19, 2009. 332. Fulcher, C.A., Roberts, J.R., Holland, L.Z., and Zimmerman, T.S., Human factor VIII procoagulant protein. Monoclonal antibodies deine precursor–product relationships and functional epitopes, J. Clin. Invest. 76, 117–124, 1985. 333. Eaton, D., Rodriguez, H., and Vehar, G.A., Proteolytic processing of human factor VIII. Correlation of speciic cleavages by thrombin, factor Xa, and activated protein C, Biochemistry 25, 505–512, 1986. 334. Burgess, R.R., Important but little known (or forgotten) artifacts in protein biochemistry, Meth. Enzymol. 463, 813–820, 2009.

78

Biotechnology of Plasma Proteins

335. Leung, J.P., Eshdal, Y., and Marchesi, V.T., Colonic tumor membrane-associated glycoprotein: Isolation of antigenically-active peptides after chemical cleavage, J. Immunol. 119, 664–670, 1977. 336. Peterson, D.M., Hirst, A., and Wehring, B., Comparison of normal and Bernard-Soulier platelet membrane glycoproteins. Isoelectric characteristics and surface radiolabel, J. Lab. Clin. Med. 100, 26–36, 1982. 337. Casanova, M., Lopoez-Ribot, J.L., Martinez, J.P., and Sentandreu, R., Characterization of cell wall proteins from yeast and mycelial cells of Candida albicans by labeling with biotin: Comparison with other techniques, Infect. Immun. 60, 4898–4906, 1992. 338. Miller, I., Crawford, J., and Gianazza, E., Protein stains for proteomic applications: Which, when, why?, Proteomics 6, 5385–5408, 2006. 339. Steinberg, T.H., Protein gel staining methods: An introduction and overview, Meth. Enzymol. 463, 541–563. 340. Seeman, N.C., Chen, J.H., and Kallenbach, N.R., Gel electrophoretic analysis of DNA branched junctions, Electrophoresis 10, 345–354, 1989. 341. Miernyk, J.A. and Thelen, J.J., Biochemical approaches for discovering protein–protein interactions, Plant J. 53, 597–609. 2008. 342. Schägger, H., Cramer, W.A., and von Jagow, G., Analysis of molecular masses and oligomeric states of protein complexes by blue native electrophoresis and isolation of membrane protein complexes by two-dimensional native electrophoresis, Anal. Biochem. 217, 220–230, 1994. 343. Grandier-Vazeille, X. and Guérin, M., Separation by blue native and colorless native polyacrylamide gel electrophoresis of the oxidative phosphorylation complexes of yeast mitochondrial solubilized by different detergents: Speciic staining of the different complexes, Anal. Biochem. 242, 248–254, 1996. 344. Thangthaeng, N., Sumien, N., Forster, M.J., et al., Nongradient blue native gel analysis of serum proteins and in-gel detection of serum esterase activities, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 879, 386–394, 2011. 345. Zickermann, V., Wumaier, Z., Wrzesniewska, B., et al., Native immunoblotting of blue native gels to identify conformation-speciic antibodies, Proteomics 10, 159–163, 2010. 346. Choi, N.S., Choi, J.H., Kim, B.H., et al., Mixed-substrate (glycerol tributyrate and ibrin) zymography for simultaneous detection of lipolytic and proteolytic enzymes on a single gel, Electrophoresis 30, 2234–2236, 2009. 347. Englebienne, P., Immune and Receptor Assays in Theory and Practice, CRC Press, Boca Raton, FL, 2000. 348. Popov, J., Zhukov, O., Ruden, S., et al., Performance and clinical utility of a commercial von Willebrand factor collagen binding assay for laboratory diagnosis of von Willebrand disease, Clin. Chem. 52, 1965–1967, 2006. 349. Komorowic, E., McBane III, R.D., Charleworth, J., and Fass, D.N., Reduced high shear platelet adhesion to the vascular media: Defective von Willebrand factor binding to the interstitial collagen, Thromb. Haemst. 87. 763–770, 2002. 350. Li, P.D., Galanakis, D.K., Ghebrehiwet, B., and Peerschke, E.I.B., The receptor for the globular “heads” of C1q, gC1q-R, binds to ibrinogen/ibrin and impairs its polymerization, Clin. Immunol. 90, 360–367, 1999. 351. Leppänen, A. and Cummings, R.D., Fluorescence-based solid-phase assays to study glycanbinding protein interactions with glycoconjugates, Meth. Enzymol. 478, 241–264, 2010. 352. Grinnell, F. and Feld, M.K., Adsorption characteristics of plasma ibronectin in relation to biological activity, J. Biomed. Mater. Res. 15, 363–381, 1981. 353. Hu, W.J., Eaton, J.W., Ugarova, T.P., and Tang, L., Molecular basis of biomaterialmediated foreign body reactions, Blood 98, 1231–1238, 2001. 354. Kiel, C., Beltrao, P., and Serrano, L., Analyzing protein interaction networks using structural information, Annu. Rev. Biochem. 77, 415–441, 2008.

Methods for the Analysis of Plasma and Plasma Protein Fractions

79

355. Wang, X., Peng, Y., Ma, Y., and Jahroudi, N., Histone H1-like protein participates in endothelial cell-speciic activation of the von Willebrand factor promoter, Blood 104, 1725–1732, 2004. 356. Wilton, R., Yousef, M.A., Saxena, P., et al., Expression and puriication of recombinant human receptor for advanced glycation end products in Escherichia coli, Protein Expr. Purif. 47, 25–35, 2006. 357. Kanoh, Y., Egawa, S., Baba, S., and Akahoshi, T., Association of IgG N-linked oligosaccharide chains and proteases in sera of prostate cancer patients with and without α2-macroglobulin deiciency, J. Clin. Lab. Anal. 23, 125–131, 2009. 358. Mátrai, Z., Németh, J., Miklós, K., et al., Serum β2-microglobulin measured by immunonephelometry: Expression patterns and reference intervals in healthy adults, Clin. Chem. Lab. Med. 47, 585–589, 2009. 359. Bradwell, A.R., Harding, S.J., Fourrier, N.J., et al., Assessment of monoclonal gammopathies by nephelometric measurement of individual immunoglobulin kappa/lambda ratios, Clin. Chem. 55, 1646–1655, 2009. 360. Di Stasio, E., Romitelli, F., Lancellotti, S., et al., Kinetic study of von Willebrand factor self-aggregation induced by ristocetin, Biophys. Chem. 144, 101–107, 2009. 361. Balakrishnan, K., Andrei-Selmer, L.C., Selmer, T., et al., Comparison of intravenous immunoglobulins for naturally occurring autoantibodies against amyloid-β, J. Alzheimer’s Dis. 20, 135–143, 2010. 362. de Kat Angelino, C.M., Raymkers, R., Teunesen, M.A., et al., Overestimation of serum κ chain concentration by immunonephelometry, Clin. Chem. 56, 1188–1190, 2010. 363. Kwon, K.A., Kim, S.H., Oh, S.Y., et al., Clinical signiicance of preoperative serum vascular endothelial growth factor, interleukin-6, and C-reactive protein level in colorectal cancer, BMC Cancer 10, 203, 2010. 364. Michiels, J.J., Berneman, Z., Gadisseur, A., et al., Classiication and characterization of hereditary types 2A, 2C, 2D, 2E, 2M, 2N, and 2U (unclassiiable) von Willebrand disease, Clin. Appl. Thromb. Hemost. 12, 397–420, 2006. 365. Bjerrum, O.J., Ingild, A., Lowenstein, H., and Weeke, B., Quantitation of human IgG by rocket immunoelectrophoresis at pH 5 by use of carbamylated antibodies. A routine laboratory method, Clin. Chim. Acta 46, 337–343, 1973. 366. Pajdak, W., Radwan, J., and Guzik, T.J., Cleavage of prothrombin bound in immune complexes results in high thrombin enzymatic activity, J. Physiol. Pharmacol. 55, 477–484, 2004. 367. Nielsen, H.M., Minthon, L., Londos, E., et al., Plasma and CSF serpins in Alzheimer disease and dementia with Lewy bodies, Neurology 69, 1569–1579, 2007. 368. Sapan, C.V., Lundblad, R.L., and Price, N.C., Colorimetric protein assay techniques, Biotechnol. Appl. Biochem. 29, 99–108, 1999. 369. Vistnes, M., Christensen, G., and Omland, T., Multiple cytokine biomarkers in heart failure, Expert Rev. Mol. Diagn. 10, 147–157, 2010. 370. Thillaivinayagalingam, P., Gommeaux, J., McLoughlin, M., et al., Biopharmaceutical production: Application of surface plasmon resonance biosensors, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 878, 149–153, 2010. 371. Lausted, C., Hu, Z., Hood, L., and Campbell, C.T., SPR imaging for high throughput, label-free interaction analysis, Comb. Chem. High Throughput Screen. 12, 741–751, 2009. 372. McGraw, R.A., Davis, L.M., Lundblad, R.L., et al., Structure and function of factor IX: Defects in haemophilia B, Clin. Heamatol. 14, 359–383, 1985. 373. Mikami, S., O’Brine, D.P., Mellars, G., et al., Studies on immunological assay of vitamin-K dependent factors. III. A double monoclonal immunoradiometric assay for factor IX antigen, Br. J. Haematol. 62, 513–524, 1986. 374. Tsang, T.C., Bentley, D.R., Mibashan, R.S., and Giannelli, F., A factor IX mutation, veriied by direct genomic sequencing, causes haemophilia B by a novel mechanism, EMBO J. 7, 3009–3015, 1988.

80

Biotechnology of Plasma Proteins

375. Jagt, H.J., Bekkers, M.L., van Bommel, S.A., et al., The inluence of the inactivating agent on the antigen content of inactivated Newcastle disease vaccines assessed by the use of the in vitro potency test, Biologicals 38, 128–134, 2010. 376. Cuervo, M.L., Sterling, A.L., NIcot, I.A., et al., Validation of a new alternative for determining in vitro potency in vaccines containing hepatitis B from two manufacturers, Biological 36, 375–382, 2008. 377. Keller, J.E., Characterization of new formalin-detoxiied botulinum neurotoxoids, Clin. Vaccine Immunol. 15, 1374–1379, 2008. 378. European Convention for the protection of vertebrate animals used for experimental and other scientiic purposes, ETSNo. 123, Strasbourg, France. Council of Europe, March, 1986. 379. Karnaukhova, E., Golding, B., and Ophir, Y., Development and evaluation of an ELISA for quantiication of human alpha-1-proteinase inhibitor in complex biological mixtures, Biologicals 35, 285–295, 2007. 380. López-Expósito, I., Chicón, R., Belloque, J., et al., Changes in the ovalbumin proteolysis proile by high pressure and its effect on IgG and IgE binding, J. Agric. Food Chem. 56, 11809–11816, 2008. 381. Maasch, H.J. and Marsh, D.G., Standardized extracts modiied allergens—Allergoids, Clin. Rev. Allergy 5, 89–106, 1987. 382. Singer, S.J., Structure and function of antigen and antibody proteins, in The Proteins, 2nd edn., ed. H. Neurath, Vol. III, Chapter 15, pp. 269–357, Academic Press, New York, 1965. 383. Boyd, W.C., The proteins of immune reactions, in The Proteins, eds. H. Neurath and K. Bailey, Vol. II, Pt. B., Chapter 22, pp. 755–844, Academic Press, New York, 1954. 384. Wells, H.G., Studies on the chemistry of anaphylaxis, J. Infect. Dis. 6, 506–522, 1909. 385. Wells, H.G., Studies on the chemistry of anaphylaxis, J. Infect. Dis. 5, 449–483, 1908. 386. Noda, Y., On the biuret reaction with peptides, Bull. Chem. Soc. Jpn. 40, 1264–1265, 1967. 387. Hortin, G.L. and Meilinger, B., Cross-reactivity of amino acids and other compounds in the biuret reaction: Interference with urinary peptide measurements, Clin. Chem. 51, 1411–1419, 2005. 388. Landsteiner, K., Serological reactivity of hydrolytic products from silk, J. Exp. Med. 75, 269–276, 1942. 389. Wornall, A., The immunological speciicity of chemically altered proteins, J. Exp. Med. 51, 295–317, 1930. 390. Johnson, L.R. and Wornall, A., CXLII. The immunological properties of alkali-treated proteins, Biochem. J. 26, 1202–1213, 1932. 391. von Pechmann, E., Über die enzymatische hydrolyse von xanthoproteinen und deren verwendung zur colorimetrischen bestimmung proteolytische fermente, Biochem. Zeitschrift 321, 248–250, 1950. 392. Drăguşanu, M., Petre, B.-A., and Przybylski, M., Epitope motif of an anti-nitrotyrosine antibody speciic for tyrosine-nitrated peptides revealed by a combination of afinity approaches and mass spectrometry, J. Pept. Sci., 17, 184–191, 2011. 393. Chou, M.Y., Hartvigsen, K., Hansen, L.F., et al., Oxidation-speciic epitopes are important targets of innate immunity, J. Intern. Med. 263, 479–488, 2008. 394. Schwartz-Albiez, R., Monteiro, R.Z.C., Rodriguez, M., et al., Natural antibodies, intravenous immunoglobulin and their role in autoimmunity, cancer and inlammation, Clin. Exp. Immunol. 158(Suppl. 1), 43–50, 2009. 395. Binder, C.J., Natural IgM antibodies against oxidation-speciic epitopes, J. Clin. Immunol. 30(Suppl. 1), S56–S60, 2010. 396. van Beers, M.M., Sauerborn, M., Gilli, F., et al., Aggregated recombinant human interferon beta induces antibodies but no memory in immune-tolerant transgenic mice, Pharm. Res. 27, 1812–1824, 2010.

Methods for the Analysis of Plasma and Plasma Protein Fractions

81

397. Richard, J. and Prang, N., The formulation and immunogenicity of therapeutic proteins: Product quality as a key factor, IDrugs 13, 550–558, 2010. 398. van Beers, M.M., Jiskoot, W., and Schellekens, H., On the role of aggregates in the immunogenicity of recombinant human interferon beta in patients with multiple sclerosis, J. Interferon Cytokine Res. 30, 767–775, 2010. 399. Singh, S.K., Impact of product-related factors on immunogenicity of biotherapeutics, J. Pharm. Sci. 100, 354–387, 2011. 400. Zaborsky, N., Brunner, M., Wallner, M., et al., Antigen aggregation decides the fate of the allergic immune response, J. Immunol. 184, 725–735, 2010. 401. Tieugabulova, D., Falcon, V., Penton, E., et al., Aggregation of recombinant hepatitis B surface antigen induced in vitro by oxidative stress, J. Chromatogr. B Biomed. Sci. Appl. 736, 153–166, 1999. 402. Kriegshaeuser, G., Kuechier, E., and Skern, T., Aggregation-associated loss of antigenicity observed for denatured virion protein 1 of equine rhinitis A virus in an enzymelinked immunosorbent assay, Virus Res. 143, 130–133, 2009. 403. Zhao, H.L., Xiue, D., Wang, Y., et al., Elimination of the free sulfhydryl group in the human serum albumin (HSA) moiety of human interferon α2b and HSA fusion protein increases its stability against mechanical and thermal stress, Eur. J. Pharm. Biopharm. 72, 405–411, 2009. 404. Fradkin, A.H., Carpenter, J.F., and Randolph, T.W., Immunogenicity of aggregates of recombinant human growth hormone in mouse models, J. Pharm. Sci. 96, 3247–3264, 2009. 405. Rademecker, R.P., Renard, E., and Scheen, A.J., Circulating insulin antibodies: Inluence of continuous subcutaneous or intraperitoneal insulin infusion, and impact on glucose control, Diabetes Metabol. Res. Rev. 25, 491–501, 2009. 406. Nezlin, R., Interactions between immunoglobulin G molecules, Immunol. Lett. 132, 1–5, 2010. 407. Kyte, J., Structure in Protein Chemistry, 2nd edn., Garland Science, New York, 2007. 408. Lundblad, R.L., Approaches to the Conformational Analysis of Biopharmaceuticals, CRC Press, Boca Raton, FL, 2010. 409. Sherwood, D. and Cooper, J., Crystals, X-rays and Proteins: Comprehensive Protein Crystallography, Oxford University Press, New York, 2011. 410. Wallace, K.K. and Moreira, A.R., Changes in biologics regulations: Impact on the development and validation of the manufacturing processes for well-characterized products, in Validation of Biopharmaceutical Manufacturing Processes, Chapter 13, pp. 170–179, American Chemical Society, Washington, DC, 1998. 411. White, F.M., The potential cost of high-throughput proteomics Sci. Signal. 4, pe8, 2011. 412. Schiestl, M., Stangler, T., Torella, C., et al., Acceptable changes in quality attributes of glycosylated biopharmaceuticals, Nat. Biotechnol. 29, 310–312, 2011.

4

Albumin

I started this chapter in the middle of a North Carolina winter, and it is now the beginning of a North Carolina summer. In retrospect, I see that I started with an incredible amount of naivety in expecting that this chapter would be inished in early spring. Albumin is much more complex than a standard for protein determination or even a bottle of opaque solution coming out of pasteurization in the manufacturing plant. I admit that I have been humbled by this chapter and by the distinguished group of scientists who worked on albumin in the days of World War II. The material has been assembled in a manner that I hope is most useful to those interested in the development of albumin as a biotechnology product. I would be remiss if I did not irst direct the reader to the comprehensive treatise on albumin by Theodore Peters,1 which was published in 1996. He notes that the irst isolation of albumin was accomplished from urine in 1500. The term albumin dates back to 1853 when work was done on vegetable and egg proteins.2 Thus, the substance derived from egg white was described as an albuminoid substance. The term albumin was used to describe the natural substances that were later identiied as proteins; for example, ibrin was described as coagulated albumin. The name albumin is derived from L. albus (white), as the various protein precipitates obtained from acidiied urine, egg white, or serum were white in color. Albumen, an early German term for protein,1 was the preferred term as late as 19033 and is used to describe the protein obtained from the egg white of various birds.4 Any student interested in the history of albumin is directed to both Peter’s work on albumin1 and Sandor’s broader consideration of plasma proteins.2 Sandor states that the separation of albumin from serum was irst described by Denis in 18405 and Panum in 1852.6 There was some tension between various investigators at this time as to who discovered what and when and what to call the material that had been discovered. It gives me some pleasure to realize that the same battles fought today over priority were fought much earlier with similar intensity. It would appear that Denis was also one of the irst (if not the irst) to use dialysis in the preparation of proteins.1 The initial classiication of proteins was based on solubility,5–9 with such a classiication applicable to both animal and plant proteins.10 In this scheme, albumin was described as being soluble in water and heat-coagulable, prolamines were soluble in alcohol, and globulins were soluble in neutral salt solutions. The differential solubility of albumin and the several globulin fractions11 formed the basis for the alcohol fractionation of plasma described in Chapter 2. The late Charles Tanford presented an excellent discussion of the contributions of Cohn and Edsall to the development of a globular model for a protein based on these earlier studies on protein solubility.12 This work had the by-product of providing the necessary basic science underlying the development of the alcohol fractionation of plasma (see Chapter 2).

83

84

Biotechnology of Plasma Proteins

Much of the early work13–16 on protein chemistry was performed with ovalbumin, as the preparation of this protein from eggs was more facile than the preparation of albumin from plasma or serum; the albumins, casein, and gelatin were the only proteins available in a reasonably homogeneous form and substantial quantity in the early part of the twentieth century.17 The investigation of food allergies was another driving force for the study of the immunochemistry of ovalbumin.18–21 Studies on the conversion of ovalbumin to plakalbumin22,23 provided some of the basis for the developing concept of limited proteolysis in the mechanism of the action of serpins.24 It is also important to note that the Hofmeister classiication of electrolytes25,26 is based on Hofmeister’s fundamental work on the effect of salts on ovalbumin, which was published in 1888.27 Ovalbumin continues to be an important model protein for studies on protein chemistry28,29 and immunology.30,31 The presence of two phosphorylated residues in ovalbumin has allowed this protein to serve as a model for the study of phosphoproteins.32,33 Human albumin shares extensive homology with bovine albumin,34,35 and bovine serum albumin (BSA) was evaluated for therapeutic use during World War II.36 The development of BSA as a parenteral therapeutic continued until 1943 when there was a second fatality due to “serum sickness” following the administration of the bovine product.37 A program for the development of human albumin had started in 194037 under the direction of Edwin Cohn (see Chapter 2), and human serum albumin (HSA) continues to have considerable clinical use (see below). There is a remarkably high prevalence of human anti-BSA immunoglobulin G (IgG) in the normal population showing prior exposure to the bovine antigen. Andersen and coworkers38 detected anti-BSA IgG in 95% (99 of 104 individuals in a gender-mixed population) of blood donors, while Mogues and coworkers39 found anti-BSA antibodies in 55% of the blood donor population at their institution. Mogues and coworkers39 did note that the elevated levels of anti-BSA antibody were not associated with a speciic clinical event. There are, however, sporadic reports of adverse reactions to therapeutics containing BSA as an excipient.40–43 Currently, BSA is an important model protein for studies in solution chemistry as well as a standard used in methods for the determination of protein concentration. It should be noted that most researchers with an interest in the relationship between structure and function in albumin consider the human and bovine proteins equivalent. Albumin in human plasma (HSA) constitutes approximately 40% of the total body albumin, with the remainder scattered in the various extravascular spaces.44 Hughes described albumin as an interstitial protein rather than a plasma protein in 1954.9 Rothschild and coworkers45 used 131I-labeled HSA to determine the tissue distribution of albumin in several individuals with terminal diseases. These researchers observed that 4–5 days were required for complete tissue equilibration. The major amount of radiolabel was found in plasma (40%) with 18% in skin and 15% in muscle; there was a minor amount in heart, lungs, kidney, and spleen. The presence of a large amount of interstitial luid in skin is thought to be responsible for the albumin in skin.46 A word of comment on the Rothschild study45 is appropriate before continuing the discussion. One of the coauthors, Rosemary Yalow, was a recipient of the Nobel Prize in Physiology or Medicine in 1977. She was the second woman to be awarded a Nobel Prize. The prize recognized the development of the radioimmunoassay by Yalow and Berson. Sherman Berson died in 1972 and did not share the Nobel Prize. The presence of albumin in the

Albumin

85

extravascular space has been known for more than 50 years.44,45 Increased leakage into the extravascular space is considered to be responsible for lower HSA concentrations in acute-phase reactions and other clinical situations.47–49 The term transcapillary escape rate50 is used to describe the process. The amount of work published on HSA prior to 1940 is sparse indeed. A search for publications on “HSA” appearing prior to 1940 using SciFinder yielded 14 papers; some 24,403 papers have appeared since 1940. It is important to recognize that 1940 was well before the advent of chromatographic techniques for protein puriication, and albumin was then separated from the globulins on the basis of solubility.2,51 Notwithstanding the lack of sophisticated techniques, crystalline HSA was prepared by two groups before 1940.52,53 Oswald52 obtained serum albumin from ascites luid, while Adair and Taylor53 obtained serum albumin from serum heated at 56°C for 25 min. The globulin fraction was removed in half-saturated ammonium sulfate prior to the crystallization of albumin. Most of the work prior to 1900 focused on the characterization of albumin (albumen) as a colloid. The properties of albumin as a colloid are discussed in greater detail next. Examples of this work included the differential iltration of colloids and crystalloids by C.J. Martin in 189654 and the differential heatdenaturation (heat-coagulation) of albumin (albumen) by Hewlett in 1892.55 Kylin56 determined the isoelectric point of human serum albumin and found several components, varying from four to six. The molecular weight of HSA was estimated by Roche and coworkers in 1935 to be 69,000 da.57 It is of interest to note that some years later, the molecular weight of HSA was estimated to be 67,000 da by sodium dodecyl sulfate (SDS)-electrophoresis58 or by analytical ultracentrifugation.59 The irst clinical application of HSA is likely that of Mann in 1918,60 who used homologous serum to treat shock in a canine model. It seems like the therapeutic effect observed with the infusion of serum in the canine shock model was due to the albumin present in the serum. The current work on HSA can be traced to activities on both sides of the Atlantic Ocean before and during World War II.61 This book,61 which was edited by General Douglas Kendrick (U.S. Army), compiled all of the various projects associated with the procurement, processing, and use of blood during World War II. It is one of a multivolume set assembled to ensure that the knowledge gained during World War II would not be lost. General Kendrick states in the Preface to this volume that information gained in World War II should have been readily available for the Korean conlict, but the totality of information was not available for implementation. This disconnect in the availability of information served as the basis for the compilation of the volume entitled Blood Programs in World War II,61 so that such issues would not be future problems. Even some 50 years later, the material in this volume has considerable value to current biotechnology. The clinical need was illustrated in a landmark study by Ebert and coworkers62 on the effect of blood loss on circulatory collapse. This group of researchers, including Eugene Stead, Jr., who later built the Department of Medicine at Duke University after serving at Emory University, established that luid replacement was not adequate but could be effectively treated with plasma or whole blood. The removal of blood was followed by a slow recovery of plasma volume and the replacement of a normal plasma protein complement. The infusion of saline was not suficient to return the circulatory volume to normal. As a note, Eugene Stead is also known for

86

Biotechnology of Plasma Proteins

his work in establishing the profession of physician assistant. Several years later, Heyl and coworkers36 demonstrated the effectiveness of HSA from the Cohn alcohol fractionation process in the treatment of acute blood loss. The rationale for this approach was provided by Scatchard and coworkers63 in their study on the osmotic pressure of plasma and albumin, in which they reported an increase of approximately 20 mL of plasma volume per gram of albumin. This is an example of albumin acting as a colloid, with a colloid being deined as an amorphous substance that does not diffuse through membranes such as dialysis membranes and is not readily crystallized. Other examples of colloids include dextrans; colloids are thought to be responsible for the osmotic properties of blood. Crystalloids are chemicals such as sodium chloride, which readily diffuse through membranes and can be readily crystallized. There is a lively debate on the relative merits of colloids and crystalloids for therapeutic use.64–68 There appears to be little doubt as to the intrinsic safety of albumin, but questions regarding its eficacy in all clinical situations remain.69–71 There has been concern that plasma volume expanders such as dextran, gelatin, and hydroxyethyl starch have a negative effect on hemostasis.72 I would again be remiss if I did not note in passing that George Scatchard (cited earlier) developed the mathematical procedures for evaluating the binding of molecules to receptors, including the binding of ions to proteins.73 This procedure is visualized graphically by what is known as a Scatchard plot.74

STRUCTURE OF ALBUMIN HSA is a single-chain protein with a molecular weight of approximately 67 kDa.75–79 A study by Debaiczyk and coworkers77 on the sequence of HSA derived from cDNA clone obtained from isolated mRNA showed the presence of a hydrophobic propeptide followed by a basic propeptide. The mature protein contains 585 amino acids with 17 disulide bonds and one free sulfhydryl group. The disulide bonds and the free sulfhydryl groups have been suggested to be involved in the redox function of albumin.80–82 The disulide pattern in the recombinant albumin produced in Pichia pastoris is the same as the pattern in the albumin derived from human plasma.83 The crystal structure of HSA has been reported by He and Carter84 and later by Sugio and coworkers,85 showing a three-domain structure representing a heart-shaped conformation. Leggio and coworkers86 showed the formation of a somewhat expanded conformation for HSA.87 The crystal structure of horse serum albumin has been reported to be similar to that of HSA.88 The binding of 2,3,5-triiodobenzoic acid to HSA is slightly tighter than it is to horse serum albumin, but the binding sites observed in the crystal structure are similar. A schematic structure of albumin is shown in Figure 4.1. J. L. Oncley, one of the members of the Cohn laboratory group,11,12 published a review of his work on the dielectric constant and structure of HSA in 2003.89 Oncley discussed the frustration of obtaining different values of the dipole moment with different albumin preparations. Working with Howard Dintzis, Oncley determined that the variation in the value of the dipole moment was related to the fatty acid content of the albumin preparation. Later work established the effect of fatty acids on albumin conformation.90 As will be shown later, the effect of fatty acids on albumin conformation is one of the several examples of the solution structure of HSA being

87

Albumin Cysteine 34

IIIB IB

SH IA

Site 2 IIA

IIIA

Tyr411

Site 1 IIB

Lys199

FIGURE 4.1 A schematic structure of albumin. This work has been adapted from various reviews, including Sugio et al.,85 Kragh-Hansen et al.,424 Quinlin et al.,685 Kratz421 and Leggio et al.86 It must be emphasized that the drawing is schematic and lacks the detail of a crystal structure. In particular, the Sudlow sites are not as discrete as shown but may overlap.873 There are also discrete binding sites for fatty acids, which are not shown.

inluenced by the binding of ligands and solvent properties. As an aside, Oncley’s interest in the study of albumin was driven by his interest in the use of the dielectric constant to measure the properties of proteins in solution91 and the availability of puriied albumin in the Cohn laboratory.89 The measurement of the dielectric constant of a protein is a potentially powerful technique that can be complex to interpret92 but can provide useful information.93,94 Oncley and coworkers used the determination of the dielectric constant (relaxation time) in postulating an elongated ellipsoid shape for HSA. Following up on his initial frustrations on the presence of fatty acid in his albumin samples that provided erratic results, Oncley and coworkers used studies on the dielectric constant to establish a change in solution behavior on the binding of fatty acids.95 Later studies by other researchers using dynamic light scattering96 suggested a compact conformation at pH 7.4 (Stokes radius of 60 Å), which increases to approximately 80 Å at pH 8.0 or pH 5.4. The effect of pH on albumin conformation is discussed in more detail later. Further support for the conformational lexibility of albumin is provided by studies with poly(ethylene)glycol (PEG). Farruggia and coworkers97 observed that PEG 1000 destabilized the HSA, while PEG 10000 stabilized the HSA. These researchers suggested that the smaller PEG molecules are small enough to get inserted into the hydrophobic regions of the protein, while the high-molecular-weight PEG would be excluded. There is another explanation based on the crowding concept. The highmolecular-weight PEG could be functioning as a more effective crowding agent,98,99 forcing the HSA into a more compact and likely more stable conformation. Ragi and coworkers100 studied the interaction of PEG 3500 with HSA and observed little change in its helical content at a low concentration with a reduction in α-helix and an increase

88

Biotechnology of Plasma Proteins

in β-turn at a higher concentration (1 mM). A binding (afinity) constant of 4.12 × 105 M for PEG was observed by these researchers. These researchers suggested that PEG stabilized at low concentrations but destabilized at high concentrations. The discrepancy in results with PEG may be due to contaminants such as peroxides.101 The effect of pH on the conformation of albumin has been a subject of study for the past 60 years. While the extremes of pH are unlikely to be of any direct physiological signiicance, the study of the transitions has been useful in understanding the factors involved in the binding of drugs and other organics. The conformational transition at a low pH is called the N → F transition. The initial observation of this phenomenon was reported by Luetscher102 in 1949 and was based on electrophoretic heterogeneity at a low pH. Foster103 has reviewed the earlier titration data on HSA and reported anomalous behavior below the isoelectric point. Electrophoretic heterogeneity at pH 4.0 was not observed by Schmid in the presence of Zn2+ with HSA with a blocked sulfhydryl group.104 Schmid later observed105 that the electrophoretic heterogeneity at pH 4.0 did depend on the buffer anion; the heterogeneity decreased with the increasing size of aliphatic acid in the solvent. The N (normal) → F (fast) transition was based on the faster rate of migration of albumin in free-boundary electrophoresis at an acidic pH.102 It should be noted that these changes in electrophoretic mobility do not represent heterogeneity in either HSA or BSA; electrophoretic mobility is observed with genetic variants.106 Tanford and coworkers107 studied the titration behavior of BSA and HSA and observed anomalies below pH 4.3 and above pH 10.3, which suggested an expansion of the protein. Subsequent work108 showed that BSA expanded in a solution below pH 4.3 and above pH 10.5 on the basis of the changes in the intrinsic viscosity. The observed changes were fully reversible. Wilting and coworkers109 used circular dichroism (CD) to study the conformation changes in HSA with or without warfarin in the pH range of 6–9. A change in molar ellipticity as a function of pH (5 → 9) was similar for HSA in the presence or absence of warfarin; a change occurred over a smaller pH range (5 → 8) in the presence of 2.5 mM CaCl2. Kasai-Morita and coworkers110 used intrinsic luorescence to demonstrate warfarin-binding increases with an increasing pH. An earlier work111 showed that the base transition could either increase or decrease the binding of drugs; the binding of indomethacin increased the binding of L-tryptophan and quinidine, with no change in the binding of salicylic acid or phenytoin. In other studies by Kosa and coworkers,112 it is suggested that the N (normal) → B (base) transition has a role in the binding and release of ligands. The ionization of histidyl residues in domain I is suggested by Bos and coworkers to be responsible for the N → B transition.113 The N → B transition is similar to the N → F transition but is not associated with a change in electrophoretic mobility. The binding of zinc to albumin is associated with a conformational change that is similar to that observed in the base transition.114 There is a further conformational transition observed in BSA when the pH is lowered from 4.0 to 2.0, which is characterized by a marked increase in the frictional ration.115 This conformational change is referred to as the F → E transition, which is also reversible, and likely involves the full extent of protein unfolding permitted by the 17 disulide bonds; the F → E transition has also been referred to as the acid expansion.116 Geisow and Beaven subjected HSA to digestion with pepsin at an acidic pH117 and observed that there was a difference in digestion patterns (electrophoretic

89

Albumin

analysis) between pH 3.7 and pH 3.5; there were also small differences in the patterns obtained with HSA and BSA. Subsequent work from this laboratory118 showed that the change in intrinsic luorescence in several isolated fragments (P44, residues 1–386; P29, 49–307 representing the amino–terminal portion of HSA) was less than that observed in the intact protein. This suggested that the carboxyl terminal portion of HSA is critical for N → F transition. Bilirubin bound to both fragments showed CD changes that were similar to those observed with the binding of bilirubin to intact albumin. These researchers also studied the binding of 8-anilinonaphthalene1-sulfonate to the large fragments and obtained results consistent with binding site(s) conined to the P44 fragment. Earlier, Weber and Young119 had subjected a BSAdimethylaminonapthalene-1-sulfonate complex to digestion with pepsin, observing a decrease in luorescence polarization. Weber and Young conducted their digestion studies at pH 2.0, while Geisow and Beaven worked at either pH 3.5 or pH 3.7. A consideration of the two studies suggests that the F → E transition (at pH 2.0) likely involved further conformational change in the albumin molecule, relecting the protonation of additional side chain carboxyl groups. While the average pKa for the β-carboxyl of aspartic acid in proteins is 3.5, the range is 0.5–9.2; for the γ-carboxyl of glutamic acid, the average pKa is 4.2 with a range of 2.1–8.8.120 Foster summarized the various acid–base transitions for BSA in 1977 (Table 4.1).121 The base transition at pH 11.3 is associated with a major structural transition with exposures of six “buried” tyrosine residues.122,123 While albumin is resilient to acid and organic solvents, this protein can be denatured by heat or by the addition of chaotropic agents. Since albumin can be pasteurized in the inal manufacturing step to provide safety from infectious agents, there has been considerable interest in studying the effect of heat on albumin.124–127 The cited studies show the inactivation of viral pathogens.125,127–129 There is an occasional problem with aggregation.130 Roelands and coworkers131 showed that repeated heating at 56°C interspersed with storage at 4°C resulted in reversible dimerization, with gelation occurring with 25% albumin after eight steps of heating. Finlayson and coworkers132 subsequently demonstrated the reversal of dimerization of HSA by

TABLE 4.1 pH Transitions in Albumin Designation

Transition pH

E F (N → F) A N B (N → B)

2.7 3.6–3.9 5.5 Native (pH 7) 9–9.5

Source: Adapted from Foster, J.F., in Albumin, Structure, Function, and Uses, eds. V.M. Rosenoer, M. Oratz, and M. Rothschild, pp. 53–84, Oxford/Pergammon, New York, 1977.

90

Biotechnology of Plasma Proteins

heating for 10 h at 60°C in the presence of acetyl–tryptophan and sodium caprylate. Kolthoff and Tan133 observed that BSA heated (70°C) in the presence of a mercurial agent demonstrates reversible aggregation (the aggregated material is made 8 M in urea and diluted 10-fold); in the absence of a “blocking agent,” the aggregation is irreversible due to the disulide exchange. Renaturation in the initial presence of a chaotropic agent is similar to the process used for bacterial recombinant proteins expressed as exclusion bodies.134 More recently, Moriyama and coworkers135 have reported that the helicity of BSA decreased with increasing temperature; half of the helicity was lost at 80°C, while 16% of the helicity remained (66% initial) after heating at 130°C. The presence of SDS offered protection against the helicity lost on heating up to a temperature of 90°C. Nuclear magnetic resonance (NMR) studies of SDS demonstrated the maintenance of surfactant structure up to a temperature of 130°C. Lohner and coworkers136 have reported that HSA is less stable to thermal denaturation in the N form but is protected by benoxaprofen or warfarin. Pico137 has described the thermal denaturation of HSA as a classical two-step process that proceeds through an “unfolded state,” which is reversible, and then proceeds to a terminally denaturated state. Pico notes that the second, irreversible step is quite slow. Michnik138 used differential scanning calorimetry (DSC) to show that ethanol decreased the thermal stability of HSA. Chaotropic agents, such as urea or guanidine, have also been used to study the conformation of HSA. Wallevik139 observed the reversible denaturation of HSA with guanidine (1–2.5 M); the process at pH 5.6 or 9.2 is a multistage process, while at pH 2.6, the process is continuous. Aoki and coworkers140 studied the denaturation of BSA with urea; guanidine was used to demonstrate that the changes observed were not due to carbamylation. These researchers used acrylamide gel electrophoresis (native electrophoresis)141 to measure the conformational change. The incubation of BSA with 5.0 M urea at pH 9.0 resulted in the formation of a dimer and aggregates as well as an altered monomer fraction; if the BSA was modiied with p-chloromercuribenzoate, the altered monomer fraction was not observed. The effect of the sulfhydryl group modiication133 on conformation change induced by heating has been mentioned earlier. The denaturation of HSA modiied with acrylodan (6-acroyl-2dimethylamino naphthalene) was studied by González-Jiménez and Cortijo.142 These researchers observed a transition at approximately 6.0 M urea at pH 7.4 as measured with a luorescent probe; the modiication of albumin with acrylodan did not result in conformational change as measured by CD. These researchers also reported denaturation with guanidine hydrochloride. It is noted that acrylodan is considered speciic for the modiication of sulfhydryl groups in proteins.143–145 The modiication of the sulfhydryl group128 has been shown to inluence conformational change with heating. Fatty acids that bind to HSA stabilize HSA from denaturation with guanidine hydrochloride146 or urea.147 Mendez and coworkers148 observed that glycated HSA was slightly more stable than native HSA to denaturation with guanidine hydrochloride. The studies on the effect of heat and chaotropic agents on albumin are consistent with albumin as a lexible protein relatively resistant to denaturation. The results suggest that the modiication of cysteine 34 inluences protein stability. As the conformational lexibility of albumin is likely critical for binding, the next section will consider conformation studies that do not involve heating or chaotropic agents.

91

Albumin

Auranoin is a gold-containing drug (Figure 4.2) that was developed to treat rheumatoid arthritis but is of current interest for cancer therapeutics;149–152 it reacts with the cysteine residue in albumin.153 Work by Christodoulou and coworkers154 using 1H NMR showed that the reaction of auranoin with BSA is associated with a conformational change as measured by changes in the resonance of His3 (1H NMR). These latter researchers also cite the failure of cathepsin to degrade mercaptalbumin, while albumin modiied at Cys34 is degraded, suggesting a conformation change. A more recent work by Talib and coworkers155 showed that a prior modiication of Ac H O H O

Ac

Et

O

Au

S

O H

P

Et

O

Ac H

Et

H Ac Auranofin

+ Albumin*

HS

Et Et

Albumin

HS

Et P

Au

Albumin*

S

Et

Et

P

Au

Au

S

S

Albumin

Et

RSH

R

S

Au

S

R

Albumin*

S

Albumin

+ Et Et

P

O

Et

FIGURE 4.2 The structure of auranoin and its reactions with albumin. The reaction is reversible via a mixed disulide exchange mechanism as well as conversion of the triethylphosphine to the corresponding oxide by a slower reaction. It is suggested that a particular conformation of albumin designated by an asterisk is the reactive species.153,157,158

92

Biotechnology of Plasma Proteins

albumin with iodoacetamide blocked the reaction with AuPEt3Cl (Figure 4.2); the mixed disulide of cysteine with HSA was also less reactive. A consideration of the early work of Christodoulou and coworkers154 and the more recent work of Talib and coworkers155 suggests that while the reaction of gold with the thiolate anion is preferred, it is possible that the reaction with the cysteine-mixed disulide of HSA occurs with auranoin but at a much slower rate. The reduced rate of reaction with the mixed disulide and possible lack of reaction with an oxidized cysteine might provide an insight into the variable clinical response to gold-containing drugs. The interaction of auranoin with albumin is critical to the pharmacokinetics of the drug as 80% of the gold is bound to albumin after the administration of the drug. However, as with all things, the mechanism of albumin may be a bit more complex. First, there may be a difference between albumin in vivo and the isolated commercial product (human or bovine) with respect to the amount of mercaptalbumin. Christodoulou and coworkers in a later study156 used 1H NMR as a surrogate measure of the chemistry of Cys34 to show that more than 90% of the albumin in human blood plasma was mercaptalbumin. These researchers suggested that the modiication of Cys34 with auranoin resulted in a conformation transition that was consistent with their earlier observations.154 Subsequently, Roberts and coworkers157 presented a careful kinetic analysis of the reaction of auranoin and the isopropyl analog with BSA. These results show that the kinetics of the reaction of auranoin with BSA are complex. The dependence of the reaction rate on conformation provides an explanation for the lack of consistent second-order kinetics. The reaction is reversible via a disulide exchange mechanism. The reader is referred to Ahmad and coworkers158 for a current review of metal-based therapeutics. Ohkubo159 showed that HSA modiied with a mixed disulide with either cysteine or glutathione showed differences in solvent perturbation spectroscopy compared to native HSA. Triluoroethanol, a mimic of anesthetics, inluences protein conformation,160 according to considerable studies on albumin,161,162 including an increasing susceptibility to chemical modiication with acrolein.163 Anesthetics such as propofol and halothane have been reported to bind to HSA164 at sites that bind fatty acids; the binding of propofol or halothane is associated with only minor changes in the crystal structure. Earlier studies165 from another group of researchers have shown tight binding of these two drugs. Lund and coworkers166 used chromatography on phenyl-agarose to separate HSA into two fractions, one of which was bound to the column (R or retained) and the second fraction appeared in the column void volume (NR or not retained). The R fraction was eluted with 8 M ethylene glycol. Chromatography on octyl-agarose resulted in complete retention; if 1 M ethylene glycol was included, the elution pattern was similar to that observed on phenyl-agarose. The separation was reversible, supporting the concept of conformational heterogeneity. The binding was minimum at pH 6.0 with an increase in the binding up to a pH of 9.0, where the binding remained constant with increasing pH; the pH dependence is consistent with a functional group with a pKa of 7.3. These researchers had previously demonstrated heterogeneity on immunoelectrophoresis with phenyl-agarose that relected ligand (fatty acid, bilirubin) binding.167 These researchers did not mention the possible inluence of the modiication at Cys34 upon separation on hydrophobic afinity matrices. Proteins

Albumin

93

such as albumin undergo conformational changes upon binding to matrices such as phenyl-agarose.168 Huang and coworkers169 used cross-linking with diimidoesters (disuccinimidyl glutarate and disuccinimidyl suberate) to show a conformational change in HSA on the binding of fatty acids. Michnik and others170 used DSC to show that UVC (254 nm) irradiation caused a conformational change in HSA; aggregation also occurred under these conditions. There is a difference between defatted albumin and native albumin. Normally, HSA is not glycosylated, but there are some variants with N-glycosylation.171 Glycosylation in domain I extended the circulatory half-life, while glycosylation in either domain II or domain III had no signiicant effect.172 Myeloperoxidase modiies methionine, tryptophan, and tyrosine in HSA, but this reaction occurs after secretion.173 While the modiications of HSA, such as glycation, oxidation, and nitrosylation, which are discussed later, do result in modiied proteins, I do not consider such modiications to be posttranslational modiications such as γ-carboxylation, sulfation, and phosphorylation, which are enzyme-catalyzed reactions.174 Some modiications such as the reaction with 4-hydroxy-2-nonenal175 or homocysteine thiolactone176 are the result of the enzymatic modiication of a precursor to form an active species. As with other plasma proteins with signiicant circulatory half-lives, albumin is subject to nonenzymatic glycation177–181 with a marked increase in diabetes where the advanced glycation end product (AGE) is suggested to play an important role in the various pathologies.182–185 Glycated albumin has been proposed as a marker for diabetes similar to glycated hemoglobin.186 Glycation of HSA shows speciicity with respect to the site of modiication and modifying sugar. Reducing sugars that are in equilibrium between a closed-ring form (pyranose form) and an open-chain structure possessing an aldehyde function (carbonyl) can react with unprotonated amino groups in proteins (primarily ɛ-amino groups of lysine residues; Figure 4.3).187 The reactivity of a given sugar is dependent on the amount of sugar in the carbonyl form. Thus, galactose, which has 0.02% carbonyl, reacts with hemoglobin more rapidly than glucose, which has 0.002% carbonyl.187 The glycation of HSA by galactose has been studied by Frost and coworkers,188 who reported that the sites modiied have also been reported to be modiied by glucose with the exception of Lys414. While the modiication of Lys414 by glucose was not reported by Frolov and Hoffmann,189 the modiication of this residue was reported by Barnaby and coworkers.190 It should be emphasized that the modiication occurs quite slowly and the pattern observed can depend on whether the modiication is “trapped” by borohydride reduction. Frost and coworkers have discussed the potential lability of the modiication of Lys414. Most studies conirm the early observations of Garlick and Mazer191 that Lys525 is the principal site of modiication. Iberg and Flückiger192 identiied four principal sites of in vivo glycation with glucose in HSA—Lys199, Lys281, Lys439, and Lys525—with a third of the total modiication at Lys525. While there is a major site of glycation of HSA with glucose, glycation of HSA does yield a heterogeneous product as would be expected from the studies on the reaction of trinitrobenzenesulfonic acid (TNBS) with HSA as reported by Goldfarb,193 who was able to identify three groups on the basis of reactivity. Goldfarb also suggested that TNBS bound to HSA prior to the reaction (Figure 4.4). Andersson and coworkers194 studied the reaction of

94

Biotechnology of Plasma Proteins O

OH H

C

HC

Carbonyl form (0.002%)

H

Hemiacetal form

CHO H

H OH

OH

H H

HO H

OH

HO

OH

H

O

HO H

OH

OH

H

H

R

CH2OH

O H2N

H

H2 C

B O

CH2

HO

CH2

CH2

CH2

CH2

CH2

CH2

N

NH

NH

CH

CH

CH2

OH

HO

H

OH H

HO

Maillard product O

HO

H

H

OH

H

OH

H

OH

H

OH

H

OH

H

OH

CH2OH Schiff base

CH2OH Eneaminol

CH2OH Amadori product

FIGURE 4.3 Nonenzymatic glycation of proteins. A reducing sugar or analog, such as methylglyoxal, reacts with the ɛ-amino groups forming a Schiff base in a reversible reaction (the stabilization of the Schiff base by reducing agents, such as sodium cyanoborohydride, is not shown). The reaction of a reducing sugar, such as glucose (shown), is dependent on the amount of sugar in the carbonyl form.187 The Schiff base can also proceed through a series of steps to form an Amadori product.192,874 Boronic acidbased afinity chromatography can be used to separate glycated albumin from native protein.178,875–877

95

Albumin NO2

NH2

+ NO2

O2N SO3

N H

2,4,6-Trinitrobenzenesulfonic acid O

NO2

Lysine

NO2

O2N NH

N H O Albumin + TNBS

Albumin TNBS

Trinitrophenylated albumin

FIGURE 4.4 Reaction of trinitrobenzenesulfonic acid (TNBS) with proteins. TNBS reacts with amino groups in albumin and other proteins. Albumin was found to have three classes of amino groups based on reactivity with TNBS.193 This and later work identiied the most reactive lysyl residue and suggested the formation of a complex between TNBS and albumin prior to reaction.196

TNBS with BSA, observing that TNBS did bind preferentially to certain sites on the protein prior to the reaction; palmitate decreased the rate of reaction of TNBS with BSA presumably by competition for the same binding site. Kurono195 reported that the reaction of TNBS with HSA did follow Michaelis–Menten kinetics. Subsequent work by Kurono and coworkers196 showed that the fast-reacting site is Lys199 and suggested that the proximity of Trp214 was important for binding speciicity. Lys199 is a signiicant site for glycation as discussed earlier. Glycation of HSA does result in a conformational change,197–202 which may be related to the pathogenic effects described earlier. Glycation of recombinant HSA resulted in a decreased circulatory half-life in a rat and an increased hepatic uptake in a mouse model.199 As cited earlier, while AGEs are formed due to the reaction of glucose with HSA, which are suggested to be involved in the pathogenesis of diabetes,200 there is no evidence to

96

Biotechnology of Plasma Proteins O

ONOO− Peroxynitrite

O2 + NO

N

SH

NO

S

H 2C

Nitric oxide

H2C

+ CH

CH

SH

N H

N H

O

O

S-nitrosocysteine O

H2C CH N H O

N S O

O

O

CH2 H N N H

HO NH2

OH O

Glutathione

FIGURE 4.5 Nitric oxide and peroxynitrite and formation of S-nitrosyl cysteine in albumin. The single cysteine residue in albumin is subject to nitrosylation, and S-nitrosoalbumin is considered a biologically signiicant in vivo repository for nitric oxide.878 The in vivo S-nitrosylation can be mediated either by nitric oxide synthase879 or by reaction with peroxynitrite derived from nitric oxide and superoxide.880 The synthesis of S-nitrosoalbumin using S-nitrosoglutathione as a donor is also shown.252 Dahm and coworkers881 suggest preferential formation of protein thiols via transnitrosylation from S-nitrosothiols.

support the cross-linking of HSA upon the nonenzymatic reaction of glucose with HSA as observed with other proteins.192 Glycation is one of the several important in vivo modiications of albumin; other important modiications are oxidation and S-nitrosylation.202–204 Oxidation and nitrosylation are considered to be related physiological events.205–207 The in vivo oxidation of functional groups in proteins is mediated by reactive oxygen species, such as superoxide, hydrogen peroxide, and hydroxyl radical, and usually involves the oxidation of cysteine, cystine, and methionine residues.208 Nitration and nitrosylation reactions are mediated by reactive nitrogen species, including nitric oxide and peroxynitrite (Figure 4.5).209 Peroxynitrite is formed via an oxidative stress mechanism, while nitric oxide may be formed directly by nitric oxide synthase and used in situ or can be transferred to glutathione to form S-nitrosoglutathione. Nitric oxide and peroxynitrite (which can be formed by the reaction of reactive oxygen species) can work at cross purposes in homeostasis.210 The in vivo oxidation of proteins can result in a complex mixture of products. In the case of HSA, there is a single free sulfhydryl group that can be oxidized to form a mixed disulide with cysteine or glutathione or oxidized to sulfenic, sulinic, or sulfonic acids (Figure 4.6).211 The chemistry of the sulfhydryl group as derived from an

97

Albumin

SH N H

Reduction

CH

O

HC

Oxidation H2C

CH2

N H

S O

S

Cysteine

H2C CH

Reduction

Oxidation

H

HO

N H

O S

S

Cystine O H 2C

H 2C

CH

CH N H

N H

RSH O

O

Mixed disulfide

Cysteine sulfenic acid

HO

O S H2C CH N H O

Cysteine sulfinic acid OH O

S

O

H2 C CH N H O Cysteine sulfonic acid

FIGURE 4.6 Oxidation/reduction reactions of cystine and cysteine in proteins. Shown is the oxidation pathway from cysteine to cysteine sulfonic acid. Cysteine may also be oxidized to cystine, presumably through cysteine sulfenic acid. Mixed disulides may also occur via the sulfenic acid derivative.882–885

98

Biotechnology of Plasma Proteins

in vitro modiication is discussed later. In addition, oxidation can modify other groups such as lysine, resulting in carbonyl derivatives (Figure 4.7).212 Various techniques have been developed for the analysis of oxidized HSA.213 However, it is clear that oxidized HSA is heterogeneous, given the number of possible derivative forms. Temple and coworkers214 identiied the sites of carbonylation in HSA resulting from metalcatalyzed oxidation (Fe3+/ascorbic acid) or oxidation with hypochlorous acid. Metalcatalyzed oxidation modiied Lys97 and Lys186, while hypochlorous acid modiied Lys130, Lys257, Lys438, Lys499, and Lys598. The reader is referred to a work by Maisonneuve and coworkers215 for a discussion on the speciicity of carbonylation in proteins. A comparison of the lysyl residues in HSA modiied by various reagents is shown in Figure 4.7. Only a few of the residues modiied by glucose or galactose are modiied with 4-hydroxy-trans-2-nonenal (4-HNE) (Lys199 and Lys525), one residue is modiied with hypochlorous acid (Lys438/439), and there is no overlap with those residues modiied with Fe3+/ascorbic acid. Bar-Or and coworkers216 reported cysteinylation of Cys34 forming a disulide in albumin from single individuals and commercial albumin preparations. The extent of cysteine modiication was higher in commercial albumin preparations than in albumin from single donors. Nitrosylation of cysteine was also observed in both preparations as was the loss of the N-terminal Asp-Ala dipeptide glycation observed only in the commercial albumin preparations. The authors suggest that the administration of albumin preparations with a high amount of modiied cysteine could have negative consequences. Subsequent work by this group217 showed a correlation between the loss of the Asp-Ala dipeptide and the formation of the diketopiperazine derivative as well as a correlation between this combination and in vitro immunosuppressive activity. Further work suggested that the diketopiperazine derivative is responsible for the immunosuppressive activity.218 Lysine is also modiied by a reaction with aspirin (acetylsalicylic acid) in a variety of proteins,219–226 including albumin,227 where it has an effect on glycation.228 The acetyl function in aspirin is an “active” acetyl group similar to the acetyl group in p-nitrophenyl acetate, which is also known to acetylate the lysine residues in proteins.229 Lockridge and coworkers230 have reported the extensive modiication of HSA with p-nitrophenyl acetate, including the acetylation of Try411 and the modiication of numerous other sites; these researchers suggest that this extensive modiication is responsible for the observed catalytic activity, not the result of turnover at a catalytic site. This is discussed later in the section on enzyme activity. The acetylation of albumin by aspirin (Figure 4.8) dates back to the early observations of Hawkins and coworkers.227,231 Jacobsen232 modiied HSA with a variety of reagents, including acetylsalicylic acid (aspirin), acetic anhydride, and N-acetylimidazole, all of which would result in the acetylation of the protein. Aspirin modiied 1.4 amino groups (from a total of 57), while acetic anhydride modiied 56 amine residues as well as 10 phenolic hydroxyl groups; N-acetylimidazole modiied 22 amino groups and 4 hydroxyl groups. In a subsequent work in 1975, aspirin was demonstrated by Walker233 to modify Lys199 in HSA. Recently, Liyasova and coworkers234 showed the acetylation of Lys199, Lys402, Lys519, and Lys525 in HSA with 0.2 mM aspirin; higher concentrations of aspirin resulted in a more extensive modiication. Bohney and coworkers235 established Lys190 as the primary site of modiication with pyridoxal-5′-phosphate.

99

Albumin −O

H3C

O

O O NH HC NH

O

H N

H N

N H

N H (a)

H3C

(c)

H N

O N H

NH

(b)

HO

H3C N

O

O

CH3

HO NH NH

H N

CHO

H2N O (d)

NH2 N H

H N

O (e)

N H (f)

O

H N N H (g)

HN

O

O OH

HO

(i) (h)

HO

FIGURE 4.7 Structures of some chemically modiied lysine residues in proteins. (a) Nɛ-Acetyllysine providing charge neutralization.886 (b) Nɛ-Succinyllysine providing charge reversal.887 (c) 2-Aminoadipyl residue obtained from metal-catalyzed oxidation of proteins.214 (d) Nɛ-Methyllysine and (e) Nɛ-Dimethyllysine, which are obtained by reaction with formaldehyde followed by reduction with sodium cyanoborohydride providing charge preservation.888 (f) The Michael addition product derived from the reaction of a lysyl residue with 4-hydroxy-2-nonenal. (g) The Schiff base derived product (after reduction) derived from the reaction of a lysyl residue with 4-hydroxy-2-nonenal.889 (h) and (i) are products derived from the reaction of a lysyl residue in albumin with 16α-hydroxyestrone.890

100

Biotechnology of Plasma Proteins

NH O CH2 O

CH3

CH2 O

HO

O

CH2

H N

H N

Acetylsalicylic acid (aspirin) 2-(acetoxy)benzoic acid 2-acetoxylbenzoid acid

N H O

OH

CH3 O NH CH2

NH

CH2 O

O

CH3 Acetaminophen; paracetamol; N-(4-hydroxyphenyl)acetamide

CH2

H N

H N N H O

P450

Protein

OH

O

CH2 S

NH

N

O

O CH3

CH3

FIGURE 4.8 Reaction of aspirin and acetaminophen with albumin. Shown is the reaction of aspirin [2-(acetoxy)benzoic acid] with the ɛ-amino group of lysine to yield Nɛ-acetyllysine.234 Also shown is the conversion of acetaminophen to N-acetyl-p-benzoquinoneimine intermediate and one of the suggested reaction products with albumin. This speciic modiication is suggested to involve Cys34.365,891

Albumin

101

Both aspirin (acetylsalicylic acid) and the hydrolysis product, salicylic acid, bind to albumin, and the albumin–salicylate complex may be responsible for the hypersensitive allergic response observed with aspirin.236–241 Nitrosylation and nitration of HSA occur during periods of oxidative stress. Nitrosylation occurs at Cys34.242 Nitrosylation (Figure 4.5) is a reversible reaction and does affect ligand binding by HSA; ligands also affect the nitrosylation of HSA by nitric oxide.243 The concentration of nitrosylated albumin in blood has been determined to be approximately 200 nM,244 and S-nitrosoalbumin has a half-life of 5.5 h in heparinized blood in vitro.245 As the in vivo half-life of nitric oxide is quite short (ca. 0.1 s),246,247 HSA can serve as a reservoir for physiologically signiicant circulating nitric oxide.248 Ascorbic acid can cause the release of nitric oxide249 S-nitrosylated derivatives, with such an effect being responsible for the increased bioavailability of nitric oxide.250 It has been suggested that ascorbate (Figure 4.9) may have a role in the release of nitric oxide radical from nitrosylated albumin.251 S-Nitrosylated HSA has a cytoprotective effect and is being developed as a therapeutic product for reperfusion injury.252 Jourd’heuil and coworkers253 observed the nitrosylation of both Trp214 and Cys34, with the products having different redox sensitivity to reactions with reduced glutathione and/or ascorbate. Peroxynitrite is derived from the reaction of nitric oxide and superoxide anion 254 and reacts with tyrosine, tryptophan, and cysteine residues in proteins (Figure 4.10).255–257 A reaction with tyrosine yields 3-nitrotyrosine, which is the product obtained on reaction with tetranitromethane (TNM).258 Nitrated proteins, including HSA, can be detected in plasma samples using an ELISA technique.259 It is suggested that the formation of 3-nitrotyrosine in HSA by peroxynitrite is a biomarker for oxidative stress.260,261 Harrohalli and coworkers used site-directed mutagenesis to suggest that Trp214 is the principal site of nitrosylation in HSA.262 Sonnenschein and coworkers263 have reported the formation of S-nitrosothiols via the intermediate formation of N-nitrosotryptophan (Figure 4.5).

IN VITRO CHEMICAL MODIFICATION OF ALBUMIN HSA and BSA have been used as model compounds for the characterization of reagents developed for the modiication of proteins. Chemical modiication of these proteins has also been used to understand the in vivo modiications described earlier. Finally, the in vitro studies provide an insight into the function of HSA as a scavenger as described later. Albumin contains the usual amounts of potential nucleophiles, such as amino groups, carboxyl groups available for modiication, 17 disulide bonds, and a single thiol group on cysteine-34. The free sulfhydryl group (Cys34) in HSA poses an interesting question regarding its potential function. However, the author posits that the presence of ive free sulfhydryl groups in factor VIII is a much bigger puzzle (see Chapter 6). There is a free sulfhydryl group in ibroblast growth factor-1, which is considered to be vestigial.264 Mutagenesis of this cysteine residue increases the functional half-life of ibroblast growth factor-1.265 Zhao and coworkers266 observed that the mutation of cysteine to serine in the HSA moiety of human interferon-α2b-HSA fusion protein increased the stability, reduced the aggregation, and decreased the immunogenicity resulting from

102

Biotechnology of Plasma Proteins HO

OH

H2C

HO

C H

OH

H2 C O

C H

−2e−

O

+2e− HO

O OH

O Dehydroascorbic acid

Ascorbic acid Cupric

Cuprous

O HN

CH

C

O

NH HN

CH2

CH

S

CH2

N

SH

C

NH

+ NO

O NO2

NO2

HOOC

HOOC

+

S

SH

S

SH

COOH

COOH

NO2

NO2 .

TRP

TRP

FIGURE 4.9 Ascorbic and dehydroascorbic acid. Ascorbic acid can serve as a reducing agent for cupric ions, causing oxidative damage mediated via cuprous ion and superoxide.892,893 Ascorbate can also cause the release of nitric oxide from S-nitrosylated derivatives.894 It is also suggested that ascorbate can reduce disulide bonds as such in dithiobis (2-nitrobenzoic acid).895,896 Ascorbate can also reduce protein-free radicals.897

103

Albumin Peroxynitrite ONOO−

O−

OH NO2

HOONO Peroxynitrous acid

N H

N H O

Tetranitromethane C(NO2)4

O 3-Nitrotyrosine

NO2 HN Peroxynitrite

HN

ONOO−

CH2 H2N

CH

CH2 C

OH H2 N

CH

C

OH

O O 6-Nitrotryptophan

FIGURE 4.10 Reactions of peroxynitrite and tetranitromethane with amino acid residues in albumin. Tyrosine residues may be nitrated by either peroxynitrite or tetranitromethane in proteins.898 Peroxynitrite has been shown to modify a tryptophanyl residue in albumin.257

mechanical and thermal stress. There was a difference in the pharmacokinetic behavior. However, the sulfhydryl group of albumin does appear to have a distinct function(s), which is described later. I would add the comment that at one time, it was thought that extracellular proteins such as albumin or factor VIII had only disulide bonds and that proteins with free sulfhydryl groups were only found inside the cell.267–269 Cysteine 34 is present in various forms in HSA, including the various oxidation states and mixed disulide forms described earlier. In addition, dehydroalanine derived from Cys34 has also been reported (Figure 4.6).270 As a result of these various modiications, commercial albumin derived from plasma as well as recombinant albumin demonstrated heterogeneity of cysteine residues.271 The majority of the modiied Cys34 was present as a mixed disulide with cysteine. Plasma-derived albumin had 55% free Cys34, while the recombinant preparation had 83% free Cys34. Ghiggeri and coworkers272 found 0.49 mol of free sulfhydryl group per mole of HSA [5,5′-dithiobis-(2-nitrobenzoic acid), DTNB] (Figure 4.11) in proteins obtained from

104

Biotechnology of Plasma Proteins O +H N 3

OH SH

[O]

CH2 Albumin

CH

C

O−

+H

3N

CH

CH2

CH2

SH

S

S

S

CH2

CH2

NO2

C

O−

Albumin

Albumin

NO2

O

+ NH3

COO−

RSSR CH2 N CH2

R

S

S

S

S

S

S

S

S

CH2

CH2

CH2

CH2

Albumin (a)

Albumin (b)

Albumin (c)

CysSSR

Albumin

FIGURE 4.11 Mixed disulide formation in albumin. Albumin in vivo has 60%–70% of Cys34 as free thiol, with the remainder present as mixed disulide or oxidized cysteine.899 The albumin-mixed disulide can undergo disulide exchange with thiols in plasma, such as homocystine.900 Also shown are the products of reaction of the thiol in albumin with 5,5′-dithiobis(2-nitrobenzoic acid), 2,2′-bis-(5-nitropyridine).272,290,301

healthy subjects. Aćimovic and coworkers273 found 0.76 mol of sulfhydryl group per mole of HSA (commercial product) in their studies. Early workers also came to the conclusion that there were less than stoichiometric amounts of the sulfhydryl group in albumin.1,103 The amount of sulfhydryl group reactivity in albumin is reduced on storage in solution, with the decrease being most rapid at neutral pH. Hughes274 was able to isolate HSA with a stoichiometric amount of sulfhydryl by the crystallization of albumin from plasma as the dimer using mercuric chloride (Figure 4.12). The mercuric ion is removed by cysteine, yielding an albumin with a stoichiometric amount of sulfur; this albumin is referred to as mercaptalbumin.275 A novel approach to mercaptalbumin has been developed by Carlsson and Svenson.276 These researchers used covalent chromatography on S-thiopyridyl-derivatized agarose (activated thiol Sepharose; Sepharose-coupled glutathione modiied with 2,2′-dithiopyridyldisulide; Figure 4.11) to prepare bovine mercaptalbumin. Funk and coworkers277 used

105

Albumin O

O

H N

H N

N H

HC

N H

CH2

HC CH2 S

SH

Hg HgCl2

SH

S

H2C

H N

CH

H2C

H N

CH

N H

N H O

O

Methylmercury (CH3Hg+)

CH3

RSH

SH

Hg

H2C

H N

CH

S N H

H2C

H N

CH

O

N H O

FIGURE 4.12 Reaction of mercuric compounds and organomercurial derivatives. Mercuric ion (mercuric chloride) and organic mercurial compounds, such as methyl mercury, can react with Cys34 in albumin.901 As shown, while mercuric ion forms a tight bond between two sulfhydryl groups, this interaction has been used to purify mercaptalbumin.275 Methylmercury, an environmental toxin, also binds to the sulfhydryl group in albumin.901

S-thiopyridyl agarose to remove mercaptalbumin as a method for enriching albumin containing derivatized Cys34. The reactivity of the sulfhydryl group at Cys34 depends on prior modiication (refer to the various studies on the presence of oxidized products and mixed disulides earlier) and nucleophilicity; the nucleophilicity depends, in part, on the pKa

106

Biotechnology of Plasma Proteins

of the functional group;278 in this case, the thiolate anion is the reactive species. The pKa for an “average” sulfhydryl group in a protein is 8.3–8.7.279 The pKa for the sulfhydryl group in HSA is approximately 7.8–8.0.280–282 Lower values for the pKa have been suggested,283 but the value determined by Spiga and coworkers281 has been obtained by a reaction with DNTB and the value determined by Bruschi and coworkers280 has been obtained by electrophoretic titration. The thiol–disulide reaction between HSA and cystine had a pH optima of approximately 8.2.282 A pKa value of 5.0 for the sulfhydryl group in HSA was cited by Narazaki and coworkers283 referring to the pKa value obtained by Lewis and coworkers284 using potentiometric titration. A pKa value of 9.0 (e.g., 1 M NH4OH)

O R

HN

S

+

R′

OH HN

N H

O

FIGURE 4.13 Reaction of cyanide with disulide bond in albumin. Cyanide reacts with disulide bonds in a disproportionation reaction, yielding a thiocyanate and sulfhydryl group.326 Cleavage at the modiied cysteine residue can be accomplished by providing a biomarker for cyanide exposure.322

coworkers273 examined the role of HSA in binding methylglyoxal in their work, emphasizing the importance of the hydrophobic environment around Cys34 in its scavenging activity. Tryptophan-214 is considered to play a role in providing this hydrophobic environment.334–336 Another example of scavenging or detoxiication is the study on the reaction of acrylamide with the sulfhydryl group albumin.337 Glycation has been described earlier, and other reactive aldehydes, including nonenal derivatives and methyl glyoxal, react with various functional groups (Figure 4.14), including sulfhydryl, amino, imidazole, and guanidino.338–341 TNM is a classic reagent for the nitration of tyrosine residues in proteins.342–344 It is of personal interest to the author who worked with this reagent some 30 years ago and was informed by a reviewer on a grant application to the National Institutes of Health that nitrotyrosine would never be of signiicance. The reaction of TNM with proteins (Figure 4.10) has been reviewed; modiication occurs primarily at tyrosine residues, but nitration of tryptophan does occur as well as oxidation of cysteine and

110

Biotechnology of Plasma Proteins H C

CH2

O

C H

H2 C

H C

1,3-Butanediene

H3 C

H C H Crotonaldehyde

O CH2

CH C H

H2C

O H3C HN OH

S CH3

CH3

O

O NH

P450

N

S

O OH HN Aminoacetophen [N-(4-hydroxyphenyl)acetamide] H3C

O

FIGURE 4.14 Reaction of electrophilic derivatives with albumin.365,902

methionine.345 TNM reaction for the modiication of HSA has been used as a model system for the nitration reaction, while other studies focus on the use of TNM for the identiication of functional sites on HSA protein. Malan and Edelhoch258 were able to modify half of the tyrosine residues in HSA with TNM at pH 8.0 and studied the spectral characteristics of the modiied protein. These researchers reported differences in the spectral characteristics allowing separation of the modiied tyrosyl residues into three groups. Fehske and coworkers346 also reported the modiication of half the tyrosine residues in HSA with TNM but reported that the modiication of two tyrosine residues decreased the binding afinity for tryptophan or benzodiazepine; there was one rapidly reacting tyrosyl residue. Subsequent work

Albumin

111

from this group.347 identiied the highly reactive tyrosine residues as Tyr411. Kim and coworkers348 used afinity capillary electrophoresis to show modiication of Trp214 and Tyr411 with 2-hydroxy-5-nitrobenzyl bromide and TNM, respectively. Modiication with TNM affected the indole–benzodiazepine binding site with no effect on warfarin binding; modiication of tryptophanyl residues with 2-hydroxy-5nitrobenzyl bromide did affect warfarin binding. Šantrůček and coworkers349 modiied HSA with TNM or iodine and observed that Ty411 readily iodinated but reacted poorly with TNM; Tyr161 and Tyr263 also showed differences in reactivity between iodine and TNM. Tyrosine 411 is also modiied by organophosphorous agents as are other tyrosine residues in HSA.350 Some of these reactions of Cys34 need to be discussed in more detail from the perspective of site-speciic chemical modiication. In addition, there are other modiication reactions, which should be considered before moving into a consideration of oxidation. The reaction of a sulfhydryl group and other functional groups in a protein can be a complex process that is dependent on the presence of other functional groups.292 Barron351 classiied sulfhydryl groups in proteins as being freely reacting, sluggish, or masked. Webb352 suggested that there is a continuum from highly reactive to unreactive. In the case of albumin, the cysteine may be unavailable due to prior modiication. The results of modiication at an extreme pH would suggest that there are steric factors that inluence modiication of Cys34 at neutral pH. However, with product characterization, the issue of disulide cleavage/disulide exchange in albumin293,294,353 has the potential to complicate the interpretation of the results. Assuming that disulide exchange or cleavage is not an issue, it could be argued that there is an increased sulfhydryl exposure at low pH and elevated pH. The problem of prior modiication by mixed disulide formation or oxidation has been discussed earlier. That leaves the issue of the effect of the local environment on the modiication of Cys34. Stewart and colleagues354 suggest the importance of Tyr84, His39, and Asp38 in the enhanced reactivity of Cys34. Several other researchers have discussed the importance of His39 in the reactivity of Cys34. One result of the microenvironment for Cys34 at neutral pH is a low pKa of 5–8.2 (see above). For the purposes of the current discussion, it is assumed that results obtained with BSA, while quantitatively different from HSA, may be used to interpret experimental results obtained with HSA. Cys34 is not available, either from the formation of a mixed disulide or oxidation, in approximately 50% of albumin in vivo, which may have consequences for the antioxidant activity of HSA. Anraku and coworkers355 observed that the oxidation of Cys34 in hemodialysis patients correlated with the thiol content and suggested that some 40% of the antioxidant activity of HSA occurs in vivo. This value was obtained from a consideration of the antioxidant activity of C34S HSA. Wilson and coworkers356 cited work showing that iodoacetamide reacts with the thiol in BSA while there is no reaction with iodoacetic acid. This difference in reactivity has been observed by other researchers and usually indicates the presence on anionic sites such as glutamic acid or aspartic acid, close to the cysteine residue being modiied.345 Wilson and colleagues studied the reaction of DTNB and other disulides with BSA. They reported a rate of 70/m/s for the reaction of DTNB with BSA at pH 8.5 compared to a calculated rate of 2.5 × 105/m/s for DTNB with

112

Biotechnology of Plasma Proteins

an unhindered thiol under the same conditions. A neutral, hydrophobic disulide, such as diethyl disulide, demonstrated a higher rate of reaction (45/m/s) than the calculated rate with unhindered thiol (0.7/m/s) under the same conditions (pH 8.0). Cystamine disulide showed a higher rate of reaction with BSA (1 × 103/m/s) than with the unhindered disulide (3.66 × 102/m/s). While not directly relevant to the speciic albumin problem, Gitler and colleagues357 have demonstrated that cationic detergents enhance the reactivity of sulfhydryl groups in proteins by lowering the pKa (presumably increasing nucleophilicity). Takeda and coworkers299 showed an increase in the rate of reaction of DTNB with BSA in the presence of urea with a suggestion of saturation kinetics in the reaction, which would indicate the formation of a reagent–BSA complex prior to the modiication of Cys34. Zhang and coworkers301 studied the reaction of DTNB as well as a zwitterionic derivative [5-(2-aminoethyl)dithio-2-nitrobenzoate, ADNB] (Figure 4.11) with BSA, canine serum albumin, and equine serum albumin. The objective was to assess the importance of Glu82 in the reactivity of Cys34 (it should be noted that Cys34 is a conserved residue in all serum albumins). The rate of reaction of the neutral reagent, ADNB, was 190 times more rapid that that observed with DTNB, which carries a negative charge. ADNB was sixfold more reactive than DTNB with canine albumin where Glu82 is replaced by an aspartic acid residue. The two reagents, ADNB and DTNB, were equally reactive with equine albumin where Glu82 is replaced by an alanine residue. This group also observed an increase in the rate of modiication with DTNB with increased ionic strength; the effect of ionic strength was also observed by Wilson and coworkers.356 In a study that adds to the results obtained by Zhang and colleagues,301 Stewart and coworkers358 studied the effect of site-speciic mutagenesis of HSA on reaction with DTNB. The rate of reaction of DTNB with the Tyr84Phe mutant was some 170-fold more rapid than the rate with the wild-type protein (pH 9.0) and fourfold more rapid with His39Leu (pH 9.0); the rate of reaction of the His39Leu was less than the wildtype at pH < 7.5. The decrease in rate below pH 7.5 would be consistent with the suggested role of His39 in decreasing the pKa of Cys34. These researchers also suggest that the hydroxyl group of Tyr84 forms a hydrogen bond with the thiolate anion, which would decrease reactivity. In more recent work, Spiga and coworkers359 have studied the reaction of DTNB with the various serum albumins to gain more insight into structural features inluencing Cys34 reactivity. HSA and BSA showed similar reactivity with DTNB at pH 7.4 (~75/m/s) at pH 7.4, while the rate reaction was faster with rat (7.4 × 102/m/s) or porcine (2.6 × 103/m/s) albumin. This group suggests the presence of additional anionic sites in the vicinity of Cys34. The reader is directed to several earlier effective studies that provide insight into the inluence of surrounding functional groups on sulfhydryl reactivity. Chaiken and Smith360,361 and Gerwin362 showed major differences in the reactivity of chloroacetic acid and chloroacetamide with a cysteine residue at active sites of papain and streptococcal proteinase, respectively. The reaction of the chloroacetamide, a neutral reagent, showed a sigmoidal curve when rate was plotted versus pH with either papain or streptococcal proteinase consistent with the critical importance of the thiolate anion in the reaction. The reaction of chloroacetic acid with either papain or streptococcal proteinase shows a bell-shaped curve when reaction rate is plotted versus pH. Chaiken and Smith postulated that the enhanced reactivity of papaion at pH 5–6 is due to the electrostatic

Albumin

113

interaction of chloroacetate with the imidazole ring of histidine and/or the enhanced ionization of the cysteine residue by the imidazolium ions. Thus, there is ample support from other systems for the role of His38 in enhancing the reaction of Cys34 in albumin. As stated earlier, there is a report of the modiication of Cys34 in HSA with iodoacetamide but not with iodoacetate. Cysteine 34 in HSA is a point of attachment of a variety of electrophilic agents,175 such as chemical warfare agents and other chemical toxins including α,β-unsaturated aldehydes (Figure 4.14).363 Another example is provided by the reaction of metabolites of 1,3-butanedione (Figure 4.14) with albumin where reaction occurs at multiple sites.364 Another example is provided from the studies of Damsten and coworkers365 who showed that N-acetyl-pbenzoquinoneimine, which is derived from the action of P450 on acetaminophen (Figure 4.8), reacts with Cys34 in HSA. Modiication of lysine residues in albumin has been somewhat less studied than sulfhydryl reactivity. The studies of Goldfarb with TNBS193 have been cited earlier and have provided a basis for classifying side chain amino groups on the basis of reactivity. Succinylation of albumin has been the subject of considerable study over the past several years. Part of this work has been directed at the preparation of succinylated HSA as a therapeutic agent against HIV.366–370 Scavenger receptors were originally described as receptors for oxidized and acetylated low-density lipoproteins, and succinylation can be used to target soluble antigens to cytotoxic T lymphocytes.371 There are a variety of studies where succinylation has been used to target albumin to scavenger receptors in various tissues.372–374 Lysine residues in albumin are available for succinylation and such modiication does cause signiicant change in conformation375,376 (succinylation causes charge reversal), while guanidation (creates more basic residue) has a lesser effect.377 Table 4.2 reviews lysine modiication in albumin.

ALBUMIN AS A CARRIER The circulating half-life of HSA is about 20 days.44–46,378 Bennhold and Kallee379 reported a half-life of 16 days for 131I-labeled albumin in a normal subject and 50–70 days in a subject with analbuminemia. As an aside, analbuminemia (congenital absence of albumin) is a comparatively rare condition with apparently minor clinical consequences.380–382 The half-life of albumin does decrease in critically ill patients as does cholinesterase.383 Anraku and coworkers384 found that substantial oxidation of HSA with chloramine T (50 mM chloramine T; 15 μM HSA, pH 8.0, 1 h) resulted in a protein with a decrease in circulatory half-life in mice (8.7 vs. 20 min) and increased uptake (146 vs. 13 μL/h) in liver. Modiication at lower concentrations of chloramine T (10 μM) did not result in a signiicant change in either circulatory half-life or liver uptake. This group385 also showed that metal ion–catalyzed oxidation of Arg410 reduces half-life; reduced half-life was also observed for the R410A mutant. Earlier studies by Anhorn and others386 observed the pharmacokinetics of human albumin in rabbits. Human plasma albumin was shown to have a half-life in rabbits of approximately 6 days (143 h); modiication with 2-phenyl-4-ethoxy methylene oxazolone386 decreased the half-life to 3.3 days (79.9 h). 2-Phenyl-4-ethoxy methyl oxazolone [(E)-4-(ethoxymethylene)-2-phenyloxazol-5(4H)-one; oxazolone]

114

Biotechnology of Plasma Proteins

TABLE 4.2 Modification of Lysine Residues in Human Serum Albumin

Lysine 12 93 97 130 159 186 195 199 212 233 257 276 281 414 438/439e 499 525 598 a

b c d e

Fe3+/ Ascorbic Acid

HOCL

4-HNEa

MDAb

Glycation (Glucose)c

Glycation (Galactose)d

+ +

+

+ + + + + +

+ + +

+

+

+ + +

+ + + +

+ +

+

+

+

4-Hydroxy-2-nonenal.338 Lysine modiication by 4-HNE can occur via Schiff base formation, as with glycation (Lys195) or both Schiff base and Michael addition (Lys199 and Lys525). Malondialdehyde.872 A number of studies on glycation with glucose are cited in the text. From Frost et al.187 There is some confusion in the literature about the location of this residue in the amino acid sequence of HSA. The sequence obtained from albumin mRNA by Dugaiczyk and coworkers77 has the lysine residue at position 439.

was one of the earliest described haptens387,388 (described as pro-antigenic in its ability to combine with a protein to produce a new antigen387). Oxazolone is a potent skin sensitizer in forming stable products with lysine and sulfhydryl groups in proteins (Figure 4.15). The half-life of albumin has encouraged the development of fusion proteins with biologically active peptides. The fusion protein between albumin and interferon-β has a half-life of 36–40 h compared to 8 h for interferon-β and retained useful biological activity.389 A fusion protein between albumin and interleukin-2 (IL-2) demonstrated longer circulatory half-life (6–8 h vs. 19–57 min for recombinant IL-2) with retention of therapeutic eficacy.390 Other useful albumin fusion products have been obtained with recombinant antibody fragments,391 hirudin,392 butyrylcholinesterase,393 blood coagulation factor IX,394 and tumor necrosis factor (TNF).395 This approach396 is

115

Albumin

O

O

N

N

O

O NH2

O Oxazolone

C H2

CH2

NH

CH2

CH2

CH2

CH2

SH CH2

CH2 CH2

C H2

S

N O

O O

C H2

S O O

Additional products with protein nucleophiles

HN

Hippuryl derivative

FIGURE 4.15 The structure of oxazolone and related compounds and their reaction with albumin. The reaction of oxazolone [(E)-4-(ethoxymethylene)-2-phenyloxazol-5(4H)] with lysine and sulfhydryl residues in proteins is shown.387,903

based on the recycling of albumin by the neonatal Fc receptor (FcRn).397–399 The study by Andersen and coworkers399 cites the failure of an albumin variant to interact with the FcRn receptor, providing an explanation for the analbuminemic state where the catabolic rate is increased due to lack of albumin recycling. IgG is also recycled by the same mechanism.400–402 McCurdy and coworkers403 prepared rabbit serum albumin with an N-terminal hexahistine tag and rabbit serum albumin with the

116

Biotechnology of Plasma Proteins

N-terminal hexahistidine tag where Cys34 is replaced by an alanine (C34A). Both of these proteins had the same half-life (~5 days). A dimer of the C34A protein joined by a hexahistidine spacer had a half-life of 3.0 days; this was not expected as, in principle, a larger protein should demonstrate slower clearance. While a certain part of the prolongation of half-life is due to the use of FcRn recycling, a large portion is due to decreased renal clearance as a result of increased size as is observed with PEGylation.404,405 These fusion proteins are examples of the use of genetic engineering to attach a biopharmaceutical protein to an albumin carrier. Techniques of solution chemistry may also be used to attach “drugs” to albumin to improve pharmacokinetics.406,407 The example of nitrosylation of albumin to prepare a therapeutic agent for the delivery of nitric oxide408 has been mentioned earlier.252 Ishima and coworkers409 evaluated the use of an albumin variant (albumin Liprizzi), R410C, as a nitric oxide carrier; a half-life of 20.4 min was observed in a mouse model. In subsequent work,410 this group engineered sites of additional carbohydrate attachment in HSA and prepared nitrosylated albumin-targeted nitric oxide delivery to the liver. Antiviral nucleoside analogs (luorodeoxyuridine) have been coupled to lactosaminated human albumin via the imidazolide derivatives (Figure 4.16).411–413 99mTc-Labeled lactosaminated albumin has been used for imaging the hepatic asiaoglycoprotein receptor in liver,414 proving superior to the previously developed 99mTc-galactosyl-albumin415 (lactose is 4-O-β-d-galactopyranosyl-d-glucose). The 99mTc is bound to the lactosaminated albumin after reduction of lactosaminated albumin (~70 mm 2-mercaptoethanol at approximately pH 8.3, 37°C, 1 h). Analysis of the reduced albumin showed 21 sulfhydryl groups (Ellman’s reagent). Lee and Hirose416 have shown that reduced albumin retains considerable structure. Kim and coworkers417 coupled glucagon-like peptide to albumin with retention of biological activity and improved pharmacokinetics. Warnecke and coworkers418 have used maleimide chemistry to couple carboplatin drugs to Cys34 of HSA. Several groups have coupled insulin to HSA, also with maleimide chemistry.419,420 Schechter and coworkers419 developed the insulin– albumin conjugate as a prodrug as did Warnecke and coworkers for the carboplatin– albumin conjugate (Figure 4.17).418 The general use of albumin as a carrier for drugs and drug products has been reviewed by Kratz421 and by Sasson and coworkers.422 Sasson and coworkers422 were able to obtain prolongation of half-life by improving noncovalent binding of prodrugs to albumin. Drugs that contain a sulfhydryl function can bind to albumin as demonstrated by the reaction of bucillamine derivatives and HSA,423 providing for the reversible association of a sulfhydryl-containing drug and HSA (Figure 4.18).424,425 Captopril is a sulfhydryl-containing drug (Figure 4.18), which can react with albumin to form a mixed disulide.426 The captopril–HSA complex may be involved in adverse reactions.427,428 The effect of Cys34 modiication on albumin conformation has been mentioned earlier.133,354 As an example, Glowacki and Jakubowski429 observed the oxidation of cysteine-enhanced homocysteinylation at lysine residues. Thus, it is not surprising that the modiication of albumin with captopril results in conformation change.430,431 A novel approach takes advantage of the propensity of Cys34 to form mixed disulide bonds. Wu and coworkers432 engineered a cysteine on the C-terminus of glucagon-like peptide-1 and formed a mixed disulide with HSA. The key to using Cys34 as a point of chemical attachment depends on

117

Albumin O F NH

N

O N

O

P

O

O

N H

O−

H

H

H OH

O F NH

N

O

O H2 C

Protein

H N

P

O

O H

O−

H

H

H OH

Glucose

Galactose H

H HO

H OH

H

H

OH

HO

Point of attachment to protein O

O O

H

HO

H

H OH OH

H

OH OH

Lactose

FIGURE 4.16 The coupling of luorodexoyuridine to lactoaminated albumin using imidazolide chemistry. Shown is the structure of the imidazolide derivative of luorodeoxyuridine.411,413

the availability of the thiolate anion. Commercial albumin, either plasma-derived or recombinant, is modiied at Cys34 either by oxidation or mixed disulide formation. As such, it is necessary to convert the modiied cysteine to free cysteine in order to maximize product formation. Kratz and coworkers407,421 used dithiothreitol to reduce the Cys34 residue in human albumin, while Jeong and coworkers414 used 2-mercaptoethanol to reduce more than half of the disulide bonds. A variety of approaches,

118

Biotechnology of Plasma Proteins O

N

CH2

H2 C

O

CH3

N

SH

+

H N

O

N H

O N-ethylmaleimide

CH3

S O H N

O

N H

O

O

O O O

O

Carboplatin

O O

NH2

Pt

H2N

O O

O

O

N O O

NH2

O

Pt O

O NH2

FIGURE 4.17 Coupling of carboplatin and carboplatin prodrug to albumin using maleimide chemistry. Shown at the top is the reaction of N-ethylmaleimide to cysteine. Below are labile maleimide derivatives of carboplatin developed for coupling to Cys34 of albumin.418,421

including afinity chromatography on a thiol-agarose matrix, were mentioned earlier for the production of mercaptoalbumin. Albumin, both human and bovine, as well as ovalbumin, is used as a carrier for small molecules, including peptides, for the development of antibodies. The preactivation of albumin with divinylsulfone has seen some use433,434 as has the iodoacetyl ester of N-hydroxysuccinimide433,435 (Figure 4.19). The latter work by Houen and coworkers435 performed the modiication with the carrier protein (ovalbumin) adsorbed to an ion exchange matrix. Other conjugate chemistries have been used with BSA and ovalbumin for the preparation of immunogens.436–439 One example is the glutaraldehyde coupling of Nɛ-carboxymethyllysine to albumin as a hapten for the generation of antibodies against AGEs.440 Thierse and coworkers441 were able to use HSA as a carrier for metal ions as haptens. There are several studies on the technology of using albumin for hapten production, and

119

Albumin Cysteine O

CH3 O H3C

C

H N

C

H C H2C

SH pKa = 8.39

C

OH

SH pKa = 10.22

Bucillamine[N-(2-mercapto-2-methylpropanoyl--cysteine] CH3 O H3 C

C

O H N

C

H C

C

OH CH3

H2 C

SH pKa = 10.50

S

N-(3,3-dimethyl-1-mercaptobutyryl)--methyl--cysteine HO O O C H2 C

C HC

HS

H C

CH CH2

CH3 H2C

CH2 Captopril 1-[(2S)-3-mercapto-2-methyl-1oxypropyl]--proline HO O H2 C HS H2 C

C HC

H C

CH3 H2 C S

S

R

O

C CH

HO CH2

O H2 C

CH2 S

C HC

S Cys34 in albumin

H C

CH3 H2 C

H2C

O

C CH

CH2 CH2

+ RSH

FIGURE 4.18 Reversible association of sulfhydryl-containing drugs with Cys34 in human serum albumin. Shown is the example of a structure of bucillamine and related compounds.423 Either sulfhydryl could react with Cys34 to form a covalent complex. Also shown is a bucillamine derivative, N-(3,3-dimethyl-1-mercaptobutyryl)-S-methyl-l-cysteine, which was used in a subsequent study to show binding differences between normal and pathological serum.904 The structure of captopril, an antihypertensive drug with a sulfhydryl group, is also shown as is the reaction of captopril with Cys34 in albumin.

they are worth consideration.442–445 Jenkinson and coworkers446 used studies with p-phenylenediamine–albumin adducts to show that such adducts are determinants in patients with contact dermatitis. It has been known for some time that bead-like structures for the delivery of drugs can be manufactured from albumin.447–449 Albumin has been used with

120

Biotechnology of Plasma Proteins O

OH O +

H2 C

C H

OH

S

C H

CH2

O Divinylsulfone

O H2 C O

H2 C

S

C H

CH2

R

O

NH RNH2

CH2

OH R

CH2

RSH S

O

CH2

S

O

CH2

CH2

H2C O

O

S

O OH

CH2 H2C O OH

FIGURE 4.19 Use of divinylsulfone or haloalkyl for coupling to albumin. Divinylsulfone and N-hydroxysuccinimide ester of iodoacetic acid. These can be used to activate albumin prior to the coupling of a smaller molecule.433

poly(lactide-co-glycolide) for encapsulation of tetanus vaccine.450 These researchers used spray-drying for the preparation of microcapsules. Heparin-coated albumin has been used as a matrix to bind the basic ibroblast growth factor,451 anti-mucus antibody has been coupled to albumin microspheres via carbodiimide chemistry in developing a drug delivery system with delayed gastrointestinal transit,452 and the RGD (arginine-glycine-aspartic acid) peptide has been coupled to albumin microsphere for targeting tumor cells.453 Albumin microspheres have been used to deliver radionucleotides.454,455 Albumin has been used to coat calcium alginate microspheres to improve

Albumin

121

stability.456,457 Albumin-coated magnetic beads have been used to measure drug– albumin interactions458 and, after attachment of antibody, to measure biomarkers.459

LIGAND-BINDING BY ALBUMIN Albumin is well known for binding metal ions, other cations, anions, including fatty acids, and various drugs. The structure of albumin has been discussed in several earlier sections. Briely, albumin has two major binding sites (sites I and II) and other sites such as Cys34 and metal ion binding sites.424,460 Site I binds large heterocyclic compounds and dicarboxylic acids, and examples include warfarin, bilirubin, and salicylates,424 while site II binds smaller molecules such as indoles and diazepam. The designation of site I and site II on albumin stems from the early work of Sudlow and coworkers,461,462 and the sites are occasionally referred to as Sudlow Site I and Sudlow Site II.463–465 The binding sites for metal ions are separate from the Sudlow sites.466 Albumin also binds fatty acids at several sites, which may overlap with Sudlow sites.467–471 Crystallographic analysis of binding of myristic acid (C14; tetradecanoic acid) to HSA showed six binding sites.471 As noted by Curry and coworkers,472 0.1–2 mol of fatty acid are bound per mole of albumin under normal physiological conditions and there are likely three high afinity sites for the binding of fatty acids. Other researchers have suggested seven binding sites,473 while other studies suggest the binding of eight myristic acids.474 The binding of fatty acids to albumin is associated with conformational change in albumin475–484 and can change the characteristics of the site of interaction on the albumin molecule such as the binding of warfarin.485,486 The binding of fatty acids inluences the polarity of the microenvironment around Cys34480 inluencing the redox properties of albumin.487 Site I is located in subdomain IIA and contains the single tryptophanyl residue (Trp214) in albumin,424 and it has been reported that the modiication of Trp214 with 2-hydroxy5-nitrobenzyl bromide inluences the binding of warfarin.348 Bromocresol purple is used to measure albumin and binds to the warfarin-binding site (site I) and a site II region as well.488 Sudlow Site I is frequently referred to as the warfarin-binding site,489–495 with the binding to albumin being of importance for the pharmacokinetics of this anticoagulant drug.496–498 There are clinical situations where the administration of a drug can cause an increase in the effective concentration of warfarin, most likely by competition for the same binding site on albumin such as observed for zafirkukast.498 In another situation, there was a decrease in the effective concentration of warfarin with a high-protein diet.499 Site II is frequently referred to as the indole site or diazepam site346,500–508 to distinguish it from site I, the warfarin site. The binding of molecules to site II is stereospeciic, and immobilized albumin can be used for chiral separations.509–517 While site I is referred to as the warfarin site, it is not a homogeneous site but contains regions of differing speciicity.490 As noted, modiication of the sole tryptophan (Trp214) in HSA with 2-hydroxy-5-nitrobenzyl bromide inluences warfarin binding with little effect of azopropazone; however, azopropazone can displace warfarin from HSA as effectively as azopropazone can displace itself (14C-labeled drug). Diazepam that binds to site II has little effect on the binding of either warfarin or azopropazone to HSA. Subsequently, Yamasaki and coworkers518 proposed

122

Biotechnology of Plasma Proteins

the existence of three binding regions within site I: site Ia, which was characterized by the binding of the marker ligand acenocoumarol; site Ib, which was characterized by the binding of the marker ligand dansyl-asparagine (DNSA); and site Ic, which was characterized by the binding of the marker ligand phenylbutazone. These sites overlap but are separate from site II (diazepam site) and a third site referred to as binding area III (digitoxin). The digitoxin site, referred to as site 2 by Sjöholm and coworkers,519 was determined by the study of binding to HSA immobilized by emulsion polymerization in polyacrylamide microparticles.520 Subsequent work has shown some peculiarities in digitoxin binding by albumin,521,522 but studies by Hage and Sengupta523 using high-performance liquid chromatography (HPLC) with an immobilized HSA matrix to measure binding show clearly that digitoxin binds to albumin, independent of site I or site II. More recently, Yamasaki and coworkers524 extended their earlier studies on site I subregions showing interaction between site Ib (binding of dansyl-L-asparagine, DNSA) and site Ic (n-alkyl p-aminobenzoates). pH has an effect with higher binding of DNSA at alkaline pH. This group had previously reported a change in the spatial relationship between site I and site II caused by the N → B transition.525 The importance of understanding the N → B transition for drug binding by albumin has been discussed earlier. Albumin also binds heme526,527 and such binding has encouraged the development of heme–albumin as an oxygen carrier in artiicial blood development.528–530 Heme binding to HSA also inluences the binding of other ligands.531–533 Ascenzi and coworkers have advanced the concept of allostery in a monomeric protein base on the binding of heme to HSA binding.534 In this model, albumin is envisioned as a protein with three discrete domains with independent binding characteristics, but able to interact, inluencing the binding characteristics of another domain much in the same way as the binding of an effector to one subunit in an oligomeric protein. It can be concluded that albumin is responsible for the bound portion of drugs in the circulation and serves as a reservoir for the release of these drugs over time. Other compounds, including medication, can compete for the binding of drugs to albumin and inluence pharmacokinetics. It is also clear that fatty acid binding can inluence the binding of drugs to albumin. Albumin is also important for binding metal ions, with the binding of zinc of particular importance. I commend the reader to an excellent article535 by Fehske and coworkers, published in 1981, where they concluded that “Thus, the structure and the location of drug binding sites of HSA might be more complicated than it seemed a few years ago ….” It would seem like the same comment holds true some 30 years later, and it is clear that albumin is a lexible molecule capable of complex interactions in the circulation. The various sites can be probed by various techniques. Modiication of HSA with 2-hydroxy-5-nitrobenzyl bromide affects warfarin binding, while TNM modiication affects indole/benzodiazapine binding.347,348 Phillips and Marmorstein536 studied the binding of 6-nitrotryptophan to HSA; binding was associated with a blue shift and hyperchromicity. These changes are similar to those observed with transfer into apolar solvent and were used to determine a K D of 8 × 10−5 M and stoichiometric binding (n = 0.95). There are a number of studies on the binding of tryptophan by HSA, suggesting 104 –105 M binding constants for tryptophan and HSA and that such binding is inluenced by the state of the albumin protein.537–541 Pressure denaturation

Albumin

123

of HSA eliminated binding at site II (based on dansylsarcosine binding) at pressures above 300 MPa, while warfarin binding was retained.542 This study also observed the effect of urea denaturation, and dansylsarcosine binding was lost in 6 M urea, while retaining warfarin binding. Data cited earlier show that modiication of the single tryptophanyl residue (Trp214) inluences binding in site I, while modiication of Tyr411 inluences binding in site II. A variety of the site probes used are also luorescent, such as DNSA, and cause changes in the intrinsic luorescence of the protein, or exhibit other spectral changes. An example of the latter is provided by the studies of Yamasaki and coworkers524 on the subregions of site I, where changes in the CD spectra and luorescence anisotropy of DNSA bound to site Ic were inluenced by the binding of n-butyl-p-aminobenzoate (and related n-alkyl-p-aminobenzoates).

PURIFICATION AND CHARACTERIZATION OF HSA FOR CLINICAL USE The Cohn fractionation process described in Chapter 2 was driven by the clinical requirements of albumin. The Cohn Fraction V contains albumin and is usually the last biotherapeutic obtained in the Cohn fractionation. Plasma protein fraction (PPF) is similar to albumin with lower protein concentration (~5%) and is obtained from the Cohn Fraction IV-4.543,544 PPF, however, is not the same as 5% albumin; albumin is supplied as a 5% solution and 25% solution. Albumin is provided as a liquid formulation, which is subjected to pasteurization in the inal manufacturing step.545–553 The pasteurization process has been demonstrated to inactivate various pathogens, such as hepatitis virus545 and, more recently, H1N1 virus.550,553 Sodium caprylate and N-acetyl-tryptophan were included as stabilizers for albumin.554,555 Pinya Cohen has described the development of stabilizers for use in the pasteurization process for albumin.556 The albumin derived from plasma fractionation (the Cohn Fraction V) was obtained as a powder in the original process and supplied to the U.S. Navy (by contract) as a 25% solution to minimize the space required for transport.557 This contract also called for stability at 50°C, as the product would be shipped and used in tropical climates.558 The basic work on thermal stability of human albumin was led by George Scatchard, more famous to most from the Scatchard plot as discussed earlier. In the irst study,559 Scatchard and others showed that the 25% solution of HSA was stable at pH 6.8 in 0.3 M NaCl for 16 h at 50°C and that storage in the cold was not an obligate requirement. The salt concentration was required for stability. In subsequent studies,560 Scatchard and coworkers built on studies561 by Ballou and coworkers who demonstrated stabilization of albumin by alkyl acids and nonpolar anions, such as N-acetyltryptophan, and developed a pasteurization process for albumin, obviating the earlier requirement for the addition of merthiolate for bacteriostasis. Boyer and Luck continued their studies at Stanford for a period after World War II on interaction of fatty acids and other organics,562–566 which led to studies such as that by Yu and Finlayson554 cited earlier providing strong scientiic support for the manufacture. Other researchers, including John Edsall, contributed to this understanding.567–569 Paul D. Boyer worked on the albumin project at Stanford University with J. Murray Luck. Boyer went onto UCLA for an illustrious career on enzyme chemistry, including winning the Nobel Prize in Chemistry in 1997; he was

124

Biotechnology of Plasma Proteins

also a founder of the Molecular Biology Institute at UCLA. Luck stayed in Palo Alto and retired in 1965, having served on Stanford faculty since 1926. He is best known for starting the nonproit enterprise “Annual Reviews.” The point here, as with other individuals who worked on blood during World War II, is that these representatives of the “greatest generation” made outstanding contributions to the early biotechnology of blood and then moved onto incredibly distinguished careers in academia, all of which begs the question as to whether albumin is really a dull protein. Protein aggregation has been observed with albumin and is suggested to be due to proteins other than albumin.548,552 The inluence of proteins other than albumin on the stability of therapeutic preparations was, in fact, demonstrated in early studies on albumin by Scatchard and coworkers560 and more recently by Lin and coworkers.552 Lin and coworkers suggest that contamination of albumin with haptoglobin was responsible for the observed aggregation in albumin preparation. Lin and coworkers suggest that haptoglobin, being the less stable protein, aggregates and entraps albumin, which is present at a much higher concentration. Girard and coworkers570 used a variety of techniques to demonstrate heterogeneity in HSA used as an excipient for recombinant human growth hormone. Aggregation can be measured by gel permeation chromatography.571 There has been some recent work on the process of plasma albumin production. Johnston and coworkers572 used caprylate at a low pH to provide an additional viral inactivation step in the manufacture of albumin. This was based on the ability of unionized caprylate to disrupt the lipid membrane of enveloped viruses. More recent work has used proteomic technologies, such as 2-DIGE, for the characterization of albumin.573 There has been considerable activity directed toward the development of recombinant HSA in the past two decades.574–582 These studies have shown that recombinant albumin is safe and effective. On the positive side, the absence of glycosylation and other posttranslational modiications (see above) permits production in a variety of cells, including prokaryotic systems, which should allow for more eficient manufacture in large quantities. On the negative side, the drivers (pathogen issues) that resulted in the production of products, such as recombinant factor VIII (see Chapter 6), are not signiicant for plasma albumin because of the pasteurization step. In work cited earlier, there may be greater availability of Cys34, but it is not clear whether this quality is signiicant, as mercaptalbumin can be easily obtained from albumin isolated from blood plasma. Approved products include Delta’s Recombumin, which was originally marketed by Sanoi-Pasteur but currently a product of Novozymes. Recombinant albumin has also been developed in Japan.582,583 Recombinant HSA has been used as a biomaterial584 and has served as a cell culture additive.585 There has also been radiobiopharmaceutical application with labeling with 99mTc for lung perfusion imaging.586,587 In an approach that did not use recombinant HSA but was suitable for use with the recombinant protein, Hoppmann and coworkers588 did couple an HER2 Afibody analog to HSA (previously modiied with metal ion chelator [1,4,7,10-tetraazacyclododecane1,4,7,10-tetraacetic acid mono-N-hydroxysuccinimide]) using a bifunctional crosslinking agent. Preclinical studies suggest that the recombinant albumin is as effective as plasma albumin.589 While it is dificult to evaluate its potential use as a replacement for plasma HSA, there may well be a market for specialized products such as S-nitroso-albumin.252

Albumin

125

The manufacture of plasma-derived HSA is based on the alcohol fractionation scheme developed by Cohn (Chapter 2), which provides a precipitate fraction that is then dissolved and subjected to pasteurization. Recombinant HSA is puriied from yeast culture media by chromatographic techniques.574,590 Curling and colleagues591 developed an ion exchange method for the puriication of albumin from human plasma based on earlier work by Björling.592 There have been subsequent studies on the use of chromatography to obtain albumin of higher purity and/or to remove potential pathogens.593–596 One commercial product, Albumex, is produced by CSL Bioproducts using chromatography.597,598 Chromatography on histidyl-Sepharose 4B was used to separate glycated albumin from native albumin599 as well as chromatography on phenyl boronate columns.600 A phenyl boronate matrix is also used to capture glycated albumin for mass spectrometry.601 Kremer602 developed a method for isolated smaller quantities of HSA from plasma, which has been used for ligandbinding studies.603 As a inal note, albumin is sometimes referred to as a commodity when compared to other therapeutics, as albumin is in “competition” with other colloids and crystalloids for luid therapy. On the other hand, while not directly relevant to HSA, bovine albumin is considered too complex to be a commodity.604

BIOLOGICAL ACTIVITY OF ALBUMIN The aforementioned early work on albumin focused on its role in oncotic pressure (osmotic pressure, colloid osmotic pressure, which is one of the four Starling forces that affect net iltration from the capillaries).605,606 Thus, the clinical application of albumin as described earlier was driven, in part, by studies showing the effect of albumin on osmotic pressure. The drug-binding activity is described earlier as is the transport of fatty acids, redox activity, and a role in detoxiication, while the putative catalytic activity of albumin is described later. There is some reason to think that the transport function may be more critical than the oncotic function in certain clinical situations.607 There are some other observations on the biological activity, which, while not extensively described, should be mentioned. Wilkinson and McKay608 showed that native HSA does not elicit a chemotactic response in neutrophils, while acidiication (pH 2–4), reduction/carboxymethylation, or foam denaturation of HSA produced material that was chemotactic for neutrophils. More recently, Körmöczi and coworkers609 produced a modiied protein that binds to and stimulates neutrophils via oxidation of HSA by neutrophil-derived reactive oxygen intermediates (superoxide anion, hydrogen peroxide, and hydroxyl radical). Polzer and colleagues610 observed that HSA oxidized with hypochlorous acid binds to the GP120 proteins on HIV-1 variants blocking interaction with CD4. More recent work by Mera and cowokers611 showed that albumin from hemodialysis patients possessed decreased antioxidant activity and stimulated neutrophil oxidative burst. These researchers found an altered conformation for the oxidized HSA with increased exposure of hydrophobic amino acids. Jaisson and coworkers612 found that carbamylated HSA was an inhibitor of neutrophil oxidative burst. Lang and colleagues613 reported that albumin had anti-inlammatory effects that were not dependent on thiol content. In vitro studies with bovine aortic endothelial cells showed that HSA (70%– 85% oxidized with ~25% resistant to reduction with 2-mercaptoethanol or sodium

126

Biotechnology of Plasma Proteins

borohydride) inhibited binding of neutrophil-derived myeloperoxidase and blocked the hypochlorous acid-mediated release of adenine. The binding of neutrophils to the endothelial cells was enhanced by hydroxyethyl starch. It should be noted that Powers and coworkers614 had earlier reported that 25% HSA modulated adhesive interaction between neutrophils and endothelium in a shock model. Collison and coworkers615 showed that glycated albumin (AGEs) bound to neutrophils with mixed consequences (inhibited transendothelial migration and Staphylococcus aureusinduced but not fMLP-induced production of reactive oxygen species). Binding of glycated albumin to neutrophils was blocked by antibodies to either receptor for advanced glycation end products (RAGE) or Nɛ-carboxymethylsine, an AGE in albumin.616 However, it is not clear that the ability of albumin to modulate inlammation is translated into clinical effectiveness.617 In work cited earlier, Wilkinson and McKay608 suggested that the exposure of hydrophobic regions was important in HSA stimulation of neutrophils. In subsequent work,618,619 Wilkinson and McKay observed that the modiication of HSA with hydrophobic derivatives increased chemotactic activity. Wilkinson later reviewed the importance of hydrophobic interactions in the nonspeciic stimulation of phagocytic cells.620

ENZYMATIC ACTIVITY OF ALBUMIN The enzymatic activity of albumin has been debated over the last 50 years or more since the paper by Tove in 1962621 describing the hydrolysis of β-naphthyl esters by BSA. The late Dr. Samuel Tove was a distinguished and lively member of the faculty at North Carolina State University for more than 35 years. Tove was also a careful researcher who could take advantage of laboratory observations. In the course of his studies on lipid metabolism, Tove observed the hydrolysis of β-naphthyl palmitate. The activity was lost on thermal denaturation (autoclave, 15 min, 120°C), and chromatography on DEAE-cellulose did not separate the hydrolytic activity from albumin protein. As Tove noted, there had been an observation on the relationship of plasma esterase activity and activity ascribed to albumin. Casida and Augustinsson622 had studied the reaction of 1-naphthyl-N-methylcarbamate (Sevin insecticide) with various animal plasmas and human plasma fractions. The reaction was followed by the release of 1-naphthol. Hydrolytic activity was observed with various human animal plasmas as well as with human plasma fraction V. These researchers also used other ester substrates, including p-nitrophenyl acetate. They concluded that albumin possessed intrinsic esterase activity. Chapuis and coworkers623 stated that the effect of serum albumin on the hydrolysis of p-nitrophenyl acetate was irst observed in 1951, but the citation to the source is not available. The work by Chapuis and colleagues623 suggested that the esterase activity of HSA preparations might be due to contamination with cholinesterase. The contaminant issue has been consistent over the years. The relationship of plasma esterase activity with enzymatic activity of albumin dates back, at least, to the work by Kunkel and Ward624 at the Rockefeller Institute for Medical Research in 1947. Patients with infectious hepatitis or cirrhosis showed defective function of plasma esterase (acetylcholine) that parallels changes in albumin concentration. Although administration of

Albumin

127

HSA increased plasma esterase activity in nephrotic patients, there was no effect on plasma esterase activity in cirrhotic patients until there was an improvement in liver function. These results were interpreted as albumin supplying a component important for liver function. One rather interesting part of this study was the use of diisopropyl phosphoroluoridate (intramuscular administration) to inhibit plasma esterase in cirrhotic, nephrotic, and normal individuals. This particular observation adds to the chemical lore of early albumin research. Before heat treatment was developed for albumin,121 merthiolate (thimersol and sodium ethylmercurithiosalicylate)625,626 was used in early albumin preparations.627 I also found mention of the use of azides to preserve biologicals (albumin) as a resource at the Library of Congress628 but could not determine the exact citation. As far as I could tell, there was a lack of adverse reactions to these materials. Much of the work on the enzymatic activity of albumin used p-nitrophenyl esters such as p-nitrophenyl acetate. p-Nitrophenyl acetate is an extremely sensitive substrate for a variety of enzymes with vastly different speciicity. While it is generally accepted that either BSA or HSA accelerates the rate of hydrolysis of p-nitrophenyl acetate, the question of its mechanism remains unanswered. There are studies on the reaction of p-nitrophenyl acetate with proteins, which can be useful. A paper from Brian Hartley’s laboratory at Cambridge is instructive for the evaluation of the work on the esterase activity of albumin. Hartley was a very bright protein chemist, who laid much of the foundation for modern scientists. Hartley and Kilby629 studied the reaction of p-nitrophenyl acetate with chymotrypsin, insulin, and some low-molecular-weight compounds. While chymotrypsin is a protease with speciicity for peptide bonds where the carboxyl group is contributed by an aromatic amino acid, such as tyrosine or phenylalanine, this enzyme as well as other proteases, such as thrombin, can catalyze the hydrolysis of p-nitrophenyl acetate.630,631 As demonstrated in these studies, the major site of hydrolysis occurs at the enzyme-active site and, depending on solvent conditions and the acyl group, a stable O-acyl derivative can be obtained. Hartley and Kilby629 observed that the reaction of chymotrypsin with diisopropyl phosphoroluoridate, which modiies the active-site serine with concomitant inactivation of the enzyme-catalyzed reaction with peptide, anilide, and many esterase substrates, results in a modiied protein, which still promotes the hydrolysis of p-nitrophenyl acetate. The native enzyme exhibited a “burst” of p-nitrophenol production, which is not observed with the inactivated chymotrypsin. Hartley and Kilby629 further observed that simple tyrosine-like compounds could stimulate the hydrolysis of p-nitrophenyl acetate as could insulin. It is suggested that the observed hydrolysis of p-nitrophenyl acetate by inactivated chymotrypsin, tyrosine derivatives, and insulin involves the participation of amino and phenolic groups. Park and coworkers at Vanderbilt showed that p-nitrophenyl acetate was a substrate for 3-phosphoglyceraldehyde dehydrogenase.632 This activity required a free sulfhydryl group but, unlike the dehydrogenase activity, it did not require the coenzyme NAD (DPN). Another observation that deserves consideration is provided by De Caro and coworkers,633 who reported that a peptide chain fragment (336–449) derived from porcine pancreatic lipase by chymotrypsin could catalyze the hydrolysis of p-nitrophenyl acetate. Small peptides containing histidine and cysteine (Z-His-Ala-Gly-Gly-Cys-NH2) or histidine, cysteine, and aspartic acid

128

Biotechnology of Plasma Proteins

(Z-His-Ala-Asp-Gly-Cys-NH2) have been shown to demonstrate saturation kinetics in the hydrolysis of p-nitrophenyl acetate.634 These various observations suggest that there are a variety of mechanisms for a protein to accelerate the hydrolysis of p-nitrophenyl acetate. The issue of a contaminant being responsible for the observed activity of albumin was discussed by Tove in his 1962 paper,621 and Chapuis and coworkers623 ascribed the observed activity in albumin to contamination with cholinesterase. Cholinesterase activity is present in plasma635 and is found in the Cohn Fraction IV.636 There is current interest in butyrylcholinesterase,637–639 which is found in the Cohn Fraction IV-4.640 The relationship between the observed total cholinesterase activity and the butyrylcholinesterase activity in plasma is not clearly understood (Chapter 9). There is a suggestion of association of butyrylcholinesterase with albumin.641 According to this author, while cholinesterase activity may well be a contaminant of albumin preparations, the esterase activity observed by the various researchers is likely intrinsic to the albumin protein. The hydrolysis of p-nitrophenyl acetate by BSA (bovine mercaptalbumin; 0.86 mol sulfhydryl per mole of protein) was reported by Tildon and Ogilivie in 1972.642 They observed a “burst” reaction followed by a steady-state hydrolysis of p-nitrophenyl acetate. Analysis of BSA after the reaction showed the presence of O-acetyl groups (hydroxamic acid), and spectral (UV) analysis suggested the formation of O-acetyltyrosine. The data are consistent with the modiication of two tyrosine residues. Comparison of the rate of reaction of p-nitrophenyl acetate with tyrosine showed a 500-fold increase with BSA. The “burst” reaction was abolished when the BSA was treated with 8.0 M urea. Means and Bender643 reported that one site in HSA was acetylated quite rapidly by p-nitrophenyl acetate; the reaction was somewhat more rapid at alkaline pH after the removal of bound fatty acids. These researchers also observed inhibition of the reaction of p-nitrophenyl acetate with HSA by long-chain fatty acids (e.g., octanoate, myristate, and palmitate). Kurono and colleagues have also made an important series of contributions to our understanding of the reaction of p-nitrophenyl esters and other esters with HSA.644–650 A number of researchers also suggest that there is one strongly reactive site on HSA, which is located near Try411, that is involved in catalytic activity and also reacts with diisopropyl phosphoroluoridate and related organophosphorous compounds.651–660 p-Nitrophenyl acetate tends to a nonspeciic, highly reactive substrate for hydrolysis and, since albumin has multiple functional groups and the ability to bind organic compounds with some speciicity, it is not surprising that albumin can catalyze the hydrolysis of p-nitrophenyl acetate. It is more surprising that albumin does have enzymatic activity toward more complex low-molecular-weight compounds, implying enhanced speciicity. Rigg and Baird661 observed alkaline phosphatase activity in commercial preparations of HSA. These researchers, as others,633,641 did suggest that the observed activity might not be intrinsic to the albumin but an indication of an enzyme associated with the particular preparation of albumin. Elsbach and Pettis662 reported phospholipase A2 activity associated with albumin preparations; phospholipase activity in albumin was also reported by Singleton and Killian.663 Cha and Kim664 reported peroxidase activity in albumin, which was enhanced by either reduced glutathione or dithiothreitol; the activity was inhibited by iodoacetate. These

Albumin

129

researchers subsequently demonstrated the presence of thioredoxin-dependent lipid peroxidase activity in albumin.665 Yang and coworkers666 have reported the conversion of prostaglandin 15-keto-PGE2 to 15-keto-PGA2 and 15-keto PGB2 by albumin in reactions involving several basic amino acid residues. Sogorb and coworkers667 reported the hydrolysis of carbaryl (1-napthalene methylcarbamate; methyl carbamic acid 1-naphthyl ester) by rabbit albumin. The speciic activity of rabbit albumin (4.6 nmol/h/mg protein) is higher than that for human albumin (3.2 nmol/h/mg protein). A kcat of 7.1 × 10−5/s and a K M of 240 μM were obtained at pH 7.4 (37°C). The activity was strongly inhibited by fatty acids (caprylic acid) and fatty acid derivatives (p-nitrophenyl butyrate). This group later showed that albumin was a physiologically signiicant catalyst in the detoxiication of carbaryl668 and paraoxon.669 Another group670 has made the remarkable inding that BSA retains robust esterase activity (p-nitrophenyl palmitate) at 160°C. One group has reported the hydrolysis of RNA oligonucleotides.671–673 Jones and coworkers674 demonstrated intrinsic proteolytic activity in native and recombinant HSA preparations under reducing conditions. The proteolytic cleavage was speciic to cleavage at Arg18-Ala199 in synaptosomal-associated protein 25 (SNAP25).

CLINICAL USE OF ALBUMIN The clinical use of albumin can be separated into three different areas, two of which are related to similar biological properties, while the third is based on the availability of albumin in bulk. The irst is the original use of albumin as a plasma expander,36,675,676 which continues to the present day with perhaps more scrutiny.677–679 There is evidence for an increased use of albumin for clinical situations other than blood loss, but it is clear that there is continuing controversy over the use of albumin as a parenteral drug.680–683 The author is not a clinician and does not feel competent enough to comment on the ongoing use of albumin as a parenteral drug. There are several recent reviews, articles, and chapters on the clinical use of albumin.684–686 The second clinical application used albumin in an extracorporeal shunt in liver failure.687,688 This clinical approach is frequently referred to as “bridge to transplant.” 689 Liver failure presents a plethora of clinical problems, some of which are related to the failure of the liver to synthesize albumin, which can bind toxic materials. Riordan and colleagues690 separated extracorporeal liver support into two broad and somewhat overlapping categories. The term artiicial is used to describe a system that does not contain a biological component, while the term bioartiicial would describe a system that used hepatocytes in a bioreactor or liver tissue. Bañares and Catalina691 have summarized the various extracorporeal systems in an excellent review. The technical issue or clinical problem is the development of toxic materials in the circulation resulting from the failure of the liver and kidney to remove these toxic materials. Liver failure can also result in the development of hepatorenal disease,692 which adds an additional complication to the problem of in vivo blood puriication. It is possible to remove some of the toxic materials by dialysis, but substances such as bilirubin that are bound tightly to albumin are not readily removed by common renal dialysis methods,693 although there are some new, promising technologies

130

Biotechnology of Plasma Proteins

such as plasma diailtration.694 Plotz and coworkers693 did develop albumin coupled to agarose as a method for the afinity removal of free and bound bilirubin from blood. In 1997, Kaplan and Epstein695 reviewed the various approaches to the management of this clinical situation, which are discussed later. While this article has an approximate date, consideration of this work and that cited earlier provided a background to the clinical and technical issues. Early attempts to address this problem used extracorporeal circulation using cadaveric liver696 or porcine liver.697 A more sophisticated approach has used isolated porcine hepatocytes698–701 and more recently, stem cells differentiating into hepatocytes.702 The use of porcine hepatocytes, while demonstrating considerable success, did have histocompatibility issues.701 Miki and coworkers702 showed that the use of 3D bioreactor technology was useful in promoting the differentiation of human stem cells into primary adult hepatocytes. HSA has been used in extracorporeal support devices, as this protein binds many toxins (vida infra), is relatively inexpensive, and is accepted by the regulatory community. The use of albumin was irst demonstrated for extracorporeal detoxiication by Etteldorf and coworkers in 1961703 and by Grollman and Odell in 1962704 using peritoneal dialysis. Falkenhagen and colleages developed plasma sorption as a combination of plasma iltration and plasma perfusion over an adsorbent as an approach to hepatic failure.705,706 Hemoperfusion is a technique that uses extracorporeal circulation through an adsorbent, such as ion exchange resin and charcoal.707 HSA has been used to coat the adsorbent material to improve biocompatibility.708,709 The current technical approach for albumin dialysis, which is described by Kaplan and Epstein695 as ingenious, is the molecular adsorbent recirculating system (MARS) described by Stange and coworkers in 1993.710 The system is based on a high lux dialysis system with an albumin circulating on the dialysate side. The retentate side of the dialysis membrane is impregnated with albumin. The principle is based on the selectivity of albumin on the dialysate side to remove both free and albumin-bound toxins on the retentate side. The albumin on the dialysate side circulates in a loop passing through a combination of ion exchange and charcoal columns to regenerate the albumin.711,712 The third area of application is the use of albumin as a tissue solder. Casein from milk and blood proteins have been used as adhesives713 in bonding wood and other materials.714 It is noted that bovine albumin is used with glutaraldehyde as a surgical adhesive and suture support,715 which is marketed as Bioglue.716,717 Bioglue has seen considerable clinical use in a variety of applications.718–724 The use of the bovine material has not been without complication, including one foreign body reaction.724 Tissue soldering or laser soldering is a wound closure technology that uses an exogenous macromolecule, usually a protein, as patch material or solder in sealing a wound. The use of protein solders dates back to 1988 with studies by Poppas and coworkers725 on urethral surgery. Subsequent work from another group established the value of including a dye to increase the sensitivity of the system.726,727 The classic deinition of a solder is a metallic alloy used for uniting metal surfaces or parts, while a patch is deined as a piece of material attached to something to repair a hole or a tear so as to strengthen or protect a weak area.728 Tissue soldering uses a protein that is “melted” to repair a lesion or strengthen a weak area. Tissue soldering has the advantage over laser

Albumin

131

welding in providing greater bond strength, lesser collateral tissue damage, and a wider parameter window for providing a satisfactory bond or seal.729 The tissue solder must establish an adhesive bond with tissue and cohesive bonds within the solder. Establishment of an appropriate balance between adhesion and cohesion is essential to the formation of a strong bond between opposing tissue surfaces. The primary requisite for a solder is the ability to form strong adhesive/cohesive bonds in response to laser irradiation or other thermal challenge. The observed physical effect of laser irradiation/heat is to denature or melt the protein. Protein denaturation can be deined as a physical, intramolecular change in the native protein structure.730 Denaturation is not a chemical change but a conformation change in the protein and is not associated with the cleavage of protein bonds. However, the use of the term, even in 1954, has become so broad as to obfuscate its value.731 Denaturation is not an irreversible process and, in fact, is used in the processing of recombinant proteins expressed as inclusion bodies in bacterial systems.732–735 Aggregation, the subsequent loss of solubility, and loss of biological activity are the most commonly observed manifestations of protein denaturation. Aggregation results from the exposure of aromatic amino acids/hydrophobic regions,736–739 which then interact with other proteins via hydrophobic interactions.740–744 Aggregation is the property of protein denaturation that is important in laser soldering. The key in laser soldering is the precise application of energy to denature the protein to produce a useful aggregate with suficient adhesive and cohesive properties.745 An issue, therefore, in the development of this technology is the measurement of “melting of the solder” with respect to the application of energy. Measurement of the physical changes such as aggregation or loss of activity is likely occurring too late in the “melting” process to be of value. The abundance of albumin as a material, combined with the extensive clinical experience as a parenteral drug, made it a reasonable choice for a solder material to be used in tissue welding. As noted elsewhere, blood has been used as glue in the wood industry with some success, and it is likely that the albumin content was responsible for the adhesive property.746 Albumin is also noted for its ability to interact with various dyes, and the binding of bromocresol green and bromocresol purple is used for the clinical assay of albumin.747 This is likely an advantage when dyes are included to improve energy transfer from lasers. The ability of albumin (or any protein) to serve as a tissue solder is based on the ability of the protein to “melt” on denaturation and subsequently congeal (aggregate). Differing from, for example, metals, proteins can aggregate without cooling. As such, you need a protein that will “melt” at a temperature that will not cause collateral tissue damage during the process of coaptation.748 The “melting” of albumin can be measured by DSC. The Tm for defatted (fatty acid–free) human albumin is approximately 65°C, and there are species differences in albumin thermal stability, with bovine albumin considered less stable (lower Tm).749,750 The binding of fatty acids to albumin increases thermal stability (higher Tm).751,752 The goal of tissue soldering or tissue welding is to establish a strong bond with satisfactory coaptation with acceptable collateral tissue damage. Reduction of thermal damage is a major part of this goal. Thus, the use of fatty acid–free albumin is highly desirable. Second, the use of a homogeneous product will produce a “tighter”

132

Biotechnology of Plasma Proteins

Tm, which will allow more uniform heating/denaturation of the solder. Finally, the inclusion of a suitable dye in the solder permits lower energy input.753 Indocyanine green is the best example754 with an adsorption maximum of 805 nm and is used with diode laser (808 nm). Application of albumin solders to human clinical surgery has been limited to urology studies,755 which seem to have been successful. A particularly intriguing application has been the fabrication and use of stents using congealed albumin.756–758 In these studies, the 25% commercial human albumin is concentrated by ultrailtration to 50% and then further concentrated to yield a gel that can be fabricated into a structure. Another group has also developed an albumin stent referred to as Bioweld.759 Genipin is a cross-linking agent that has increased bond strength of albumin solder welds.760 Development of albumin solders continues.761

ALBUMIN AS DIAGNOSTIC/BIOMARKER HSA is modiied in vivo by a variety of environmental agents and drug metabolites. One example is the modiication of tyrosine and serine residues by organophosphorous compounds such as pesticides.350 HSA is also modiied by electrophilic agents derived from the metabolism of compounds, such as 1,3-butanediene364 and acetoaminophen.365 Modiication of HSA with HNE is suggested as a biomarker for systemic oxidative stress.338 Albumin is modiied by α,β-unsaturated aldehydes, such as crotonaldehyde derived from cigarette smoke.363 Albumin modiied by cyanide has been described earlier as a biomarker for cyanide exposure.322,323 The concentration of albumin in blood has been suggested as a biomarker for nutritional status but may be more relective of a disease state.762,763 Ischemia-modiied albumin is an albumin-derived biomarker of increasing interest. Ischemia-modiied albumin is deined as a defect in the binding of cobalt.764 Ischemia-modiied albumin is produced during myocardial ischemia and is considered to be a biomarker for that clinical condition.765 Following the development of ischemia-modiied albumin as a biochemical event and as a biomarker based on cobalt binding has been challenging. As far as the author could ascertain, a 1993 patent766 preceded the publication’s substance on ischemia-modiied albumin. Studies on the interaction of cobalt with albumin date back to studies with BSA by Rao and Lal767,768 in the 1950s. These researchers showed that modiication of lysine residues in BSA with methyl alcohol/HCl eliminated cobalt binding, while acetylation with acetic anhydride did not appear to affect binding. Nandedkar and colleagues769 showed that cobalt (Co2+) added to either human or rabbit plasma in vitro preferentially bound to albumin. These researchers subsequently demonstrated preferential binding in vivo using a rabbit model system.770 Lakusta and Sarkar771 studied the binding of Co2+ to tripeptide analogs to the amino terminus of HSA. Cobalt binding was studied as a model of zinc binding. These researchers reported that while Co2+ is a useful analog of Zn2+ binding to proteins, it is less useful with small peptides. The data did suggest that a histidyl residue and a carboxyl group were involved in the binding of zinc and cobalt to albumin. Trueba and coworkers772 observed a conformational change in BSA, with cobalt nitrate associated with binding to carboxyl groups. Later,773 Sadler and coworkers suggested the

Albumin

133

involvement of a lysine residue (Lys4) in addition to the N-terminal Asp-Ala-His sequence in binding Co2+. The formation of ischemia-modiied albumin has been suggested to involve modiication of the amino terminal sequence of HSA with reactive oxygen species,774 resulting in decreased binding of Co2+. Roy and coworkers775 showed that the hydroxyl radical (generated by the Fenton reaction) could modify albumin in human serum to form ischemia-modiied albumin. Ischemia-modiied albumin was not formed in serum with either hydrogen peroxide or superoxide. These researchers do not comment on the stability of ischemia-modiied albumin. It is clear that there is a narrow window (4–5 h) for the detection of ischemia-modiied albumin.775–778 It is not clear as to whether this suggests reversibility of modiication or rapid removal of the modiied albumin. Notwithstanding these issues, ischemia-modiied albumin has generated interest in the diagnosis of myocardial ischemia.778–784 However, there is evidence to suggest that the expression of ischemia-modiied albumin is somewhat nonspeciic, relecting other ischemic conditions and oxidative stress.785–791 A group of researchers from Genoa have identiied an oxidized form of albumin where Cys34 is converted to a sulfonic acid derivative in patients with focal glomerulosclerosis.792–795

PHARMACOKINETICS OF ALBUMIN A half-life of 15–20 days is commonly accepted for HSA.44–46,379,796–799 As with other proteins, chemical modiication can decrease half-life. Studies have shown that the oxidation of HSA by chloramine-T800 has a major effect on albumin, reducing binding of ligands, such as warfarin; there is also a shorter half-life for the oxidized proteins. Earlier studies by Berson and coworkers44 showed that “heavily iodinated” albumin was cleared somewhat more rapidly (t1/2, 10 days) than albumin with a lower degree of modiication (t1/2, 17 days). Gamsjäger and coworkers801 reported a half-life of 11.8 days for albumin in a retrospective study of more than 1400 intensive care unit patients. As discussed earlier, a substantial amount of albumin is outside the vascular space. Ding and coworkers802 observed a half-life of approximately 13 days (307 h) for albumin modiied with 5-aminoluorescein via triazine chemistry. Albumin dimer has been suggested to have lower vascular permeability than native HSA.803 An earlier study with an albumin dimer suggested a more rapid clearance.804 Albumin is also subject to renal clearance805 as well as to tubular reabsorption.806,807 Thus, the renal handling of albumin is a complex process808 that may relect albumin conformation.809 Clavant and Comper809 observed that a batch of tritiated BSA (tritium incorporated by reductive methylation810) showed accelerated renal clearance (fractional clearance determined with perfused rate kidney). 14C-Labeled rat serum albumin (reductive methylation with 14C-labeled formaldehyde811) has a lower fractional clearance. Heating at acid pH (70°C at pH 3) markedly reduced fractional clearance, while heating at basic pH (70°C at pH 12.5) increased fractional clearance. The acid expansion transition (N → F transition) described earlier could explain the reduced fractional clearance; the base treatment likely results in the rupture of the disulide bonds, resulting in the “extension” of the albumin molecule with an increase in lexibility. Rennke and Venkatachalan812 compared the clearance of

134

Biotechnology of Plasma Proteins

dextran with that of horseradish peroxidase, both of which have a Stokes–Einstein radius of 2.85 nm. Dextran has a fractional clearance rate of 0.485, whereas the horseradish peroxidase has a fractional clearance of 0.068. These researchers suggest that shape and lexibility are important factors in glomerular iltration. It is not unreasonable, then, to speculate that the base-treated albumin in the Clavant studies809 has its structure disrupted by disulide bond cleavage and such a structure is more lexible than the native protein. Nevertheless, molecular size as expressed by Stokes–Einstein radius is considered a major factor in renal clearance but not the only factor.813 Lundström and coworkers814 showed that bikunin, which is similar in size to albumin (Stokes–Einstein radius of ~3.6 nm), has a fractional clearance rate some 80-fold higher than that of albumin. These authors argue that the increased lexibility of bikunin is responsible for the increased rate of clearance. Nephrotic syndrome is a kidney disorder815 characterized by high protein content in the urine (proteinuria816,817) and a decreased plasma protein concentration. Plasma albumin concentration in the circulation is markedly decreased in nephrotic syndrome.818–820 The value of 15–20 days is considerably longer than the half-lives of the various coagulation factors, which are of similar shape and size but similar to that of IgG. It is not surprising then that IgG and albumin share a protection receptor mechanism, in which the protein after endocytosis is not presented for lysosomal degradation but rather returned to the circulation.821–823 The processing of IgG seems a bit more complicated than that of albumin,824 and the function of the neonatal receptor is more diverse than just recycling.825 The role of the FcRn receptor in maternal transmission of immunoglobulin is an example of the complex function of the FcRn receptor. The comparatively long half-life has encouraged the use of albumin as a fusion partner for therapeutic use and has been discussed earlier. It is not clear if prolongation consistent with the recycling of albumin has been achieved. It seems more likely that, as with PEGylation,826 prolongation of half-life relects primarily decreased renal clearance, although decreased proteolysis by endogenous serum proteases may also be a factor.827,828

ANALYTICAL METHODS FOR ALBUMIN, INCLUDING USE OF ALBUMIN AS STANDARD FOR ANALYTICAL METHODS The measurement of albumin in serum or plasma can be accomplished by a variety of methods. The early approaches were based on the Tselius free-boundary electrophoretic system described in Chapter 3, as used by Longsworth and coworkers to determine the albumin/globulin ratio,829 and/or the differential solubility of the albumin fraction and globulin fraction of plasma.830–832 The albumin/globulin ratio was popular as a diagnostic tool.2,829,832 The albumin/globulin ratio is no longer used extensively in human medicine but is still of great interest in veterinary medicine.833–835 Current approaches to the measurement of albumin in plasma and serum use dye binding, immunonephelometry, immunoturbidity, radial immunodiffusion, and “dipstick.” 836,839 Point-of-care methodology has been developed for the measurement of albumin in urine.838,839 The clinical measurement of albumin in blood uses the spectral changes on the binding of bromcresol green or bromcresol purple.840 The relatively speciic binding

Albumin

135

of dyes by albumin in plasma and serum has been known for some time. It was possible to locate citations to work in the early part of the last century when albumin was used as a medium for printing. The development of dye-binding for the analysis of albumin in serum and urine has been reviewed by Peters1 and Doumas and Peters.840 Before the days of pH meters, which provide a value for the pH of the solution together with a personality analysis of the operator, pH in solution was measured with a series of dyes that underwent spectral changes at certain pH.841 Following an observation by Klotz and Walker842 observed that the presence of protein caused changes in the observed spectrum of dyes used for the measurement of pH; the spectrum of a dye in combination with a protein may differ in intensity (hyperchromic or hypochromic) and wavelength for maximum absorbance (red or bathochromic shift or blue or hypsochromic shift). This change in the absorbance characteristics of dyes bound to protein stimulated efforts over the next 20 years to make this method useful for the speciic measurement of albumin in serum, but it was not until the work of Doumas and coworkers that this resulted in the use of bromcresol green for the measurement of albumin.843 The use of bromcresol green for albumin measurement remains popular,844 and the binding reaction remains under study.845–847 Before moving on, it would be useful to spend some time on Irving Klotz. Klotz was one of the several giants of physical biochemistry in the period immediately following World War II. Professor Klotz spent his career at Northwestern University and made major contributions to our understanding of receptor–ligand interactions,848 providing a strong foundation for pharmaceuticals and biopharmaceuticals directed at cell surface receptors. Dye binding to albumin is used for purposes other than determination of albumin concentration. Carroll849 used a dye–albumin complex (Orange I-BSA) as a substrate for proteolytic enzymes. Tang and coworkers850 used albumin labeled with two different luorophores (lucifer yellow and rhodamine) as a substrate for proteases. Lucifer yellow luorescence was quenched by rhodamine; subtilisin reduced the luorescence-quenching transfer. A similar approach used relief of luorescence quenching of a luorophor-labeled albumin to visualize ibroblast cell migration in a hydrogel subject to proteolytic degradation.851 Most methods for the determination of protein concentration in solution require the use of a reference standard. The author has published a review on the methods for the determination of protein concentration,852 and it is clear that BSA is the reference standard of choice. It is best to purchase a standard solution of BSA; if a “homemade” standard is prepared, it should be validated by either absorbance at 280 nm or amino acid analysis such as for the clinical albumin standard.853,854 However, BSA as a standard is a generalization and may not be useful for all assays and all proteins855 with serious potential consequences for therapeutic proteins.856 BSA is used far more often than the human protein as a standard for the determination of protein concentration as well as for other analytical methods. Yang and coworkers used both BSA and ovalbumin as model proteins for the study of nitration,857 while Mortstedt and coworkers used BSA as a model protein for oxidation studies.858 A number of other researchers have used albumin as a model protein for the development of techniques involving mass spectrometry.859–863 In a study cited earlier,802 Ding and coworkers used HSA labeled with

136

Biotechnology of Plasma Proteins

5-aminoluorescein to discriminate between brain tumor tissue and normal tissue for glioma surgery.

ALBUMIN AS EXCIPIENT Albumin, both of bovine and human origin, has a long history of being used to stabilize biologicals.864,865 There are few studies on any effect, other than stability, produced by the inclusion of HSA (or BSA) as an excipient/stabilizing agent. Zhi and coworkers at Roche866 showed that the presence of albumin did not affect the pharmacokinetics/bioequivalency of interferon α-2a in volunteer human subjects. Hawe and Friess867 observed that the inluence of excipients, including albumin on the freezing process, must be considered to avoid collapse and product stability problems during lyophilization. This is a useful study that employed DSC, cryomicroscopy, and powder x-ray diffraction to study the effect of excipients on the freezing process. In a later study,868 the same researchers observed that the positive effect of NaCl on the stabilization of a hydrophobic cytokine was due to an effect on HSA and subsequent overall stabilization of the product. In one interesting recent study, Warren and coworkers869 observed that progastrin-releasing peptide assay standards were far more stable in BSA pasteurized at pH 3.0 than at neutral pH.

CONCLUSION Albumin continues to be a protein with many facets remaining to be explored: • Further research is needed to ind out whether the binding of fatty acids is useful just for transport or whether there is a more sophisticated function in pathologies, such as metabolic syndrome.870,871 • The molecular basis for the development of ischemia-modiied albumin needs to be clariied. Regardless of speciicity, it is an interesting transition that might provide more information about oxidative stress and the binding of fatty acids. • The MARS system has the potential of serving as a viable “bridge-to-transplant,” which may be more critical with an aging population. • The potential of albumin as a drug carrier should be explored in more detail. There is considerable potential for the S-nitroso product and for albumin as a carrier of prodrugs.

REFERENCES 1. Peters, T., Jr., All About Albumin. Biochemistry, Genetics, and Medical Applications, Academic Press, San Diego, CA, 1996. 2. Sandor, G., Historical developments of the physicochemical and immunological methods of protein identiication and separation, in Serum Proteins in Health and Disease, Chapter 1, pp. 3–51, Williams & Wilkins, Baltimore, MD, 1966. 3. Index Medicus, Carnegie Institute of Washington, 1903. 4. Deeming, D.C., Double-yolked pheasant eggs provide an insight into the albumen secretion in bird eggs, Br. Poult. Sci. 52, 40–47, 2011.

Albumin

137

5. Denis, Memoire sur de Sang, Bailliere et Fils, Paris, France, 1840. 6. Panum, P., Ueber einen constanten, mit dem Casein übeeinsimmenden Bestandtheil des Blutes, 3, 251–264, 1853. 7. Loeb, J. and Loeb, R.F., The inluence of electrolytes on the solution and precipitation of casein and gelatin, J. Gen. Physiol. 4, 187–211, 1921. 8. Cohn, E.J., Jr., The physical chemistry of the proteins, Physiol. Rev. 5, 349–437, 1925. 9. Hughes, W.L., Interstitial proteins: The proteins of blood plasma and lymph, in The Proteins, eds. H. Neurath and K. Bailey, Vol. II, Part B, Chapter 21, pp. 664–734, Academic Press, New York, 1954. 10. Osbourne, T.B., in The Vegetable Proteins, 2nd edn., Monographs in Biochemistry, eds. R.H.A. Plimmer and F.G. Hopkins, Longmans, Green, and Co., London, UK, 1924. 11. Pillemer, L., Ecker, E.E., Oncley, J.L., and Cohn, E.J., The preparation and physicochemical characterization of the serum protein component of complement, J. Exp. Med. 74, 297–308, 1941. 12. Tanford, C., ‘Cohn and Edsall’: Physical chemistry conclusively supports a protein model, Biophys. Chem. 100, 81–90, 2003. 13. Chick, H. and Martin, C.J., XXXIX. The precipitation of egg-albumin by ammonium sulphate: A contribution to the theory of the “salting-out” of proteins, Biochem. J. 7, 380–388, 1917. 14. Cohn, E.J. and Conant, J.B., Molecular weight determination of proteins in phenol, Zeit. Physiol. Chem. 159, 93–101, 1926. 15. Prideaux, E.B. and Howitt, F.O., The electrophoretic velocities of gelatin and ovalbumin in different concentrations of their mixtures and the effect of ultraviolet irradiation, Biochem. J. 25, 391–402, 1931. 16. Villee, C.A., Linderstrøm-Lang, K., Ottesen, M., et al., The enzymatic conversion of ovalbumin to plakalbumin, Biol. Bull. 99(2), 322–323, 1950. 17. Loeb, J., Proteins and the Theory of Colloid Behavior, 2nd edn., McGraw-Hill, New York, 1924. 18. Bayne-Jones, S., Equilibria in precipitin reactions: The coexistence of a single free antigen and its antibody in the same serum, J. Exp. Med. 25, 837–853, 1917. 19. Rothen, A. and Landsteiner, K., Serological reactions of protein ilms and denatured proteins, J. Exp. Med. 76, 437–450, 1942. 20. Cohn, M., Wetter, L.R., and Deutsch, H.F., Immunological studies on egg white proteins: Precipitation of chicken-ovalabumin and conalbumin by rabbit- and horse-antisera, J. Immunol. 61, 283–296, 1949. 21. Sherman, W.B. and Coulson, E.J., Passive sensitization of human skin by sera of guinea pigs anaphylactically sensitized to ovalbumin, Proc. Soc. Exp. Biol. Med. 77, 245–247, 1951. 22. Eeg-Larsen, N., Lindestrøm-Lang, K., and Ottesen, M., Transformation of ovalbumin into plakalbumin, Arch. Biochem. 19, 340–344, 1948. 23. Smith, M.B., The isolation of a large peptide from denatured plakalbumin, Biochim. Biophys. Acta 154, 263–266, 1968. 24. Gettins, P., Absence of large scale conformational change upon limited proteolysis of ovalbumin, the proteolytic serpin, J. Biol. Chem. 264, 3781–3785, 1989. 25. Cacace, M.G., Landau, E.M., and Ramsden, J.J., The Hofmeister series: Salt and solvent effects on interfacial phenomena, Q. Rev. Biophys. 30, 241–277, 1997. 26. Zhang, Y. and Cremer, P.S., Interactions between macromolecules and ions: The Hofmeister series, Curr. Opin. Chem. Biol. 10, 658–663, 2006. 27. Hofmeister, F., Zur Lehre von der Wirkung der Salze, Arch. Exp. Pathol. Pharmacol. 24, 247–260, 1888. 28. Otani, M., Taniguchi, T., Sakai, A., et al., Phosphoproteome proiling using a luorescent phosphosensor dye in two-dimensional polyacrylamide gel electrophoresis, Appl. Biochem. Biotechnol. 164(6), 804–818, 2011.

138

Biotechnology of Plasma Proteins

29. Ishimaru, T., Ito, K., Tanaka, M., et al., The role of the disulide bridge in the stability and structural integrity of ovalbumin evaluated by site-directed mutagenessis, Biosci. Biotechnol. Biochem. 75(3), 544–549, 2011. 30. Martos, G., Lopez-Exposito, I., Becharitiwon, R., et al., Mechanisms underlying differential food allergy response to heated egg, J. Allergy Clin. Immunol. 127(4), 990–997, 2011. 31. Lundblad, L.K., Rinaldi, L.M., Poynter, M.E., et al., Detrimental effects of albuterol on airway responsiveness requires airway inlammation and is independent of β-receptor afinity in murine models of asthma, Respir. Res. 12(1), 27, 2011. 32. Stensballe, A., Andersen, S., and Jensen, O.N., Characterization of phosphoproteins from electrophoretic gels by nanoscale Fe(III) afinity chromatography with off-line mass spectrometry analysis, Proteomics 1, 207–222, 2001. 33. Novotna, L., Hruby, M., Benes, M.J., and Kucerova, K., Immobilized metal afinity chromatography of phosphorylated proteins using high performance sorbents, Chromatographia 68, 381–386, 2008. 34. Bradshaw, R.A. and Peters, T., Jr., Amino acid sequence of peptide 1–24 of rat and human serum albumins, J. Biol. Chem. 244, 5582–5589, 1969. 35. Brown, J.R., Structural origins of mammalian albumin, Fed. Proc. 35, 2141–2144, 1976. 36. Heyl, J.T., Gibson 2nd, J.G., and Janeway, C.A., Studies on the plasma proteins. V. The effect of concentrated solutions of human and bovine serum albumin on blood volume after acute blood loss in man, J. Clin. Invest. 22, 763–773, 1943. 37. Blood Programs in World War II, ed. D.B. Kendrick, The Human and Bovine Albumin Programs, Chapter 12, Ofice of the Surgeon General, Department of the Army, United States Publishing Ofice, Washington, DC, 1964. 38. Andersen, D.C., Koch, C., Jensen, C.H., et al., High prevalence of human anti-bovine IgG antibodies as the major cause of false positive reactions n two-site immunoassays based on monoclonal antibodies, J. Immunoassay Immunochem. 25, 17–30, 2004. 39. Mogues, T., Li, J., Coburn, J., and Kuter, D.J., IgG antibodies against bovine serum albumin in humans—Their prevalence and response to exposure to bovine serum albumin, J. Immunol. Meth. 300, 1–11, 2005. 40. Moneret-Vautrin, A., Wal, J.M., Guillet-Rossof, F., et al., Bovine serum albumin immunization: A new risk of allergy during protocols for in vitro fertilization, Allergy 46, 228–234, 1991. 41. Linetsky, E., Kenyon, N., Li, H., et al., Increased immunogenicity of human vertebral body marrow after processing in bovine versus human serum albumin, Transplant. Proc. 29, 1960, 1997. 42. Mackensen, A., Dräger, R., Schlesier, M., et al., Presence of IgE antibodies to bovine serum albumin in a patient developing anaphylaxis after vaccination with human peptide-pulsed dendritic cells, Cancer Immunol. Immunother. 49, 152–156, 2000. 43. Pagán, J.A., Postigo, I., Rodriguez-Pacheco, J.R., et al., Bovine serum albumin contained in culture medium used in artiicial insemination is an important anaphylaxis risk factor, Fertil. Steril. 90, 2013.e17–2013.e19, 2008. 44. Berson, S.A., Yalow, R.S., Schreiber, S.S., and Post, J., Tracer experiments with I131 labeled serum albumin: Distribution and degradation studies, J. Clin. Invest. 32, 746– 768, 1953. 45. Rothschild, M.A., Bauman, A., Yalow, R.S., and Berson, S.A., Tissue distribution of I131 labeled human serum albumin following intravenous administration, J. Clin. Invest. 34, 1354–1358, 1955. 46. Bert, J.L., Pearce, R.H., and Mathieson, J.M., Concentration of plasma albumin in its accessible space in postmortem human dermis, Microvasc. Res. 32, 211–223, 1986. 47. Henriksen, J.H. and Schlichting, P., Increased extravasation and lymphatic return rate of albumin during diuretic treatment of ascites in patients with liver cirrhosis, Scand. J. Clin. Lab. Invest. 41, 589–599, 1981.

Albumin

139

48. Galatius, S., Bent-Hansen, L., Wroblemwski, H., et al., Plasma disappearance of albumin and impact of capillary thickness in idiopathic dilated cardiomyopathy and after heart transplantation, Circulation 102, 319–325, 2000. 49. Fleck, A., Hawker, F., Wallace, P.I., et al., Increased vascular permeability: A major cause of hypoalbuminemia in disease and injury, Lancet I, 781–783, 1985. 50. Parving, H.P. and Gyntelberg, F., Transcapillary escape rate of albumin and plasma volume in essential hypertension, Circ. Res. 32, 643–651, 1973. 51. Atkinson, J.P., The fractional precipitation of the globulin and albumin of normal horse serum and diptheria antitoxic serum, and antitoxic strength of the precipitates, J. Exp. Med. 5, 67–76, 1900. 52. Oswald, A., Crystallization of human serum albumin, Z. Physiol. Chem. 95, 102–103, 1915. 53. Adair, M.E. and Taylor, G.L., Crystallization of human serum albumin, Nature 135, 207, 1935. 54. Martin, C.J., A rapid method of separating colloids from crystalloids in solutions containing both, J. Physiol. 20, 364–371, 1896. 55. Hewlett, M.B., On fractional heat-coagulation, J. Physiol. 13, 493–512, 1892. 56. Kylin, E., Isoelectric point of native human serum albumin, Acta Med. Scand. 87, 536–550, 1938. 57. Roche, A., Doner, M., and Marquet, F., Molecular weight of human serum albumin, Compt. Rendus Seanc. 119, 1150–1151, 1935. 58. Zwaan, J., Estimation of molecular weights of proteins by polyacrylamide gel electrophoresis, Anal. Biochem. 21, 155–168, 1967. 59. Squire, P.G., Moser, P., and O’Kansk, C.T., The hydrodynamic properties of bovine serum albumin and dimer, Biochemistry 7, 4261–4271, 1968. 60. Mann, F.C., Further experimental study of surgical shock, J. Am. Med. Assoc. 71, 1184– 1188, 1918. 61. Blood Programs in World War II, ed. D.B. Kendrick, Ofice of the Surgeon General, Department of the Army, United States Publishing Ofice, Washington, DC, 1964. 62. Ebert, R.V., Stead, E.A., and Gilbson 2nd, J.G., Response of normal individuals in acute blood loss, Arch. Intern. Med. 68, 578–590, 1941. 63. Scatchard, G., Batchelder, A.C., and Brown, A., Chemical, clinical, and immunological studies on the products of human plasma fractionation. VI. The osmotic pressure of plasma and of serum albumin, J. Clin. Invest. 23, 458–464, 1944. 64. Bisonni, R.S., Holtgrave, D.R., Lawler, F., and Marley, D.S., Colloids versus crystalloids in luid resuscitation: An analysis of randomized controlled trials, J. Fam. Pract. 32, 387–390, 1991. 65. Hemington-Gorse, S.J., Colloid or crystalloid for resuscitation of major burns, J. Wound Care 14, 256–258, 2005. 66. Perel, P. and Roberts, I., Colloids versus crystalloids for luid resuscitation in critically ill patients, Cochrane Database Syst. Rev. 17(4), CD000567, 2007. 67. Hiltebrand, L.B., Kimberger, O., Arnberger, M., et al., Crystalloids versus colloids for goal directed luid therapy in major surgery, Crit. Care 13(2), R40, 2009. 68. Guidet, B., Soni, N., Della Rocca, G., et al., A balanced view of balanced solutions, Crit. Care 14(5), 325, 2010. 69. Fan, E. and Steward, T.E., Albumin in critical care: SAFE, but worth its salt?, Crit. Care 8, 297–299, 2004. 70. Myburgh, J.A. and Finfer, S., Albumin is a blood product too—Is it safe for all patients?, Crit. Care Resusc. 11, 67–70, 2009. 71. Chalidis, B., Kanakaris, N., and Giannooudis, P.V., Safety and eficacy of albumin administration in trauma, Expert Opin. Drug Saf. 6, 407–415, 2007. 72. Levi, M. and de Jonge, E., Clinical relevance of the effects of plasma expanders on coagulation, Semin. Thromb. Hemost. 33, 810–815, 2007.

140

Biotechnology of Plasma Proteins

73. Scatchard, G., The attraction of proteins for small molecules and ions, Ann. N. Y. Acad. Sci. 51, 660–672, 1949. 74. Hamacek, J. and Piguet, C., How to adapt Scatchard plot for graphically addressing cooperativity in multicomponent self-assemblies, J. Phys. Chem. B 110, 7783–7792, 2006. 75. Meloun, B., Morávek, L., and Kostka, V., Complete amino acid sequence of human serum albumin, FEBS Lett. 58, 134–137, 1975. 76. Lawn, R.M., Adelman, J., Bock, S.C., et al., The sequence of human serum albumin cDNA and its expression in E. coli, Nucleic Acids Res. 9, 6103–6114, 1981. 77. Debaiczyk, A., Law, S.W., and Dennison, O.E., Nucleotide sequence and the encoded amino acids of human serum albumin mRNA, Proc. Natl. Acad. Sci. USA 78, 71–75, 1982. 78. Ho, J.X., Holowachuk, E.W., Norton, E.J., et al., X-ray and primary structure of horse serum albumin (Equus caballus) at 0.27-nm resolution, Eur. J. Biochem. 215, 205–212, 1993. 79. Hein, K.L., Kragh-Hansen, U., Morth, J.P., et al., Crystallographic analysis reveals a unique lidocaine binding site on human serum albumin, J. Struct. Biol. 171, 353–360, 2010. 80. Nagasawa, T. and Era, S., Alteration of redox state of human serum albumin before and after hemodialysis, Blood Purif. 22, 525–529, 2004. 81. Cha, M.K. and Kim, I.H., Disulide between Cys392 and Cys438 of human serum albumin is redox-active, which is responsible for the thioredoxin-supported lipid peroxidase activity, Arch. Biochem. Biophys. 445, 19–25, 2006. 82. Oettl, K. and Marsche, G., Redox state of human serum albumin in terms of cysteine-34 in health and disease, Meth. Enzymol. 474, 181–195, 2010. 83. Ikegaya, K., Hirose, M., Ohmura, T., and Nokihara, K., Complete determination of disulide forms of puriied recombinant human serum albumin, secreted by the yeast Pichia pastoris, Anal. Chem. 69, 1986–1991, 1997. 84. He, X.M. and Carter, D.C., Atomic structure and chemistry of human serum albumin, Nature 358, 205–215, 1992. 85. Sugio, S., Kashima, A., Mochizuki, S., et al., Crystal structure of human serum albumin at 2.5 Å resolution, Protein Eng. 12, 439–446, 1999. 86. Leggio, C., Galantini, L., and Pavel, N.V., About the albumin structure in solution: Cigar expanded form versus heart normal shape, Phys. Chem. Chem. Phys. 10, 6741–6750, 2008. 87. Oncley, J.L. and Ferry, J.D., Studies on the dielectric properties of protein solutions. II. The water soluble proteins of horse serum, J. Am. Chem. Soc. 60, 1123–1132, 1938. 88. Ho, J.X., Holowachuk, E.W., Norton, E.J., et al., X-ray and primary structure of horse serum albumin (Equus caballus) at 0.27-nm resolution, Eur. J. Biochem. 215, 205–212, 1993. 89. Oncley, J.L., Dielectric behavior and atomic structure of serum albumin, Biophys. Chem. 100, 151–158, 2003. 90. Curry, S., Mandelkow, H., Brick, P., and Franks, N., Crystal structure of human serum albumin complexed with fatty acids reveals an asymmetric distribution of binding sites, Nat. Struct. Biol. 5, 827–835, 1998. 91. Oncley, J.L., Jensen, C.C., and Gross, P.M., Jr., Dielectric constant studies of zein solutions, J. Phys. Colloid Chem. 53, 162–175, 1949. 92. Warshel, A. and Papazyan, A., Electrostatic in macromolecules: Fundamental concepts and practical modeling, Curr. Opin. Struct. Biol. 8, 211–217, 1998. 93. Matthew, J.B. and Richards, F.M., Anion binding are pH dependent electrostatic effects in ribonuclease, Biochemistry 21, 4989–4999, 1982. 94. Bircan, C. and Barringer, S.A., Use of dielectric properties to detect protein denaturation, J. Microw. Power Electromagn. Energy 37, 89–96, 2002.

Albumin

141

95. Schneider, W., Dintzis, H.W., and Oncley, J.L., Changes in the electric dipole vector of human serum albumin due to complexing with fatty acids, Biophys. J. 16, 417–431, 1976. 96. Luik, A.I., Naboka, Y.N., Mogilevich, S.E., et al., Study of the structure of human serum albumin structure by dynamic light scattering: Two types of reaction under different pH and interaction with physiologically active compounds, Spectrochim. Acta A 54, 1503–1507, 1998. 97. Farruggia, B., Garcia, G., D’Angelo, C., and Picóm, G., Destabilization of human serum albumin by polyethylene glycol studied by thermodynamic equilibrium and kinetic approaches, Int. J. Biol. Macromol. 20, 43–51, 1997. 98. Totani, K., Ihara, Y., Matsuo, I., and Ito, Y., Effects of macromolecular crowding on glycoprotein processing enzymes, J. Am. Chem. Soc. 130, 2101–2107, 2008. 99. Okamoto, D.N., Oliveira, L.C.G., Kundo, M.Y., et al., Increase of SARS CoV 3CL peptidase activity due macromolecular crowding effects in the mileus composition, Biol. Chem. 391, 1461–1468, 2010. 100. Ragi, C., Sedaghat-Herati, M.R., Quameur, A.A., and Tajmir-Rishi, H.A., The effects of poly(ethylene)glycol on the solution structure of human serum albumin, Biopolymers 78, 231–236, 2005. 101. Wasylaschuk, W.R., Harmon, P.A., Wagner, G., et al., Evaluation of hydroperoxides in common pharmaceutical excipients, J. Pharm. Sci. 96, 106–116, 2007. 102. Luetscher, J., Serum albumin II. Identiication of more than one albumin in horse and human serum by electrophoretic in acid solution, J. Am. Chem. Soc. 61, 2888–2890, 1949. 103. Foster, J.F., Plasma albumin, in The Plasma Proteins, ed. F.W. Putnam, Vol. 1, Academic Press, New York, 1960. 104. Schmid, K., Characterization of an “electrophoretically homogeneous” human serum albumin, J. Am. Chem. Soc. 79, 4679–4682, 1957. 105. Schmid, K., Electrophoresis of human serum albumin at pH 4.0. I. A systematic study on the effect of organic acids and alcohols upon the electrophoretic behavior of albumin, J. Biol. Chem. 234, 3163–3168, 1959. 106. Nishimukai, H., Kera, Y., Sano, Y., et al., Two new slow-moving variants of human serum albumin, Vox Sang. 42, 313–317, 1982. 107. Tanford, C., Swanson, S.A., and Shore, W.S., Hydrogen ion equilibria of bovine serum albumin, J. Am. Chem. Soc. 77, 6414–6421, 1955. 108. Tanford, C., Buzzell, J.G., Rands, D.G., and Swanson, S.A., The reversible expansion of bovine serum albumin in acid solution, J. Am. Chem. Soc. 77, 6421–6428, 1955. 109. Wilting, J., Weideman, M.M., Doomer, A.C.J., et al., Conformational changes in human serum albumin around the neutral pH from circular dichroism measurements, Biochim. Biophys. Acta 579, 469–473, 1979. 110. Kasai-Morita, S., Horie, T., and Awazu, S., Inluence of the N-B transition of human serum albumin on the structure of the warfarin-binding site, Biochim. Biophys. Acta 915, 277–283, 1987. 111. Wanwimolruk, S. and Birkett, D.J., The effects of N-B transition of human serum albumin on the speciic drug-binding sites, Biochim. Biophys. Acta 709, 247–255, 1982. 112. Kosa, T., Maruyuma, T., Sakai, N., et al., Species differences of serum albumin. III. Analysis of structural characteristics and ligard binding properties during N-B transition, Pharm. Res. 15, 592–598, 1998. 113. Bos, O.J.M., Labra, J.F.A., Fisher, M.J.E., et al., The molecular mechanism of the neutral-to-base transition of human serum albumin. Acid/base titration and proton nuclear magnetic resonance studies on a large petpdie and a large tryptic fragment of albumin, J. Biol. Chem. 264, 963–969, 1989. 114. Blindauer, C.A., Harvey, I., Bunyan, K.E., et al., Structure, properties, and engineering of the major zinc binding on human albumin, J. Biol. Chem. 284, 23116–23124, 2009.

142

Biotechnology of Plasma Proteins

115. Harrington, W.F., Johnson, P., and Ottewill, R.H., Bovine serum albumin and its behavior in acid solution, Biochem. J. 62, 569–582, 1956. 116. Sogami, M. and Foster, M.F., Isomerization reactions of charcoal-defatted bovine plasma albumin. The N-F transition and acid expansion, Biochemistry 7, 2172–2182, 1968. 117. Geisow, M.J. and Beaven, M.H., Large fragments from human serum albumin, Biochem. J. 161, 619–625, 1977. 118. Geisow, M.J. and Beaven, M.H., Physical and binding properties of large fragments of human serum albumin, Biochem. J. 163, 477–484, 1977. 119. Weber, G. and Young, L.B., Fragmentation of bovine serum albumin by pepsin. I. The origin of the acid expansion of the albumin molecule, J. Biol. Chem. 239, 1415–1423, 1964. 120. Grimsley, G.R., Scholtz, J.M., and Pace, C.N., A summary of the measured pK values of the ionizable groups in folded proteins, Protein Sci. 18, 247–251, 2009. 121. Foster, J.F., Some aspects of the structure and conformational properties of serum albumin, in Albumin: Structure, Function, and Uses, eds. V.M. Rosenoer, M. Oratz, and M. Rothschild, pp. 53–84, Oxford/Pergammon, New York, 1977. 122. Steinhardt, J. and Stocker, N., Effects of high pH and sodium dodecyl sulfate on the hidden tyrosines of human serum albumin, Biochemistry 12, 1789–1797, 1973. 123. Honoré, B., Stopped-low studies of spectral changes in human serum albumin following an alkaline pH jump, J. Biol. Chem. 262, 14935–14938, 1987. 124. Cohen, P., Effect of heating on human serum albumin, in Proceedings of the Workshop on Albumin, pp. 323–332, DHEW Publication No. (NIH) 76-925, U.S. Superintendent of Documents, Washington, DC, 1975. 125. Hoofnagle, J.H., Barker, L.F., Thiel, J., and Gerety, R.J., Hepatitis B virus and hepatitis B surface antigen in human albumin products, Transfusion 16, 141–147, 1976. 126. Hansen, J.F. and Ezban, M., A new high quality albumin for therapeutic use, Dev. Biol. Stand. 48, 105–112, 1980. 127. Adcock, W.L., MacGregor, A., Davies, J.R., et al., Chromatographic removal and heat inactivation of hepatitis B virus during the manufacture of human albumin, Biotechnol. Appl. Biochem. 28, 169–178, 1998. 128. Erstad, B.L., Viral infectivity of albumin and plasma protein fraction, Pharmacotherapy 16, 996–1001, 1996. 129. Schegel, A., Immelmann, A., and Kempf, C., Virus inactivation of plasma-derived proteins by pasteurization in the presence of guanidine hydrochloride, Transfusion 41, 382–389, 2001. 130. Lin, J.J., Meyer, J.D., Carpenter, J.F., and Manning, M.C., Aggregation of human serum albumin during a thermal viral inactivation, Int. J. Biol. Macromol. 45, 91–96, 2009. 131. Roelands, J.F., Moody, J.F., and Cohen, P., Effects of repeated heating on human albumin, Vox Sang. 26, 415–424, 1974. 132. Finlayson, J.S., Reamer, B.L., and Young, A.M., Dissociation of human albumin dimer by heating, Vox Sang. 26, 457–461, 1974. 133. Kolthoff, I.M. and Tan, B.H., Reactivity of sulfhydryl and disulide in proteins. VI. Effect of heat denaturation of bovine serum albumin (BSA) on sulfhydryl and reactive disulide content, J. Am. Chem. Soc. 87, 2717–2720, 1966. 134. Fischer, B., Sumner, I., and Goodenough, P., Isolation and renaturation of bio-active proteins expressed in Escherichia coli as inclusion bodies, Arzneimittelforschung 42, 1512–1515, 1992. 135. Moriyama, Y., Watanabe, E., Kobayashi, K., et al., Secondary structural change of bovine serum albumin in thermal denaturation up to 130°C and protective effects of sodium dodecyl sulfate on the change, J. Phys. Chem. B 112, 16585–16589, 2008. 136. Lohner, K., Sen, A.C., Prankord, R., et al., Effects of drug binding on the thermal denaturation of human serum albumin, J. Pharm. Biomed. Anal. 12, 1501–1505, 1994.

Albumin

143

137. Pico, G., Thermodynamic aspects of the thermal stability of human serum albumin, Biochem. Mol. Biol. Int. 36, 1017–1023, 1995. 138. Michnik, A., DSC study of the association of ethanol with human serum albumin, J. Therm. Anal. Calorim. 87, 91–96, 2007. 139. Wallevik, K., Reversible denaturation of human serum albumin by pH, temperature, and guanidine hydrochloride followed by optical rotation, J. Biol. Chem. 248, 2650–2653, 1973. 140. Aoki, K., Murata, M., and Hiramatsu, K., Urea denaturation of bovine serum albumin at pH 9, Anal. Biochem. 59, 146–157, 1974. 141. King, J., Haase-Pettingell, C., Robinson, A.S., et al., Thermolabile folding intermediates: Inclusion body precursors and chaperonin substrates, FASEB J. 10, 57–66, 1996. 142. González-Jiménez, J. and Cortijo, M., Urea-denaturation of human serum albumin labeled with acrylodan, J. Protein Chem. 21, 75–79, 2002. 143. Clark, I.D. and Burtnick, L.D., Fluorescence of equine platelet tropomyosin labeled with acrylodan, Arch. Biochem. Biophys. 260, 595–600, 1988. 144. Baudier, J. and Cole, R.D., Reinvestigation of the sulfhydryl reactivity in bovine brain S100b (ββ) protein and the microtubule-associated tau proteins. Ca2+ stimulates disulide cross-linking between the S100b β-subunit and the microtubule-associated tau(2) protein, Biochemistry 27, 2728–2736, 1988. 145. Sommer, A., Gores, R., Kostner, G.M., et al., Sulfhydryl-selective luorescence labeling of lipoprotein(a) reveals evidence for one single disulide linkage between apoproteins(a) and B-100, Biochemistry 30, 11245–11249, 1991. 146. Varshney, A., Ahmad, B., and Khan, R.H., Comparative studies of unfolding and binding of ligands to human serum albumin in the presence of fatty acids: Spectroscopic approach, Int. J. Biol. Macromol. 42, 483–490, 2008. 147. Leggio, C., Galantini, L., Konarev, P.V., and Pavel, N.V., Urea-induced denaturation process on defatted serum albumin and in the presence of palmitic acid, J. Phys. Chem. B 113, 12590–12602, 2009. 148. Mendez, D.L., Jensen, R.A., McElroy, L.A., et al., The effect of non-enzymatic glycation on the unfolding of human serum albumin, Arch. Biochem. Biophys. 444, 92–99, 2005. 149. Gaujoux-Viala, C., Smolen, J.S., Landewé, R., et al., Current evidence for the management of rheumatoid arthritis with synthetic disease-modifying antirheumatic drugs: A systematic literature review informing the EULAR recommendations for the management of rheumatoid arthritis, Ann. Rheum. Dis. 69, 1004–1009, 2010. 150. van Roon, E.N., van den Bemt, P.M., Jansen, T.L., et al., An evidence-based assessment of the clinical signiicance of drug–drug interactions between disease-modifying antirheumatic drugs and non-antirheumatic drugs according to rheumatologists and pharmacists, Clin. Ther. 31, 1737–1746, 2009. 151. Tiekink, E.R., Anti-cancer potential of gold complexes, Inlammopharmacology 16, 138–142, 2008. 152. Kast, R.E., Glioblastoma invasion, cathepsin B, and the potential for both to be inhibited by auranoin, an old-anti-rheumatoid arthritis drug, Cen. Eur. Neurosurg. 71, 139–142, 2010. 153. Ecker, D.J., Hempel, J.C., Sutton, B.M., et al., Reactions of the metallodrug Auranoin [(1-thio-β-D-glucopyranose-2,3,4,6-tetraaceto-S)(triethylphosphine)gold] with biological ligands studied by radioisotope methodology, Inorg. Chem. 25, 3139–3143, 1986. 154. Christodoulou, J., Sadler, P.J., and Tucker, A., A new structural transition of serum albumin dependent on the state of Cys34. Detection by 1H-NMR spectroscopy, Eur. J. Biochem. 225, 363–368, 1994. 155. Talib, J., Beck, J.L., and Ralph, S.F., A mass spectrometric investigation of the binding of gold antiarthritic agents and the metabolite [Au(CN)2]‐ to human serum albumin, J. Biol. Inorg. Chem. 11, 559–570, 2006.

144

Biotechnology of Plasma Proteins

156. Christodopoulou, J., Sadler, P.J., and Tucker, A., 1H NMR of albumin in human plasma: Drug binding and redox reactions at Cys34, FEBS Lett. 376, 1–5, 1995. 157. Roberts, J.R., Xiao, J., Schliesman, B., et al., Kinetics and mechanism of the reaction between serum albumin and auranoin (and its isopropyl analogue) in vitro, Inorg. Chem. 35, 424–433, 1996. 158. Ahmad, S., Isab, A.A., Ali, S., and Al-Arfaj, A.R., Perspectives in bioinorganic chemistry or some metal based therapeutic agents, Polyhedron 25, 1633–1645, 2006. 159. Ohkubo, A., On the conformation around the sulfhydryl group in human serum albumin, J. Biochem. 45, 879–888, 1969. 160. Buck, M., Triluoroethanol and colleagues: Cosolvents come of age. Recent studies with peptides and proteins, Q. Rev. Biophys. 31, 297–355, 1998. 161. Kumar, Y., Tayyab, S., and Muzammil, S., Molten-globule like partially folded states of human serum albumin induced by luoro and alklyl alcohols at low pH, Arch. Biochem. Biophys. 426, 3–10, 2004. 162. Carrotta, R., Manno, M., Giordano, F.M., et al., Protein stability modulated by a conformational effector: Effects of triluoroethanol on bovine serum albumin, Phys. Chem. Chem. Phys. 11, 4007–4018, 2009. 163. Craig, H.D., Eklund, J.D., and Seidler, N.W., Triluoroethano increases albumin’s susceptibility to chemical modiication, Arch. Biochem. Biophys. 480, 11–16, 2008. 164. Bhattacharya, A.A., Curry, S., and Franks, N.P., Binding of the general anesthetics propofol and halothane to human serum albumin. High resolution crystal structures, J. Biol. Chem. 275, 38731–38738, 2000. 165. Liu, R., Meng, Q., Xi, J., et al., Comparative binding character of two general anesthetics for sites on human serum albumin, Biochem. J. 380, 147–152, 2004. 166. Lund, M., Bjerrum, O.J., and Bjerrum, M.J., Structural heterogeneity of the binding sites of HSA for phenyl-groups and medium-chain fatty acids. Demonstration of equilibrium between different binding conformations, Eur. J. Biochem. 260, 470–476, 1999. 167. Bjerrum, O.J., Bjerrum, M.J., and Heegaard, N.H., Electrophoretic and chromatographic differentiation of two forms of albumin in equilibrium at neutral pH: New screening techniques for determination of ligand binding to albumin, Electrophoresis 16, 1401– 1407, 1995. 168. Gospodarek, A.M., Smatlak, M.E., O’Connell, J.P., and Fernandez, E.J., Protein stability and structure in HIC: Hydrogen exchange experiments and COREX calculations, Langmuir 27, 286–295, 2011. 169. Huang, B.X., Dass, C., and Kim, H.-Y., Probing conformational changes of human serum albumin due to unsaturated fatty acid binding by chemical cross-linking and mass spectrometry, Biochem. J. 387, 695–702, 2005. 170. Michnik, A., Michalik, K., and Drzazga, Z., Effect of UVC radiation on conformational restructuring of human serum albumin, J. Photochem. Photobiol. B Biol. 90, 170–178, 2008. 171. Kragh-Hansen, U., Donaldson, D., and Jensen, P.H., The glycan structure of albumin Redhill, a glycosylated variant of human serum albumin, Biochim. Biophys. Acta 1550, 20–26, 2001. 172. Iwao, Y., Hiraike, M., Kragh-Hansen, U., et al., Altered chain-length and glycosylation modify the pharmacokinetics of human serum albumin, Biochim. Biophys. Acta 1794, 634–641, 2009. 173. Sakavej, P., Spalteholz, H., and Arnhold, J., Modiication of amino acid residues in human serum albumin by myeloperoxidase, Free Radic. Biol. Med. 40, 516–525, 2006. 174. Walsh, C.T., Garneau-Tsodikova, S., and Gatto, G.J., Jr., Protein posttranslational modiications: The chemistry of proteome diversiications, Angew. Chem. Int. Ed. Engl. 44, 7342–7472, 2005.

Albumin

145

175. Aldini, G., Vistoli, G., Regazzoni, L., et al., Albumin is the main nucleophilic target of human plasma: A protective role against pro-atherogenic electrophilic reactive carbonyl species?, Chem. Res. Toxicol. 21, 824–835, 2008. 176. Sikora, M., Marczak, L., Twardowski, T., et al., Direct monitoring of albumin lysine-525 N-homocysteinylation in human serum by liquid chromatography/mass spectrometry, Anal. Biochem. 405, 132–134, 2010. 177. Garlick, R.L. and Mazer, J.S., The principal site of nonenzymatic glycosylation of human serum albumin in vivo, J. Biol. Chem. 258, 6142–6146, 1983. 178. Wa, C., Cerny, R.L., Clarke, W.A., and Hage, D.S., Characterization of glycation adducts on human serum albumin by matrix-assisted laser desorption/ionization time-of-light mass spectrometry, Clin. Chim. Acta 385, 48–60, 2007. 179. Barnaby, O.S., Wa, C., Cerny, R.L., et al., Quantitative analysis of glycation sites on human serum albumin using 16O/18O-labeling and matrix-assisted laser desorption/ ionization time-of-light mass spectrometry, Clin. Chim. Acta 411, 1102–1110, 2010. 180. Frolov, A. and Hoffman, R., Identiication and relative quantiication of speciic glycation sites in human serum albumin, Anal. Bioanal. Chem. 397, 2349–2356, 2010. 181. Frost, L., Chaudhry, M., Bell, T., et al., In vitro galactosylation of human serum albumin: Analysis of the putative galactosylation site by mass spectrometry, Anal. Biochem. 410, 248–256, 2011. 182. Heidland, A., Sebekova, K., and Schinzel, R., Advanced glycation end products and the progressive course of renal disease, Am. J. Kidney Dis. 38 (4 Suppl. 1), S100–S106, 2002. 183. Schalkwijk, C.G., Lieuw-a-Fa, M., van Hinsbergh, V.W., and Stehouwer, C.D., Pathophysiological role of Amadori-glycated proteins in diabetic microangiopathy, Semin. Vasc. Med. 2, 191–197, 2002. 184. Cohen, M.P., Intervention strategies to prevent pathogenetic effects of glycated albumin, Arch. Biochem. Biophys. 419, 25–30, 2003. 185. Lapolla, A., Fedele, D., Seraglia, R., and Traldi, P., The role of mass spectrometry in the study of non-enzymatic protein glycation in diabetes: An update, Mass Spectrom. Rev. 25, 775–797, 2006. 186. Koga, M. and Kasayama, S., Clinical impact of glycated albumin as another glycemic control marker, Endocr. J. 57, 751–762, 2010. 187. Bunn, H.F. and Higgins, P.J., Reaction of monosaccharides with proteins: Possible evolutionary signiicance, Science 213, 222–224, 1981. 188. Frost, L., Chaudhry, M., Bell, T., and Cohenford, M., In vitro galactation of human serum albumin: Analysis of the protein’s galactation sties by mass spectrometry, Anal. Biochem. 410, 248–256, 2011. 189. Frolov, A. and Hoffmann, R., Identiication and relative quantiication of speciic glycation sites in human serum albumin, Anal. Bioanal. Chem. 397, 2349–2356, 2010. 190. Barnaby, O.S., Cerny, R.L., Clarke, W., and Hage, D.S., Comparison of modiication sites formed on human serum albumin at various stages of glycation, Clin. Chim. Acta 412, 277–285, 2011. 191. Garlick, R.L. and Mazer, J.H., The principle site of nonenzymic glycosylation of human serum albumin in vitro, J. Biol. Chem. 258, 6142–6146, 1983. 192. Iberg, N. and Flückiger, R., Nonenzymatic glycosylation of albumin in vivo. Identiication of multiple glycosylation sites, J. Biol. Chem. 261, 13542–13545, 1986. 193. Goldfarb, A.R., Heterogeneity of amino groups in proteins. I. Human serum albumin, Biochemistry 5, 2574–2578, 1966. 194. Andersson, L.-O., Brandt, J., and Johansson, S., The use of trinitrobenzenesulfonic acid in studies on the binding of fatty acid anions to bovine serum albumin, Arch. Biochem. Biophys. 146, 428–440, 1971.

146

Biotechnology of Plasma Proteins

195. Kurono, Y., Kinetic study on rapid reaction of trinitrobenzene sulfonate with human serum albumin, J. Pharm. Sci. 70, 1297–1298, 1981. 196. Kurono, Y., Ichioka, K., and Ikeda, K., Kinetics of the rapid modiication of human serum albumin with trinitrobenzene sulfonate and localization of its site, J. Pharm. Sci. 72, 432–435, 1983. 197. Shaklai, N., Garlick, R.L., and Bunn, H.F., Nonenzymatic glycosylation of human serum albumin alters its conformation and function, J. Biol. Chem. 259, 3812–3819, 1984. 198. Vorum, H., Fisker, K., Otagiri, M., et al., Calcium ion binding to clinically relevant chemical modiications of human serum albumin, Clin. Chem. 41, 1654–1661, 1995. 199. Natajou, K., Watanabe, H., Kragh-Hansen, U., et al., The effect of glycation on the structure, function and biological fate of human serum albumin as revealed by recombinant mutants, Biochim. Biophys. Acta 1623, 88–97, 2003. 200. Rodiño-Janeiro, B.K., González-Peteiro, M., Uciedo-Somoza, R., et al., Glycated albumin, a precursor of advanced glycation end-products, up-regulates NADPH oxidase and enhances oxidative stress in human endothelial cells: Molecular correlate of diabetic vasculopathy, Diabetes Metab. Res. Rev. 26, 550–559, 2010. 201. Eble, A.S., Thorpe, S.R., and Baynes, J.W., Nonenzymatic glucosylation and glucosedependent cross-linking of protein, J. Biol. Chem. 258, 9406–9412, 1983. 202. Otagiri, M. and Chuang, V.T., Pharmaceutically important pre- and posttranslational modiications on human serum albumin, Biol. Pharm. Bull. 32, 527–534, 2009. 203. Raikova, O., Raikov, R., and Nudler, E., Catalysis of S-nitrosothiols formation by serum albumin: The mechanism and implication of vascular control, Proc. Natl. Acad. Sci. USA 99, 5913–5918, 2002. 204. Meucci, E., Mordente, A., and Martorana, G.E., Metal-catalyzed oxidation of human serum albumin: Conformational and functional changes. Implications of protein aging, J. Biol. Chem. 266, 4692–4699, 1991. 205. Mikkelsen, R.B. and Wardman, P., Biological chemistry of reactive oxygen and nitrogen and radiation-induced signal transduction mechanisms, Oncogene 22, 5734–5754, 2003. 206. Shen, Y.-C., Wang, Y.-H., Chou, Y.-C., et al., Dimemorfan protects rats against ischemic stroke through activation of sigma-1 receptor-mediated mechanisms by decreasing glutamate accumulation, J. Neurochem. 104, 558–572, 2008. 207. Wang, L., Azad, N., Kongkaneramit, L., et al., The Fas death signaling pathway connecting reactive species generation and FLICE inhibitory protein down-regulation, J. Immunol. 180, 3072–3080, 2008. 208. Hawkes, W.C. and Alkan, Z., Regulation of redox signaling by selenoproteins, Biol. Trace Elem. Res. 136, 235–251, 2010. 209. Van Der Vliet, A., Eiserich, J.P., Kaur, H., et al., Nitrotyrosine as biomarker for reactive nitrogen species, Meth. Enzymol. 269, 175–184, 1996. 210. Förstermann, U., Nitric oxide and oxidative stress in vascular disease, Plugers Arch. 459, 923–939, 2010. 211. Kawai, K., Hayashi, T., Matsuyama, Y., et al., Difference in redox status of serum and aqueous humor in senile cataract patients as monitored via the albumin thiol-redox state, Jpn. J. Ophthalmol. 54, 584–588, 2010. 212. Oettl, K. and Stauber, R.E., Physiological and pathological changes in the redox state of human serum albumin critically inluence its binding properties, Br. J. Pharmacol. 151, 580–590, 2007. 213. Guedes, S., Vitorino, R., Domingues, R., et al., Oxidation of bovine serum albumin: Identiication of oxidation products and structural modiications, Rapid Commun. Mass Spectrom. 23, 2307–2315, 2009. 214. Temple, A., Yen, T.Y., and Gronert, S., Identiication of speciic protein carbonylation sites in model oxidations of human serum albumin, J. Am. Soc. Mass Spectrom. 17, 1172–1180, 2006.

Albumin

147

215. Maisonneuve, E., Ducet, A., Khoueiry, P., et al., Rules governing selective protein carbonylation, PLoS One 4, e7269, 2009. 216. Bar-Or, D., Bar-Or, R., Rael, L.T., et al., Heterogeneity and oxidation status of commercial human albumin preparations in clinical use, Crit. Care. Med. 33, 1638–1641, 2005. 217. Bar-Or, D., Thomas, G.W., Bar-Or, R., et al., Commercial human albumin preparations for clinical use are immunosuppressive in vitro, Crit. Care Med. 34, 1707–1712, 2006. 218. Shimonkevitz, R., Thomas, G., Slone, D.S., et al., A diketopiperazine fragment of human serum albumin modulates T-lymphocyte cytokine production through rap1, J. Trauma 64, 35–41, 2008. 219. Bridges, K.R., Schmidt, G.J., Jensen, M., The acetylation of hemoglobin by aspirin. In vitro and in vivo, J. Clin. Invest. 56, 201–207, 1975. 220. Villanueva, G.B. and Allen, N., Acetylation of antithrombin III by aspirtin, Semin. Thromb. Hemost. 12, 213–215, 1986. 221. Upchurch, G.R., Jr., Ramdev, N., Walsh, M.T., and Loscalzo, J., Prothrombotic consequences of the oxidation of ibrinogen and their inhibition by aspirtin, J. Thromb. Thromobolysis 5, 9–14, 1998. 222. Macdonald, J.M., LeBlanc, D.A., Haas, A.L., and London, R.E., An NMR analysis of the reaction of ubiquitin with [acety-1-13C] aspirin, Biochem. Pharmacol. 57, 1233–1244, 1999. 223. Xu, A.S., Ohba, Y., Vida, L., et al., Aspirin acetylation of βLys-82 of human hemoglobin. NMR study of acetylated hemoglobin Tsurumai, Biochem. Pharmacol. 60, 917– 922, 2000. 224. Anotovic, A., Perneby, C., Ekman, C.J., et al., Marked increase of ibrin gel permeability with very low dose ASA treatment, Thromb. Res. 116, 509–517, 2005. 225. Alfonso, L.F., Srivenugopal, K.S., and Bhat, G.J., Does aspirin acetylate multiple cellular proteins?, Mol. Med. Rep. 2, 533–537, 2009. 226. Jung, S.B., Kim, C.S., Naqvi, A., et al., Histone deacetylase 3 antagonizes aspirinstimulated endothelial nitric oxide production by reversing aspirin-induced lysine acetylation of endothelial nitric oxide synthase, Circ. Res. 107, 877–887, 2010. 227. Hawkins, D., Pinckard, R.N., Crawford, I.P., and Farr, R.S., Structural changes in human serum albumin induced by ingestion of acetylsalicylic acid, J. Clin. Invest. 48, 536–542, 1969. 228. Rendell, M., Nierenberg, J., Brannan, C., et al., Inhibition of glycation of albumin and hemoglobin by acetylation in vitro and in vivo, J. Lab. Clin. Med. 108, 286–293, 1986. 229. Jabusch, J.R. and Deutsch, H.F., Localization of lysines acetylated in ubiquitin reacted with p-nitrophenyl acetate, Arch. Biochem. Biophys. 238, 170–177, 1985. 230. Lockridge, O., Xue, W., Gaydess, A., et al., Pseudo-esterase activity of human albumin: Slow turnover on tyrosine 411 and stable acetylation of 82 residues including 59 lysines, J. Biol. Chem. 283, 22582–22589, 2008. 231. Hawkins, D., Pinckard, R.N., and Farr, R.S., Acetylation of human serum albumin by acetylsalicylic acid, Science 160, 780–781, 1968. 232. Jacobsen, C., Chemical modiication of the high-afinity bilirubin-binding site of human-serum albumin, Eur. J. Biochem. 27, 513–519, 1972. 233. Walker, J.E., Lysine residues 199 of human serum albumin is modiied by acetylsalicylic acid, FEBS Lett. 66, 173–175, 1976. 234. Liyasova, M.S., Schopfer, L.M., and Lockridge, O., Reaction of human albumin with aspirin in vitro: Mass spectrometric identiication of acetylated lysines 190, 402, 519, and 545, Biochem. Pharmacol. 79, 784–791, 2010. 235. Bohney, J.P., Fonda, M.L., and Feldhoff, R.C., Identiication of Lys190 as the primary site for pyridoxal-5′-phosphate in human serum albumin, FEBS Lett. 298, 266–268, 1992. 236. Yurchak, A.M., Wicher, K., and Arbesman, C.E., Immunologic studies on aspirin. Clinical studies with aspiryl-protein conjugates, J. Allergy 46, 245–253, 1970.

148

Biotechnology of Plasma Proteins

237. Honma, K., Nakamura, M., and Ishikawa, Y., Acetylsalicylate-human serum albumin interaction as studied by NMR spectroscopy—Antigenicity-producing mechanism of acetylsalicylic acid, Mol. Immunol. 28, 107–113, 1991. 238. Jurkowski, W., Porebski, G., Obtulowicz, K., and Roterman, I., Serum albumin complexation of acetylsalicylic acid metabolism, Curr. Drug Metab. 10, 448–458, 2009. 239. Ali, M.A. and Routh, J.I., The protein binding of acetylsalicylic acid and salicylic acid, Clin. Chem. 15, 1027–1038, 1969. 240. Blanca, M., Perez, E., Garcia, J.J., et al., Angioedema and IgE antibodies to aspirin: A case report, Ann. Allergy 62, 295–298, 1989. 241. Jurkowski, W., Porebski, G., Obtulowicz, K., and Roterman, I., Serum albumin complexation of acetylsalicylic acid metabolites, Curr. Drug Metab. 10, 448–458, 2009. 242. Kashiba-Iwatsuki, M., Miyamoto, M., and Inoue, M., Effect of nitric oxide on the ligand-binding activity of albumin, Arch. Biochem. Biophys. 345, 237–242, 1997. 243. Ishima, Y., Akaike, T., Kragh-Hansen, U., et al., Effects of endogenous ligands on the biological role of human serum albumin in S-nitrosylation, Biochem. Biophys. Res. Commun. 364, 790–795, 2007. 244. Tsikas, D., Sandmann, D., and Frölich, J.C., Measurement of S-nitrosoalbumin by gas chromatography-mass spectrometry. III. Quantitative determination in human plasma after speciic conversion of the S-nitroso group to nitrite by cysteine and Cu2+ via intermediate formation of S-nitrosocysteine and nitric oxide, J. Chromatogr. B 772, 335–346, 2002. 245. Tsikas, D., Sandmann, J., Rossa, S., et al., Measurement of S-nitrosoalbumin by gas chromatography-mass spectrometry I. Preparation, puriication, isolation, characterization and metabolism of S-[15N]nitrosoalbumin in human blood in vitro, J. Chromatogr. B 726, 1–12, 1999. 246. Kelm, M. and Schrader, J., Control of coronary vascular tone by nitric oxide, Circ. Res. 66, 1561–1575, 1990. 247. Moshage, H., Kok, B., Hulzenga, J.R., and Jansen, P.L.M., Nitrite and nitrate determinations in plasma: A critical evaluation, Clin. Chem. 41, 892–896, 1995. 248. Hallström, S., Gasser, H., Neumayer, C., et al., S-Nitroso human serum albumin treatment reduces ischemia/reperfusion injury in skeletal muscle via nitric oxide release, Circulation 105, 3032–3038, 2002. 249. Al-Ani, B., Hewett, P.W., Ahmed, S., et al., The release of nitric oxide from S-nitrosothiols promotes angiogenesis, PLoS One 1, e25, 2006. 250. Biesalski, H.K. and McGregor, G.P., Antioxidant therapy in critical care—Is the microcirculation the primary target?, Crit. Care Med. 35(9 Suppl.), S577–S583, 2007. 251. Scorza, G., Pietraforte, D., and Minetti, M., Role of ascorbate and protein thiols in the release of nitric oxide from S-nitroso-albumin and S-nitroso-glutathione in human plasma, Free Radic. Biol. Med. 22, 633–642, 1997. 252. Ishima, Y., Hiroyama, S., Kragh-Hansen, U., et al., One-step preparation of S-nitrosylated human serum albumin with high biological activity, Nitric Oxide 23, 121–127, 2010. 253. Jourd’heuil, F.L., Lowery, A.M., Melton, E.M., et al., Redox-sensitivity and sitespeciicity of S- and N-denitrosylation in proteins, PLoS One 5(12), e14400, 2010. 254. Pryor, W.A. and Squadrito, G.L., The chemistry of peroxynitrite: A product from the reaction of nitric oxide with superoxide, Am. J. Physiol. 268, L699–L722, 1995. 255. Alvarez, B., Ferrer-Sueta, G., Freeman, B.A., and Radi, R., Kinetics of peroxynitrite reaction with amino acid and human serum albumin, J. Biol. Chem. 274, 842–848, 1999. 256. Carballal, S., Radi, R., Kirk, M.O., et al., Sulfenic acid formation in human serum albumin by hydrogen peroxide and peroxynitrite, Biochemistry 42, 9906–9914, 2003. 257. Yamakura, F. and Ikeda, K., Modiication of tryptophan an tryptophan residues in proteins by reactive nitrogen species, Nitric Oxide 14, 152–161, 2006.

Albumin

149

258. Malan, P.G. and Edelhoch, H., Nitration of human serum albumin and bovine and human goiter thyroglobulins with tetranitromethane, Biochemistry 9, 3205–3214, 1970. 259. Khan, J., Brennand, D.M., Bradley, N., et al., 3-Nitrotyrosine in the proteins of human plasma determined by an ELISA method, Biochem. J. 330, 795–801, 1998. 260. Gow, A., Duran, D., Thom, S.R., and Ischiropolous, H., Carbon dioxide enhancement of peroxynitrite-mediated protein tyrosine nitration, Arch. Biochem. Biophys. 33, 42–48, 1996. 261. Nikov, G., Bhat, V., Wishnok, J.S., and Tannenbaum, S.R., Analysis of nitrated proteins by nitrotyrosine-speciic afinity probes and mass spectrometry, Anal. Biochem. 320, 214–222, 2003. 262. Harrohalli, K., Petersen, C.E., Ha, C.E., et al., Site-directed mutgenesis studies of human serum albumin deine tryptophan at amino acid position 214 as the principal site for nitrosation, J. Biomed. Sci. 9, 47–58, 2002. 263. Sonnenschein, K., de Groot, H., and Kirsch, M., Formation of S-nitrosothiols from regiospeciic reaction of thiols with N-nitrosotryptophan derivatives, J. Biol. Chem. 279, 45433–45440, 2004. 264. Lee, J. and Blaber, M., Structural basis of conserved cysteine in the ibroblast growth factor family: Evidence for a vestigial half-cysteine, J. Mol. Biol. 393, 128–139, 2009. 265. Lee, J. and Blaber, M., The interaction between thermodynamic stability and buried free cysteines in regulating the functional half-life of ibroblast growth factor-1, J. Mol. Biol. 393, 113–127, 2009. 266. Zhao, H.L., Xue, C., Wang, Y., et al., Elimination of the free sulfhydryl group in the human serum (HSA) moiety of human interferon-α2b and HSA fusion protein increases its stability against mechanical and thermal stress, Eur. J. Pharm. Biopharm. 72, 405– 411, 2009. 267. Thornton, J.M., Disulphide bridges in globular proteins, J. Mol. Biol. 151, 261–287, 1981. 268. Fiser, A. and Simon, I., Predicting the redox states of cysteine in proteins, Meth. Enzymol. 353, 10–44, 2002. 269. Holmgren, A., The role of thioredoxin and glutaredoxin systems in disulide reduction and thiol redox control, in Cellular Implications of Redox Signaling, eds. C. Giller and A. Dannon Imperial College Press, London, UK, 2003. 270. Bar-Or, R., Rael, L.T., and Bar-Or, D., Dehydroalanine derived from cysteine is a common post-translational modiication in human serum albumin, Rapid Commun. Mass Spectrom. 22, 711–716, 2008. 271. Ikegaya, K., Nokihara, K., and Yasuhara, T., Characterization of sulfhydryl heterogeneity in human serum albumin and recombinant human serum albumin for clinical use, Biosci. Biotechnol. Biochem. 74, 2232–2236, 2010. 272. Ghiggeri, G.M., Candiano, G., Delino, G., and Queiroio, C., A modiication of the 5,5′-dithiobis(2-nitrobenzoic acid)(DTNB) method for the determination of the sulfhydryl content of human serum albumin, Clin. Chim. Acta 130, 257–261, 1983. 273. Aćimovic, J.M., Stanimirović, B.D., Todorović, N., et al., Inluence of the microenvironment of thiol groups in low molecular mass thiols and serum albumin on the reaction with methylglyoxal, Chem. Biol. Interact. 188, 21–30, 2010. 274. Hughes, W.L., Jr., An albumin fraction isolated from human plasma as a crystalline mercuric salt, J. Am. Chem. Soc. 69, 1836–1837, 1947. 275. Hughes, W.L., Jr. and Dintzis, H.M., Crystallization of the mercury dimer of human and bovine mercaptalbumin, J. Biol. Chem. 239, 845–849, 1964. 276. Carlsson, J. and Svenson, A., Preparation of bovine mercaptalbumin by means of covalent chromatography, FEBS Lett. 42, 183–186, 1974. 277. Funk, W.E., Li, H., Iavarone, A.T., et al., Enrichment of cysteinyl adducts of human serum albumin, Anal. Biochem. 400, 61–68, 2010.

150

Biotechnology of Plasma Proteins

278. Bunnett, J.F., Nucleophilic reactivity, Annu. Rev. Phys. Chem. 14, 271–290, 1963. 279. Thurkill, R.L., Grimsley, G.R., Schultz, J.M., and Pace, C.N., pK values for ionizable groups of proteins, Protein Sci. 15, 1214–1218, 2006. 280. Bruschi, M., Musante, L., Candiano, G., et al., Transitions of serum albumin in patients with glomerulosclerosis ‘in vivo’ characterization by electrophoretic titration curves, Electrophoresis 27, 2960–2969, 2006. 281. Spiga, O., Summa, D., Cirri, S., et al., A structurally driven analysis of thiol reactivity in mammalian albumins, Biopolymers 95, 278–285, 2011. 282. Edwards, F.B., Rombauer, R.B., and Campbell, B.J., Thiol-disulide interchange reactions between serum albumin and disulides, Biochim. Biophys. Acta 194, 234–245, 1969. 283. Narazaki, R., Hamada, M., Harada, K., and Otagiri, M., Covalent binding between bucillamine derivatives and human serum albumin, Pharm. Res. 13, 1317–1321, 1996. 284. Lewis, S.D., Misra, D.C., and Shafer, J.A., Determination of interactive thiol ionizations in bovine serum albumin, glutathione, and other thiols by potentiometric difference titration, Biochemistry 19, 6129–6137, 1980. 285. Ohkubo, A., Conformation around the sulfhydryl group in human serum albumin, J. Biochem. 65, 879–888, 1969. 286. Zhang, H., Le, M., and Means, G.E., A kinetic approach to characterize the electrostatic environments of thiol groups in proteins, Bioorg. Chem. 26, 356–364, 1998. 287. Baker, A., Santos, B.D., and Powis, G., Redox control of caspase-3 activity by thioredoxin and other reduced proteins, Biochem. Biophys. Res. Commun. 268, 78–81, 2000. 288. Cha, M.-K. and Kim, I.-H., Disulide between Cys392 and Cys438 of human serum albumin is redox-active, which is responsible for the thioredoxin-supported lipid peroxidase activity, Arch. Biochem. Biophys. 445, 19–25, 2006. 289. Edwards, F.B., Rombauer, R.B., and Campbell, B.J., Thiol-disulide interchange reactions between serum albumin and disulide, Biochim. Biophys. Acta 194, 234–245, 1969. 290. Pedersen, A.O. and Jacobsen, J., Reactivity of the thiol group in human and bovine albumin at pH 3–9, as measured by exchange with 2,2′-dithiopyridine, Eur. J. Biochem. 106, 291–295, 1980. 291. Svenson, A. and Carlsson, J., The thiol group of bovine serum albumin. High reactivity at acid pH as measured by the reaction with 2,2′-dithiopyridyl disulide, Biochim. Biophys. Acta 400, 433–438, 1975. 292. Liu, T.-Y., The role of sulfur in proteins, in The Proteins, 3rd edn., eds. H. Neurath and R.L. Hill, Vol. 3, Chapter 3, pp. 239–402, Academic Press, New York, 1977. 293. Kuwata, K., Era, S., and Sogami, M., The kinetic studies on the intramolecular SH, S–S exchange reaction of bovine mercaptalbumin, Biochim. Biophys. Acta 1205, 317–324, 1994. 294. Cha, M.-K. and Kim, I.-H., Disulide between C392 and Cys438 of human serum albumin is redox-active, which is responsible for the thioredoxin-supported lipid peroxidase activity, Arch. Biochem. Biophys. 445, 19–25, 2006. 295. Lindley, H. and Haylett, T., Disulphide interchange reactions involving cyclocystine and their relevance to problems of α-keratin structure, Biochem. J. 108, 701–703, 1968. 296. Tatsumi, E. and Hirose, M., Highly ordered molten globule-like state of ovalabumin at acidic pH: Native-like fragmentation by protease and selective modiication of cys367 with dithiopyridine, J. Biochem. 122, 300–308, 1997. 297. Tatsumi, E., Yoshimatsu, D., and Hirose, M., Conformational state of ovalabumin at acidic pH as evaluated by a novel approach utilizing intrachain sulfhydryl-mixed disulide exchange reactions, Biochemistry 37, 12351–12359, 1998. 298. Brocklehurst, K. and LIttle, G., Reactions of papain and of low-molecular weight thiols with some aromatic disulphides, Biochem. J. 133, 67–80, 1973.

Albumin

151

299. Takeda, K., Shigemura, A., Hamada, S., et al., Dependence of reaction rate of 5,5′-dithiobis-(2-nitrobenzoic acid) to free sulfhydryl groups of bovine serum albumin and ovalbumin on the protein conformation, J. Protein Chem. 11, 187–192, 1992. 300. Chamani, J., Conformation and stability of the α-helical intermediate of intact and thiolmodiied β-lactoglobulin induced by sodium dodecyl sulfate, Asian J. Chem. 19, 4203– 4213, 2007. 301. Zhang, H., Le, M., and Means, G.E., A kinetic approach to characterize the electrostatic environments of thiol groups in proteins, Bioorg. Chem. 26, 356–364, 1998. 302. Wynn, R. and Richards, F.M., Chemical modiication of protein thiols: Formation of mixed disulides, Meth. Enzymol. 251, 351–356, 1995. 303. Bradshaw. R.A., Kanarek, L., and Hill, R.L., The preparation, properties, and reactivation of the mixed disulide derivative of egg white lysozyme, J. Biol. Chem. 242, 3789–3798, 1967. 304. Kozlov, G., Määttänen, P., Thomas, D.Y., and Gehring, K., A structural overview of the PDI family of proteins, FEBS J. 277, 3924–3936, 2010. 305. Wang, Y.-H. and Narayan, M., pH dependence of the isomerase activity of protein disulide isomerase: Insights into its functional relevance, Protein J. 27, 181–185, 2008. 306. Sengupta, S., Chen, H., Togawa, T., et al., Albumin thiolate anion is an intermediate in the formation of albumin-S-S-homocysteine, J. Biol. Chem. 276, 30111–30117, 2001. 307. Zinellu, A., Lepedda, A., Jr., Sotigia, S., et al., Albumin-bound low molecular weight thiol analysis in plasma and carotid plaque by CE, J. Sep. Sci. 33, 126–133, 2010. 308. Hortin, G.L., Seam, N., and Hoehn, G.T., Bound homocysteine, cysteine, and cysteinylglycine distribution between albumin and globulins, Clin. Chem. 52, 2258–2264, 2006. 309. Beck, J.L., Ambahera, S., Yong, S.R., et al., Direct observation of covalent adducts with Cys34 of human serum albumin using mass spectrometry, Anal. Biochem. 325, 326– 336, 2004. 310. Berggård, T., Thelin, N., Falkenberg, C., et al., Prothrombin, albumin and immunoglobulin A form covalent complexes using α1-microglobulin in human plasma, Eur. J. Biochem. 245, 676–683, 1997. 311. Tejler, L. and Grubb, A.O., A complex-forming glycoprotein heterogeneous in charge and present in human plasma, urine, and cerebrospinal luid, Biochim. Biophys. Acta 439, 82–94, 1976. 312. Grubb, A.O., López, C., Tejler, L., et al., Isolation of human complex-forming glycoprotein, heterogeneous in charge (protein HC), and its IgA complex from plasma. Physicochemical and immunochemical properties, normal plasma concentration, J. Biol. Chem. 258, 14698–14707, 1983. 313. Wojcik, E.G.C., Simioni, P., Berg, M.V.D., et al., Mutations which introduce free cysteine residues in the Gla-domain of vitamin K-dependent proteins result in the formation of complexes with α1-microglobulin, Thromb. Haemost. 75, 70–75, 1996. 314. Mosher, D.F. and Johnson, R.B., In vitro formation of disulide-bonded ibronectin multimer, J. Biol. Chem.258, 6595–6601, 1983. 315. Fujihara, N., Tozuka, M., Yamauchi, K., et al., Characterization of factor XII Tenri, a rare CRM-negative factor XII deiciency, Ann. Clin. Lab. Sci. 34, 218–221, 2004. 316. McCann, K.B., Vucica, Y., Famulari, S., and Bertolini, J., Effect of processing methods on colouration of human serum albumin preparations, Biologicals 37, 32–36, 2009. 317. Anson, M.L., The sulfhydryl groups of egg albumin, J. Gen. Physiol. 24, 399–421, 1941. 318. Yamazoe, H., Yamauchi, K., and Tanabe, T., Preparation of S-sulfo albumin ilm and its cell adhesive properties, Mater. Sci. Eng. C. Mater. Biol. Appl. 29, 1105–1108, 2009. 319. Sarkar, N., Kumar, M., and Dubey, V.K., Effect of sodium tetrathionate on amyloid ibril: Insight into the role of disulide bond in amyloid progression, Biochimie 93, 962–968, 2011.

152

Biotechnology of Plasma Proteins

320. Happer, D.A.R., Mitchell, J.W., and Wright, G.J., Nucleophilic cleavage of diaryl disulide, Aust. J. Chem. 26, 121–134, 1973. 321. Klein, I.B. and Kirsch, J.F., The mechanism of the activation of papain, Biochem. Biophys. Res. Commun. 34, 575–581, 1969. 322. Fasco, M.J., Hauer III, C.R., Stack, R.E., et al., Cyanide adducts with human plasma proteins: Albumin as a potential exposure surrogate, Chem. Res. Toxicol. 20, 677–684, 2007. 323. Fasco, M.J., Strack, R.F., Lu, S., et al., Unique cyanide adduct in human serum albumin: Potential as a surrogate exposure marker, Chem. Res. Toxicol. 24, 505–514, 2011. 324. Tang, H.-Y. and Speicher, D.W., Identiication of alternative products and optimization of 2-nitro-5-thiocyanatobenzoic acid cyanylation and cleavage at cysteine residues, Anal. Biochem. 334. 48–61, 2004. 325. Walker, E., CLIV. A colour reaction for disulphides, Biochem. J. 19, 1085–1087, 1925. 326. Catsimpoolas, N. and Wood, J.L., The reaction of cyanide with bovine serum albumin, J. Biol. Chem. 239, 4132–4137, 1964. 327. Carballal, S., Radi, R., Kirk, M.C., et al., Sulfenic acid formation in human serum albumin by hydrogen peroxide and peroxynitrite, Biochemistry 42, 9906–9914, 2003. 328. Turell, L., Caballal, S., Botti, H., et al., Oxidation of the albumin thiol to sulfenic acid and its implications in the intravascular compartment, Braz. J. Med. Biol. Res. 42, 305– 311, 2009. 329. Turell, L., Botti, H., Carballal, S., et al., Reactivity of sulfenic acid in human serum albumin, Biochemistry 47, 358–367, 2008. 330. Priora, R., Coppo, L., Salzano, S., et al., Measurement of mixed disulides including glutathionylated proteins, Meth. Enzymol. 473, 149–159, 2010. 331. Jacob, C., Knight, I., and Winyard, P.G., Aspects of the simple redox response to sophisticated signalling pathways, Biol. Chem. 387, 1385–1397, 2006. 332. Rehder, D.S. and Borges, C.R., Possibilities and pitfalls in quantifying the extent of cysteine sulfenic acid modiication of speciic proteins within complex bioluids, BMC Biochem. 11, 25, 2010. 333. Alvarez, B., Carballal, S., Turell, L., and Radi, R., Formation and reactions of sulfenic acid in human serum albumin, Meth. Enzymol. 473, 117–136, 2010. 334. Suzukida, M., Le, H.P., Shahid, F., et al., Resonance energy transfer between cysteine-34 and tryptophan-214 in human serum albumin. Distance measurements as a function of pH, Biochemistry 22, 2415–2420, 1983. 335. Hagag, N., Birnbaum, E.R., and Darnell, D.W., Resonance energy transfer between cysteine-34, tryptophan-214, and tyrosine-411 of human serum albumin, Biochemistry 22, 2420–2427, 1983. 336. Krishnakumar, S.S. and Panda, D., Spatial relationship between the prodan site, Trp214, and Cys-34 residues in human serum albumin and loss of structure through incremental unfolding, Biochemistry 41, 7443–7452, 2002. 337. Tong, G.C., Cornwell, W.K., and Means, G.E., Reactions of acrylamide with glutathione and serum albumin, Toxicol. Lett. 147, 127–131, 2004. 338. Aldini, G., Gamberoni, L., Orioli, M., et al., Mass spectrometric characterization of covalent modiication of human serum albumin by 4-hydroxy-trans-2-nonenal, J. Mass Spectrom. 41, 1149–1161, 2006. 339. Colombo, G., Aldini, G., Orioli, M., et al., Water-soluble α,β-unsaturated aldehydes of cigarette smoke induce carbonylation of human serum albumin, Antioxid. Redox Signal. 12, 349–364, 2010. 340. Ishino, K., Wakita, C., Shibata, T., et al., Lipid peroxidation generates body odor component trans-2-nonenal covalently bound to protein in vivo, J. Biol. Chem. 285, 15302– 15313, 2010.

Albumin

153

341. Mera, K., Takeo, K., Izumi, M., et al., Effect of reactive-aldehydes on the modiication and dysfunction of human serum albumin, J. Pharm. Sci. 99, 1614–1625, 2010. 342. Wormall, A., The immunological speciicity of chemically altered proteins: Halogenated and nitrated proteins, J. Exp. Med. 51, 295–317, 1930. 343. Ehrenberg, L., Fischer, I., and Lofgren, N., Inhibitory effect of tetranitromethane on diphtheria toxin–antitoxin precipitin reaction, Nature 157, 730, 1946. 344. Sokolovsky, M., Riordan, J.F., and Vallee, B.L., Tetranitromethane. A reagent for the nitration of tyrosyl residues in proteins, Biochemistry 5, 3582–3589, 1966. 345. Lundblad, R.L., Chemical Reagents for Protein Modiication, 3rd edn., CRC Press, Boca Raton, FL, 2004. 346. Fehske, K.J., Müller, W.E., and Wollert, U., A highly reactive tyrosine residue as part of the indole and benzodiazepine binding site of human serum albumin, Biochim. Biophys. Acta 577, 346–359, 1979. 347. Fehske, K.J., Müller, W.E., and Wollert, U., Direct demonstration of the highly reactive tyrosine residue of human serum albumin located in fragment 299–585, Arch. Biochem. Biophys. 205, 217–221, 1990. 348. Kim, H.S., Austin, J., and Hage, D.S., Identiication of drug-binding sites on human serum albumin using afinity capillary electrophoresis and chemically modiied proteins as buffer additives, Electrophoresis 23, 956–963, 2002. 349. Šantrůček, J., Strohalm, M., Kadlčik, V., et al., Tyrosine residues modiication studied by MALDI-TOF mass spectrometry, Biochem. Biophys. Res. Commun. 323, 1151–1156, 2004. 350. Ding, S.-J., Carr, J., Carlson, J.E., et al., Five tyrosines and two serines in human albumin are labeled by the organophosphorous agent FP-biotin, Chem. Res. Toxicol. 21, 1787–1794, 2008. 351. Barron, E.S.G., Thiol groups of biological importance, Adv. Enzymol. 11, 201–266, 1951. 352. Webb, J.L., in Enzyme and Metabolic Inhibitors, Sulfhydryl Reagents, Vol. 2, Chapter 4, p. 643, Academic Press, New York, 1966. 353. Koithoff, I.M. and Tan, B.H., Reactivity of sulfhydryl and disulide in proteins, VI. Effect of heat denaturation of bovine serum albumin (BSA) on sulfhydryl and reactive disulide content, J. Am. Chem. Soc. 87, 2717–2720, 1965. 354. Stewart, A.J., Blindauer, C., Berezenko, S., et al., Role of Tyr84 in controlling the reactivity of Cys34 of human albumin, FEBS J. 272, 353–362, 2004. 355. Anraku, M., Takeuchi, K., Watanabe, H., et al., Quantitative analysis of cysteine-34 on the antioxidative properties of human serum albumin in hemodialysis patients, J. Pharm. Sci. 100, 3968–3976, 2011. 356. Wilson, J.M., Wu, D., Motiu-DeGrood, R., and Hope, D.J., A spectrophotometric method for studying the rate of reaction of disulides with protein thiol groups applied to bovine serum albumin, J. Am. Chem. Soc. 102, 359–363, 1980. 357. Gitler, C., Zarmi, B., and Kalep, E., Use of cationic detergents to enhance reactivity of protein sulfhydryls, Meth. Enzymol. 251, 366–375, 1995. 358. Stewart, A.J., Blindauer, C.A., Berenzo, S., et al., Role of Tyr84 in controlling the reactivity of Cys34 of human albumin, FEBS J. 272, 353–362, 2004. 359. Spiga, O., Summa, D., Cirri, S., et al., A structurally driven analysis of thiol reactivity in mammalian albumins, Biopolymers 95, 278–285, 2011. 360. Chaiken, I.M. and Smith, E.L., Reaction of chloroacetamide with the sulfhydryl group of papain, J. Biol. Chem. 244, 5087–5094, 1969. 361. Chaiken, I.M. and Smith, E.L., Reaction of the sulfhydryl group of papain with chloroacetic acid, J. Biol. Chem. 244, 5095–5099, 1969. 362. Gerwin, B.J., Properties of the single sulfhydryl group of streptococcal proteinase. A comparison of the rate of alkylation by chloroacetic acid and chloroacetamide, J. Biol. Chem. 242, 451–456, 1967.

154

Biotechnology of Plasma Proteins

363. Columbo, G., Aldini, G., Orioli, M., et al., Water-soluble-α,β-unsaturated aldehydes of cigarette smoke induce carbonylation of human serum albumin, Antioxid. Redox Signal. 12, 349–364, 2010. 364. Lindh, C.H., Kristiansson, M.H., Berg-Andersson, U., and Cohen, A.S., Characterization of adducts formed between human serum albumin and the butadiene metabolite epoxybutanediol, Rapid Commun. Mass Spectrom. 19, 2488–2496, 2005. 365. Damsten, M.C., Commandeur, J.N.M., Fidder, A., et al., Liquid chromatography/ tandem mass spectrometry detection of covalent binding of acetaminophen to human serum albumin, Drug Metab. Dispos. 35, 1408–1417, 2007. 366. Jansen, R.W., Molema, G., Pauwela, R., et al., Potent in vitro anti-human immunodeiciency virus-1 activity of modiied human serum albumins, Mol. Pharmacol. 39, 818–823, 1991. 367. Swart, P.J., Sun, C.S., Kuipers, M.E., et al., The in vitro anti-HIV eficacy of negatively charged human serum albumin is antagonized by heparin, AIDS Res. Hum. Retroviruses 13, 677–683, 1997. 368. Swart, P.J., Kuipers, M.E., Smit, C., et al., The metabolic fate of the anti-HIV active drug carrier succinylated serum albumin after intravenous administration in rats, J. Drug Target. 9, 95–109, 2001. 369. Proost, J.H., Beljaars, L., Olinga, P., et al., Prediction of the pharmacokinetics of succinylated human serum albumin in man from in vivo disposition data in animals and in vitro liver slice incubations, Eur. J. Pharm. Sci. 27, 123–132, 2006. 370. Vermeulen, J.N., Meijer, D.K., Over, J., et al., A phase I/IIa study with succinylated human serum albumin (suc-HSA), a candidate HIV-1 fusion inhibitor, Antivir. Ther. 12, 273–278, 2007. 371. Yamasaki, Y., Ikenaga, T., Otsuki, T., et al., Induction of antigen-speciic cytotoxic T lymphocytes by immunization with negatively charged soluble antigen though scavenger receptor-mediated delivery, Vaccine 25, 85–89, 2007. 372. Tokuda, H., Masuda, S., Takekura, Y., et al., Speciic uptake of succinylated proteins via a scavenger receptor-mediated mechanism in culture brain microvessel endothelial cells, Biochem. Biophys. Res. Commun. 196, 18–24, 1993. 373. Yamasaki, Y., Suimoto, K., Nishikawa, M., et al., Pharmacokinetic analysis of in vivo disposition of succinylated proteins targeted to liver nonparenchymal cells via scavenger receptors: Importance of molecular size and negative change density for in vivo recognition by receptors, J. Pharmcol. Exp. Ther. 301, 467–477, 2002. 374. Yamasaki, Y., Hisazumi, J., Yamaoka, K., and Takahura, Y., Eficient scavenger receptormediated hepatic targeting of proteins by introduction of negative charges on the proteins by aconitylation: The inluence of charge density and size of the protein molecules, Eur. J. Pharm. Sci. 18, 305–312, 2003. 375. Chang, T.S. and Sun, S.F., Structural studies on the succinylated bovine serum albumin, Int. J. Pept. Protein Res. 11, 65–72, 1978. 376. Mir, M.M., Fazili, K.M., and Abul Qasim, M., Chemical modiication of buried lysine residues of bovine serum albumin and its inluence on protein conformation and bilirubin binding, Biochim. Biophys. Acta 1119, 261–267, 1992. 377. Khan, M.M. and Tayyab, S., Understanding the role of internal lysine residues of serum albumins in conformational stability and bilirubin binding, Biochim. Biophys. Acta 1545, 263–277, 2001. 378. Giovanni, I., Chiarla, C., Giuliante, M., et al., The relationship between albumin, other plasma proteins and variables, and age in the acute phase response after liver resection in man, Amino Acids 31, 463–469, 2006. 379. Bennhold, H. and Kallee, E., Comparative studies on the half-life of iodine-131-labeled albumins and nonradioactive human serum albumin in a case of analbuminemia, J. Clin. Invest. 38, 863–872, 1959.

Albumin

155

380. Weisberg, H.F., Osmotic pressure of the serum proteins, Ann. Clin. Lab. Sci. 8, 155–164, 1978. 381. Dammaco, F., Miglietta, A., D’Addabbo, A., et al., Analbuminemia: Report of a case and review of the literature, Vox Sang. 39, 153–161, 1980. 382. Koot, B.G., Howen, R., Pot, D.J., and Nauta, J., Congenital analbuminemia: Biochemical and clinical implications. A case report and literature review, Eur. J. Pediatr. 163, 664– 670, 2004. 383. Gamsjäger, T., Brenner, L., Sitzwohl, C., and Weinstabl, C., Half-lives of albumin and cholinesterase in critically ill patients, Clin. Chem. Lab. Med. 46, 1140–1142, 2008. 384. Anraku, M., Kragh-Hansen, U., Kawai, K., et al., Validation of the chloramine-T induced oxidation of human serum albumin as a model for oxidative damage in vivo, Pharm. Res. 20, 684–692, 2003. 385. Iwao, Y., Anraku, M., Yamasaki, K., et al., Oxidation of Arg-410 promotes the elimination of human serum albumin, Biochim. Biophys. Acta 1764, 743–749, 2006. 386. Anhorn, C., Sheldon, S., Laschinger, C., and Naylor, D.H., Catabolic half-lives and antigenic relationships of native, altered and commercially prepared human albumins in rabbits, Vox Sang. 42, 233–242, 1982. 387. Gell, P.G.H., Harrington, C.R., and Rivers, R.P., The antigenic function of simple chemical compounds: Production of precipitins in rabbits, Br. J. Exp. Pathol. 27, 267–286, 1946. 388. Yoshimura, M. and Cinader, B., The effect of tolerance on the speciicity of antibody response: Antibody to oxazolonated albumin of animals tolerant to the protein carrier, J. Immunol. 97, 959–968, 1966. 389. Sung, C., Nardelli, B., LaFleur, D.W., et al., An IFN-β-albumin fusion protein that displays improved pharmacokinetic and pharmacodynamic properties in nonhuman primates, J. Interferon Cytokine Res. 23, 25–36, 2003. 390. Melder, R.J., Osborn, B.L., Riccobene, T., et al., Pharmacokinetics and in vitro and in vivo anti-tumor response of an interleukin-2-human serum albumin fusion protein in mice, Cancer Immunol. Immunother. 54, 535–547, 2005. 391. Müller, D., Karle, A., Meissburger, B., et al., Improved pharmacokinetics of recombinant bispeciic antibody molecules by fusion to human serum albumin, J. Biol. Chem. 282, 12650–12660, 2007. 392. Shefield, W.P., Gataiance, S., and Eltringham-Smith, L.J., Combined administration of barbourin—Albumin and hirudin—Albumin fusion proteins limits ibrin(ogen) deposition on the rabbit balloon-injured aorta, Thromb. Res. 119, 195–207, 2007. 393. Huang, Y.-J., Lundy, P.M., Lazaris, A., et al., Substantially improved pharmacokinetics by recombinant human butyrylcholinesterase by fusion to human serum albumin, BMC Biotechnol. 8, 50, 2008. 394. Metzner, H.J., Weimer, T., Kronthaler, U., et al., Genetic fusion to albumin improves the pharmacokinetic properties of factor IX, Thromb. Haemost. 102, 634–644, 2009. 395. Mueller, N., Schneider, B., Pizenmaier, K., and Wajant, H., Superior serum half-life of albumin tagged NF ligands, Biochem. Biophys. Res. Commun. 396, 793–799, 2010. 396. Andersen, J.T., Pehrsen, F., Tomachev, V., et al., Extending half-life by indirect targeting of the neonatal Fc receptor (FcRn) using a minimal albumin binding domain, J. Biol. Chem. 286, 5234–5241, 2011. 397. Kim, J., Hayton, W.L., Robinson, J.M., and Anderson, C.L., Kinetics of FcRn-mediated recycling of IgG and albumin in human: Pathophysiology and therapeutic implications using a simpliied mechanism-based model, Clin. Immunol. 122, 146–155, 2007. 398. Kenanova, V.E., Olafsen, T., Salazar, F.B., et al., Tuning the serum persistence of human serum albumin domain III: Diabody fusion proteins, Protein Eng. Des. Sel. 23, 789–798, 2010. 399. Andersen, J.T., Daba, M.B., and Sandlie, I., FcRn binding properties of an abnormal truncated analbuminemic albumin variant, Clin. Biochem. 43, 367–372, 2010.

156

Biotechnology of Plasma Proteins

400. Roopenian, D.C. and Akilesh, S., FcRn: The neonatal Fc receptor comes of age, Nat. Rev. Immunol. 7, 715–725, 2007. 401. Low, S.C. and Mezo, A.R., Inhibitors of the FcRn: IgG protein–protein interaction, AAPS J. 11, 432–434, 2009. 402. Tesar, D.B. and Björkman, P.J., An intracellular trafic jam: Fc receptor-mediated transport of immunoglobulin G, Curr. Opin. Struct. Biol. 20, 226–233, 2010. 403. McCurdy, T.R., Gataiance, S., Eltringham-Smith, H.J., and Shefield, W.P., A covalently linked recombinant albumin dimer is more rapidly cleared in vivo than are wild-type and mutant C34A albumin, Pharm. Res. 23, 882–891, 2006. 404. Werle, M. and Bernkop-Schnürch, A., Strategies to improve plasma half life time of peptide and protein drugs, Amino Acids 30, 351–367, 2006. 405. Yang, B.B. and Kido, A., Pharmacokinetics and pharmacodynamics of pegilgrastim, Clin. Pharmacokinet. 50, 295–306, 2011. 406. Chuang, V.T.G., Kragh-Hansen, U., and Otagiri, M., Pharmaceutical strategies utilizing recombinant human serum albumin, Pharm. Res. 19, 569–577, 2002. 407. Kratz, F., Abu Hjaj, K., and Warnecke, A., Anticancer carrier-linked prodrugs in clinical trials, Expert Opin. Investig. Drugs 16, 1037–1056, 2007. 408. Christen, S., Catlin, I., Knight, I., et al., Plasma S-nitrosylated status in neonatal calves: Antigenic association with tissue-speciic S-nitrosylation and nitric oxide synthase, Exp. Biol. Med. 232, 309–322, 2002. 409. Ishima, Y., Sawa, T., Kragh-Hansen, U., et al., S-Nitrosylation of human variant albumin Liprizzi (R410C) confers potent antibacterial and cytoprotective properties, J. Pharmacol. Exp. Ther. 320, 969–977, 2007. 410. Hirato, K., Maruyama, T., Watanabe, H., et al., Genetically engineered mannosylated human serum albumin as a versatile carrier for liver-selective therapeutics, J. Control. Release 145, 9–16, 2010. 411. Fiume, L., Busi, C., DiStefano, G., and Mattioli, A., Coupling of antiviral nucleoside analogs to lactosaminated human albumin by using the imidazolides of their phosphoric esters, Anal. Biochem. 212, 407–411, 1993. 412. Di Stefano, G., Busi, C., and Fiume, L., Floxuridine coupling with lactosaminated human albumin to increase eficiency on liver micrometastases, Dig. Liver Dis. 34, 439–446, 2002. 413. Di Stefano, G., Tubaro, M., Lanza, M., et al., Synthesis and physicochemical characteristics of a liver-targeted conjugates of luorodeoxyuridine monophosphate with lactosaminated human albumin, Rapid Commun. Mass Spectrom. 17, 2503–2507, 2003. 414. Jeong, J.M., Hong, M.Y., and Lee, J., 99mTc-Neoglycosylated human serum albumin for imaging the hepatic asialoglycoprotein receptor, Bioconjug. Chem. 15, 850–855, 2004. 415. Stadalnik, R.C. and Vera, D.R., The evolution of 99mTc-NGA as a clinically useful receptor-binding radiopharmaceutical, Nucl. Med. Biol. 28, 499–503, 2001. 416. Lee, J.Y. and Hirose, M., Partially folded state of the disulide-reduced form of human serum albumin as an intermediate for reversible denaturation, J. Biol. Chem. 267, 14753–14758, 1992. 417. Kim, J.G., Baggio, L.L., Bridan, D.P., et al., Development and characterization of a glucagon-like peptide 1-albumin conjugate. The ability to activate the glucagon-like peptide 1 receptor in vivo, Diabetes 52, 751–759, 2003. 418. Warnecke, A., Fichtner, I., Garmenn, D., et al., Synthesis and biological activity of water-soluble maleimide derivative of the anticancer drug carboplatin designed as albumin-binding prodrugs, Bioconjug. Chem. 15, 1349–1359, 2004. 419. Schechter, Y., Mironcheik, M., Rubinraut, S., et al., Albumin-insulin conjugate releasing insulin under physiological conditions: A new concept for long-acting insulin, Bioconjug. Chem. 16, 913–920, 2005.

Albumin

157

420. Thiabaudeau, K., Léger, R., Huang, X., et al., Synthesis and evaluation of insulin– human serum albumin conjugates, Bioconjug. Chem. 16, 1000–1008, 2005. 421. Kratz, F., Albumin as a drug carrier: Design of prodrugs, drug conjugates, and nanoparticles, J. Control. Release 132, 171–183, 2008. 422. Sasson, K., Marcus, Y., Lev-Goldman, V., et al., Engineered prolonged-acting prodrugs employing an albumin-binding probe than undergoes slow hydrolysis at physiologic conditions, J. Control. Release 142, 214–220, 2010. 423. Narazaki, R., Hamada, M., Harada, K., and Otagiri, M., Covalent binding between bucillamine derivatives and human serum albumin, Pharm. Res. 13, 1317–1321, 1996. 424. Kragh-Hansen, U., Chuang, V.T.C., and Otagiri, M., Practical aspects of the ligandbinding and enzymatic properties of human serum albumin, Biol. Pharm. Bull. 25, 695–704, 2002. 425. Summa, D., Spiga, O., Bernini, A., et al., Protein-thiol substitution of protein dethiolation by thiol/disulide exchange reactions: The albumin model, Proteins Struct. Funct. Bioinform. 69, 369–378, 2007. 426. Narazaki, R., Harada, K., Sugil, A., and Otagiri, M., Kinetic analysis of the covalent binding of captopril to human serum albumin, J. Pharm. Sci. 86, 215–219, 1997. 427. Yeung, J.H.K., Breckenridge, A.M., and Park, B.K., Drug–protein conjugates-IV. The effect of acute renal failure on the disposition of [14C]captopril in the rat, Biochem. Pharmacol. 32, 2467–2472, 1983. 428. Narazaki, R., Watanabe, H., Maruyama, T., et al., An immunological method for the detection of captopril–protein conjugate, Arch. Toxicol. 72, 203–206, 1998. 429. Glowacki, R. and Jakubowski, H., Cross-talk between Cys34 and lysine residues in human serum albumin revealed by N-homocysteinylation, J. Biol. Chem. 279, 10864– 10871, 2004. 430. Lin, S.Y., Wei, K.S., Li, M.J., and Wang, S.L., Effect of ethanol or/and captopril on the secondary structure of human serum albumin before and after protein binding, Eur. J. Pharm. Biopharm. 57, 457–464, 2004. 431. Maries, A.D. and Al-Shabanah, O., Protective ability and binding afinity of captopril towards serum albumin in an in vitro glycation model of diabetes mellitus, J. Pharm. Biomed. Anal. 41, 571–575, 2006. 432. Wu, Y.-L., Huang, J., Xu, J., et al., Addition of a cysteine to glucagon-like peptide-1 (GLP-1) conjugates GLP-1 to albumin in serum and prolongs GLP-1 action in vivo, Regul. Pept. 164, 83–89, 2010. 433. Houen, G. and Jensen, O.M., Conjugation to preactivated proteins using divinylsulfone and iodoacetic acid, J. Immunol. Meth. 181, 187–200, 1995. 434. Gabor, F., Schwarzbauer, A., and Wirth, M., Lectin-mediated drug delivery: Binding and uptake of BSA-WGA conjugates using the CaCo-2 model, Int. J. Pharm. 237, 227–239, 2002. 435. Houen, G., Olsen, D.T., Hansen, P.R., et al., Preparation of bioconjugates by solid-phase conjugation to ion exchange matrix-adsorbed proteins, Bioconjug. Chem. 14, 75–79, 2003. 436. Kubler-Kielb, J., Liu, T.Y., Mocca, C., et al., Additional conjugation methods and immunogenicity of Bacillus anthracis poly-γ-glutamic acid–protein conjugates, Infect. Immun. 74, 4744–4749, 2006. 437. Blasco, H., Lalmanach, G., Godat, E., et al., Evaluation of a peptide ELISA for the detection of rituximab in serum, J. Immunol. Meth. 325, 127–139, 2007. 438. Phillips, J.A., Morgan, E.L., Dong, Y., et al., Single-step conjugation of bioactive peptides to proteins via a self-contained succinimidyl bis-arylhydrazone, Bioconjug. Chem. 20, 1950–1957, 2009. 439. Li, G., Rodriguez, L.G., Fransworth, D.F., and Gildersleeve, J.C., Effects of hapten density on the induced antibody repertoire, Chem. Bio. Chem. 11, 1686–1691, 2010.

158

Biotechnology of Plasma Proteins

440. Mera, K., Nagai, M., Brock, J.W., et al., Glutaraldehyde is an effective cross-linker for production of antibodies against advanced glycation end-products, J. Immunol. Meth. 334, 82–90, 2008. 441. Thierse, H.J., Gamerdinger, K., Junkes, C., et al., T cell receptor (TCR) interaction with haptens: Metal ions as non-classical haptens, Toxicology 209, 101–107, 2005. 442. Liang, Y., Liu, X.Y., Liu, Y., et al., Synthesis of three haptens for the class-speciic immunoassay for O,O-dimethyl organophosphorous pesticides and effect of hapten heterology on immunoassay sensitivity, Anal. Chim. Anal. 615, 174–183, 2008. 443. Shinkaruk, S., Lamothe, V., Scmitter, J.M., et al., Synthesis of haptens and conjugates for ELISA of glycitein: Development and validation of an immunological test, J. Agric. Food Chem. 56, 6809–6817, 2008. 444. Callan, H.E., Jenkins, R.E., Maggs, J.L., et al., Multiple adduction reactions of nitroso sulfamethoxazole with cysteinyl residues of peptides and proteins: Implications for hapten formation, Chem. Res. Toxicol. 22, 937–948, 2009. 445. Fodey, T.L., Greer, N.M., and Crooks, S.R., Antibody production: Low dose immunogen vs. low incorporation hapten using salmeterol as a model, Anal. Chim. Acta 637, 328–332, 2009. 446. Jenkinson, C., Jenkins, R.E., Aleksic, M., et al., Characterization of p-phenylenediamine–albumin binding sites and T-cell responses to hapten-modiied protein, J. Invest. Dermatol. 130, 732–742, 2010. 447. Lee, T.K., Sokoloski, T.D., and Royer, G.P., Serum albumin beads: An injectable, biodegradable system for the sustained release of drugs, Science 213, 233–235, 1981. 448. Newman, J.F., Serum albumin beads possessing slow-release properties for vaccines, Adv. Biotechnol. Processes 14, 129–146, 1990. 449. Chuang, V.T., Kragh-Hansen, U., and Otagiri, M., Pharmaceutical strategies utilizing recombinant human serum albumin, Pharm. Res. 19, 569–577, 2002. 450. Johansen, P., Merkle, H.P., and Gander, B., Technological considerations related to the up-scaling of protein microencapsulation by spray-drying, Eur. J. Pharm. Biopharm. 50, 413–417, 2000. 451. Bos, G.W., Sharenborg, M.M., Post, A.A., et al., Proliferation of endothelial cells on surface-immobilized albumin–heparin conjugate loaded with basic ibroblast growth factor, J. Biomed. Mater. Res. 44. 330–340, 1999. 452. MacAdam, A.B., Shai, Z.B., Marriott, G.P., and James, S.L., Anti-mucous polyclonal antibody production, puriication and linkage to the surface of albumin microspheres, Int. J. Pharm. 195, 147–158, 2000. 453. Toublan, F.J., Boppart, S., and Suslick, K.S., Tumor targeting by surface-modiied protein microspheres, J. Am. Chem. Soc. 128, 3472–3473, 2007. 454. Wunderlich, G., Drews, A., and Kotzerke, J., A kit for labeling of [188Re] human serum albumin microspheres for therapeutic use in nuclear medicine, Appl. Radiat. Isot. 62, 915–918, 2005. 455. Schiller, E., Bergmann, R., Pietzsch, J., et al., Yttrium-86-labelled human serum albumin microspheres: Relation of surface structure with in vivo stability, Nucl. Med. Biol. 35, 227–232, 2008. 456. Callewaert, M., Laurent-Maquin, D., and Edwards-Lévy, F., Albumin–alginate-coated microspheres: Resistance to steam sterilization and to lyophilization, Int. J. Pharm. 344, 161–164, 2007. 457. Callewaert, M., Millot, J.M., Lesage, J., et al., Serum albumin–alginate coated microspheres: Role of the inner gel in binding and release of the KRFK peptide, Int. J. Pharm. 366, 103–110, 2008. 458. Marszall, M.P. and Bucinski, A., A protein-coated magnetic beads as a tool for the rapid drug–protein binding study, J. Pharm. Biomed. Anal. 52, 420–424, 2010.

Albumin

159

459. Wang, J., Wang, X., Ren, L., et al., Conjugation of biomolecules with magnetic protein microspheres for the assay of early biomarkers associated with acute myocardial infarction, Anal. Chem. 81, 6210–6217, 2009. 460. Quinlan, G.J., Martin, G.S., and Evans, T.W., Albumins: Biochemical properties and therapeutic potential, Hepatology 41, 1211–1219, 2005. 461. Sudlow, G. Birkett, D.J., and Wade, D.N., The characterization of two speciic drug binding sites of human serum albumin, Mol. Pharmacol. 11, 824–832, 1975. 462. Sudlow, G., Birkett, D.J., and Wade, D.N., Further characterization of speciic drug binding sites on human serum albumin, Mol. Pharmacol. 12, 1052–1061, 1976. 463. Joseph, K.S., Moser, A.C., Basiaga, S.B., et al., Evaluation of alternatives to warfarin as probes for Sudlow site I of human serum albumin: Characterization by highperformance afinity chromatography, J. Chromatogr. A 1216, 3492–3500, 2009. 464. Karlsson, B.C., Rosengren, A.M., Näsland, I., et al., Synthetic human serum albumin Sudlow I binding site mimics, J. Med. Chem. 53, 7932–7937, 2010. 465. Abou-Zied, O.K. and Al-Lawatia, N., Exploring the drug-binding site Sudlow I of human serum albumin: The role of water and Trp214 in molecular recognition and ligand binding, Chem. Phys. Chem. 12, 270–274, 2011. 466. Lu, J., Stewart, A.J., Sadler, P.J., et al., Albumin as a zinc carrier: Properties of its highafinity zinc-binding sites, Biochem. Soc. Trans. 36, 1317–1321, 2008. 467. Peyre, V., Lair, V., André, V., et al., Detergent binding as a sensor of hydrophobicity and polar interactions in the binding cavities of proteins, Langmuir 21, 8865–8875, 2005. 468. Mandula, H., Parepally, J.M., Feng, R., and Smith, Q.R., Role of site-speciic binding to plasma albumin in drug availability to brain, J. Pharmacol. Exp. Ther. 317, 667–675, 2006. 469. Quevedo, M.A., Ribone, S.R., Moroni, G.N., and Briñón, M.C., Binding to human serum albumin of zidovudine (AZT) and novel AZT derivatives. Experimental and theoretical analyses, Bioorg. Med. Chem. 16, 2779–2790, 2008. 470. Zhu, L., Yang, F., Chen, L., et al., A new drug binding subsite on human serum albumin and drug–drug interaction studied by X-ray crystallography, J. Struct. Biol. 162, 40–49, 2008. 471. Guo, S., Shi, X., Yang, F., et al., Structural basis of transport of lysophophospholipids by human serum albumin, Biochem. J. 423, 23–30, 2009. 472. Curry, S., Brick, P., and Franks, N.P., Fatty acid binding to human serum albumin: New insights from crystallographic analysis, Biochem. Biophys. Acta 1441, 131–140, 1999. 473. Fujiwara, S. and Amisaki, T., Identiication of high afinity fatty acid binding sites on human serum albumin by MM-PHSA method, Biophys. J. 94, 95–103, 2008. 474. Hamilton, J.A., Fatty acid interactions with proteins: What X-ray crystal and NMR solution structures tell us, Prog. Lipid Res. 43, 177–199, 2004. 475. Hamilton, J.A., How fatty acids bind to proteins: The inside story from protein structures, Prostaglandins Leukot. Essent. Fatty Acids 67, 65–72, 2002. 476. Morrisett, J.D., Pownall, H.J., and Gotto, A.M., Jr., Bovine serum albumin. Study of the fatty acid and steroid binding sites using spin-labeled lipids, J. Biol. Chem. 250, 2487–2494, 1975. 477. Aguanno, J.J. and Ladenson, J.H., Inluence of binding and conformation studies and evidence for distinct differences between unsaturated and saturated fatty acids, J. Biol. Chem. 257, 8745–8748, 1982. 478. Dröge, J.H., Janssen, L.H., and Wilting, J., Evidence for the fatty acid-induced heterogeneity of the N and B conformations of human serum albumin, Biochem. Pharmacol. 34, 3299–3304, 1985. 479. Oida, T., 1H-NMR study on the interactions of human serum albumin with free fatty acid, J. Biochem. 100, 1533–1542, 1986.

160

Biotechnology of Plasma Proteins

480. Narazaki, R., Maruyama, T., and Otagiri, M., Probing the cysteine 34 residue in human serum albumin using luorescence techniques, Biochim. Biophys. Acta 1338, 275–281, 1997. 481. Kawahara, K., Kuniyasu, A., Masuda, K., et al., Eficient identiication of photolabelled amino acid residues by combining immunoafinity puriication with MS: Revealing the semotiadil-binding site and its relevance to binding sites for myristates in domain III of human serum albumin, Biochem. J. 363, 223–232, 2002. 482. Fanali, G., Fesce, R., Agrati, C., et al., Allosteric modulation of myristate and Mn(III) heme binding to human serum albumin. Optical and NMR spectroscopy characterization, FEBS J. 272, 4672–4683, 2005. 483. Suji, G., Khedkar, S.A., Singh, S.K., et al., Binding of lipoic acid induces conformational change and appearance of a new binding site in methylglyoxal modiied serum albumin, Protein J. 27, 205–214, 2008. 484. Huang, B.X., Dass, C., and Kim, H.Y., Proving conformational changes of human serum albumin due to unsaturated fatty binding by chemical cross-linking and mass spectrometry, Biochem. J. 387, 695–702, 2005. 485. Petitpas, I., Bhattacharya, A.A., Twine, S., et al., Crystal structure analysis of warfarin binding to human serum albumin. Anatomy of drug site I, J. Biol. Chem. 276, 22804– 22809, 2001. 486. Panjehshahin, M.R., Bowmer, C.J., and Yates, M.S., Effect of valproic acid, its unsaturated metabolites and some structurally related fatty acids on the binding of warfarin and dansylsarcosine to human albumin, Biochem. Pharmacol. 41, 1227–1233, 1991. 487. Gryzunov, Y.A., Arroyo, A., Vigne, J.L., et al., Binding of fatty acids facilitates oxidation of cysteine-34 and converts copper–albumin complexes from antioxidants to prooxidants, Arch. Biochem. Biophys. 413, 53–66, 2003. 488. Ito, S. and Yamamoto, D., Identiication of two bromocresol purple binding sites on human serum albumin, Clin. Chim. Acta 411, 1536–1538, 2010. 489. Fehske, K.J., Müller, W.E., Wollert, U., et al., The lone tryptophan residue of human serum albumin as part of the speciic warfarin binding site. Binding of dicoumarol to the warfarin, indole, and benzodiazepine binding site, Mol. Pharmacol. 16, 778–789, 1979. 490. Fehske, K.J., Schläfer, U., Wollert, U., and Müller, W.E., Characterization of an important drug binding area on human serum albumin including the high-afinity binding sites of warfarin and azopropazone, Mol. Pharmacol. 21, 387–393, 1982. 491. Kasai-Morita, S., Horie, T., and Awazu, S., Inluence of the N-B transition of human serum albumin on the structure of the warfarin-binding site, Biochim. Biophys. Acta 915, 277–283, 1987. 492. Domenici, E., Bertucci, C., Salvadori, P., and Wainer, I.W., Use of a human serum albumin-based high-performance liquid chromatography chiral stationaryphase for the investigation of protein binding: Detection of the allosteric interaction between warfarin and benzodiazepaine binding sites, J. Pharm. Sci. 80, 164–166, 1991. 493. Moreno, F., Cortijo, M., and González-Jiménez, J., The lourescent probe prodan characterizes the warfarin binding sites on human serum albumin, Photochem. Photobiol. 69, 8–15, 1999. 494. Dockal, M., Chang, M., Carter, D.C., and Rüker, F., Five recombinant fragments of human serum albumin-tools for the characterization of the warfarin binding site, Protein Sci. 9, 1455–1465, 2000. 495. Petersen, C.E., Ha, C.E., Curry, S., and Bhagavan, N.V., Probing the structure of the warfarin-binding site on human serum albumin using site-directed mutagenesis, Proteins 47, 116–125, 2002. 496. Shetty, H.G., Fennerty, A.G., and Routledge, P.A., Clinical pharmacokinetic considerations in the control of oral anticoagulant therapy, Clin. Pharmacokinet. 16, 238–253, 1989. 497. Palareti, G. and Legnani, C., Warfarin withdrawal. Pharmacokinetic–pharmacodynamic considerations, Clin. Pharmacokinet. 30, 300–313, 1996.

Albumin

161

498. Dekhuijzen, P.N. and Koopmans, P.P., Pharmacokinetic proile of zairlukast, Clin. Pharmacokinet. 41, 105–114, 2002. 499. Hornsby, L.B., Hester, E.K., and Donaldson, A.R., Potential interaction between warfarin and high dietary protein intake, Pharmacotherapy 28, 536–539, 2008. 500. Roosdorp, N., Wänn, B., and Sjöholm, I., Correlation between arginyl residue modiication and benzodiazepine binding to human serum albumin, J. Biol. Chem. 252, 3876–3880, 1977. 501. Fehske, K.J. and Müller, W.E., Optical studies on interaction of biliary contrast agents with native and modiied human serum albumin, J. Pharm. Sci. 70, 549–554, 1981. 502. Maruyama, K., Nishigori, H., and Iwatsuru, M., Characterization of the benzodiazepine binding site (diazepam site) on the human serum albumin, Chem. Pharm. Bull. (Tokyo) 33, 5002–5012, 1985. 503. Dale, O., The interaction of enlurane, halothane and the halothane metabolite triluoroacetic acid with the binding of acidic drugs to human serum albumin. An in vitro study, Biochem. Pharmacol. 35, 557–561, 1986. 504. Fehske, K.J. and Müller, W.E., High-afinity binding of ethacrynic acid is mediated by the two most important drug binding sites of human serum albumin, Pharmacology 32, 208–213, 1986. 505. Kragh-Hansen, U., Octanoate binding to the indole- and benzodiazepine-binding region of human serum albumin, Biochem. J. 273, 641–644, 1991. 506. Takamura, N., Rahman, M.N., Yamasaki, K., et al., Interaction of benzothiadiazides with human serum albumin studies by dialysis and spectroscopic methods, Pharm. Res. 11, 1452–1457, 1994. 507. Pistolozzi, M. and Bertucci, C., Species-dependent stereoselective drug binding to albumin: A circular dichroism study, Chirality 20, 552–558, 2008. 508. Kimura, T., Nakanishi, K., Nakagawa, T., et al., High-performance frontal analysis of the binding of thyroxine enantiomers to human serum albumin, Pharm. Res. 22, 667–675, 2005. 509. Pettersson, C., Arvidsson, T., Karlsson, A.L., and Merle, I., Chromatographic resolution of enantiomers using albumin as complexing agent in the mobile phase, J. Pharm. Biomed. Anal. 4, 221–235, 1986. 510. Zandomeneghi, M. and Cavazza, M., Chiral recognition by biological macromolecules. Partial resolution of racemic enones by albumin, J. Chromatogr. 464, 289–295, 1989. 511. Andersson, S., Thompson, R.A., and Allenmark, S.G., Direct liquid chromatographic separation of enantiomers on immobilized protein stationary phases. IX. Inluence of the cross-linking reagent on the retentative and enantioselective properties of chiral sorbents based on bovine serum albumin, J. Chromatogr. 591, 65–73, 1992. 512. Chosson, E., Uzan, S., Gimenex, F., et al., Inluence of speciic albumin ligand markers used as modiiers on the separation of benzodiazepine enantiomers by chiral liquid chromatography on a human serum albumin column, Chirality 5, 71–77, 1993. 513. Yang, J. and Hage, D.S., Characterization of the binding and chiral separation of Dand L-tryptophan on a high-performance immobilized human serum albumin column, J. Chromatogr. 645, 241–250, 1993. 514. Yang, J. and Hage, D.S., Role of binding capacity versus binding strength in the separation of chiral compounds on protein-based high-performance liquid chromatography columns. Interactions of D- and L-tryptophan with human serum albumin, J. Chromatogr. A 725, 273–285, 1996. 515. Nakamura, M., Kiyohara, S., Saito, K., et al., Chirla separation of DL-tryptophan using porous membranes containing multilayered bovine serum albumin crosslinked with glutaraldehyde, J. Chromatogr. A 822, 53–58, 1998. 516. Haginaka, J., Enantiomer separation of drugs by capillary electrophoresis using proteins as chiral selectors, J. Chromatogr. A 875, 235–254, 2000.

162

Biotechnology of Plasma Proteins

517. Bertucci, C., Bartolini, M., Gotti, R., and Andrisano, V., Drug afinity to immobilized target bio-polymers by high-performance liquid chromatography and capillary electrophoresis, J. Chromatogr. B 797, 111–129, 2003. 518. Yamasaki, K., Maruyama, T., Kragh-Hansen, U., and Otagiri, M., Characterization of site I on human serum albumin: Concept about the structure of a drug binding site, Biochim. Biophys. Acta 1295, 147–157, 1996. 519. Sjöholm, I., Ekman, B., Kober, A., et al., Binding of drugs to human serum albumin XI. The speciicity of three binding sites as studied with albumin immobilized in microparticles, Mol. Pharmacol. 16, 767–777, 1979. 520. Ekman, B. and Sjöholm, I., Improved stability of proteins immobilized by a modiied emulsion polymerization technique, J. Pharm. Sci. 67, 693–696, 1978. 521. Tillement, J.P., Zini, R., Lecomte, M., and d’Athis, P., Binding of digitoxin, digoxin and gitoxin to human serum albumin, Eur. J. Drug Metab. Pharmacokinet. 5, 129–134, 1980. 522. Brørs, O., Fremstad, D., and Poulsson, C., The afinity of human serum albumin for [3H]-digitoxin is dependent on albumin concentration, Pharmacol. Toxicol. 72, 310–313, 1993. 523. Hage, D.S. and Sengupta, A., Characterization of the binding of digitoxin and acetyldigitoxin to human serum albumin by high-performance afinity chromatography, J. Chromatogr. B Biomed. Sci. Appl. 724, 91–100, 1999. 524. Yamasaki, K., Maruyama, T., Takadate, A., et al., Characterization of site I of human serum albumin using spectroscopic analyses: Locational relations between regions Ib and Ic of site I, J. Pharm. Sci. 93, 3004–3012, 2004. 525. Yamasaki, K., Maruyama, T., Yoshimoto, K., et al., Interactive binding to the two principle ligand binding sites of human serum albumin: Effect of the neutral-to-base transition, Biochim. Biophys. Acta 1432, 313–323, 1999. 526. Beaven, G.H., Chen, S.H., d’Albis, A., and Gratzer, W.H., A spectroscopic study of the haemin—Human-serum-albumin sytem, Eur. J. Biochem. 41, 539–546, 1974. 527. Hrkal, Z., Kodícek, M., Vodrázka, Z., et al., Haeme binding to human serum albumin and to the three large cyanogen albumin fragments, Int. J. Biochem. 9, 349–355, 1978. 528. Kolmatsu, T., Matsukawa, Y., and Tsuchida, E., Effect of heme structure on O2-binding properties of human serum albumin–heme hybrids: Intramolecular histidine coordination provides a stable O2-adduct complex, Bioconjug. Chem. 13, 397–402, 2002. 529. Komatsu, T., Ohmichi, N., Zunszain, P.A., et al., Dioxygenation of human serum albumin having a prosthetic heme group in a tailor-made heme pocket, J. Am. Chem. Soc. 126, 14304–14305, 2004. 530. Tsuchida, E., Sou, K., Nagakawa, A., et al., Artiicial oxygen carriers, hemoglobin vesicles and albumin–hemes, based on bioconjugate chemistry, Bioconjug. Chem. 20, 1419–1440, 2009. 531. Ascenzi, P., Bocedi, A., Notari, S., et al., Heme impairs allosterically drug binding to human serum albumin Sudlow’s site I, Biochem. Biophys. Res. Commun. 334, 481–486, 2005. 532. Fanali, G., Fesce, R., Agrati, C., Allosteric modulation of myristate and Mn(III) heme binding to human serum albumin. Optical and NMR spectroscopy characterization, FEBS J. 272, 4672–4683, 2005. 533. Simard, J.R., Zunszain, P.A., Hamilton, J.A., and Curry, S., Location of high and low afinity fatty acid binding sites on human serum albumin revealed by NMR drugcompetition analysis, J. Mol. Biol. 361, 336–351, 2006. 534. Ascenzi, P. and Fasano, M., Allostery in a monomeric protein: The case of human serum albumin, Biophys. Chem. 148, 16–22, 2010. 535. Fehske, K., Müller, W.E., and Wollert, U., The location of drug binding sites in human serum albumin, Biochem. Pharmacol. 30, 687–692, 1981.

Albumin

163

536. Phillips, R.S. and Marmorstein, R.Q., 6-Nitro-L-tryptophan: A novel spectroscopic probe of trp aporepressor and human serum albumin, Arch. Biochem. Biophys. 262, 337–344, 1988. 537. Sjöholm, I. and Grahnén, A., Circular dichroism studies on the binding of L-tryptophan to human serum albumin, FEBS Lett. 2, 109–122, 1972. 538. Sjöholm, I. and Ljungstedt, I., Studies on the tryptophan and drug-binding properties of human serum albumin fragments by afinity chromatography and circular dichroism measurements, J. Biol. Chem. 248, 8434–8441, 1973. 539. Cunningham, V.J., Hay, L., and Stoner, H.B., The binding of L-tryptophan to serum albumins in the presence of non-esteriied fatty acids, Biochem. J. 146, 653–658, 1975. 540. Bowmer, C.J. and Lindup, W.E., Inverse dependence of binding constants upon albumin concentration. Results of L-tryptophan and three anionic dyes, Biochim. Biophys. Acta 624, 260–270, 1980. 541. Sollenne, N.P., Wu, H.L., and Means, G.E., Disruption of the tryptophan binding site in the human serum albumin dimer, Arch. Biochem. Biophys. 207, 264–269, 1981. 542. Tanaka, N., Nishizawa, H., and Kumugi, S., Structure of pressure-induced denatured state of human serum albumin: A comparison with the intermediate in urea-induced denaturation, Biochim. Biophys. Acta 1338, 13–20, 1997. 543. Barker, L.F., Albumin products and the bureau of biologics, in Proceedings of the Workshop on Albumin, February 12–13, 1975, eds. J.T. Sgouris and A. René, Superintendent of Documents, Washington, DC, 1976. 544. Lane, R.S. and Vallet, L., Human albumin and plasma protein fraction, Lancet 323, 1245–1246, 1984. 545. Hoofnagle, J.H., Barker, L.F., Thiel, J., and Gerety, R.J., Hepatitis B virus and hepatitis B surface antigen in human albumin products, Transfusion 16, 141–147, 1976. 546. Hansen, J.F. and Ezban, M., A new quality albumin for therapeutic use, Dev. Biol. Stand. 48, 105–112, 1980. 547. Marley, P.B. and Gilbo, C.M., Temperature sensitivity within the pasteurization temperature range of prekallikrein activator in stable plasma protein solution (SPPS), Transfusion 21, 320–324, 1981. 548. Dengler, T., Stöcker, U., Kellner, S., and Fürst, G., Chemical and immunochemical characterization of polymers of aggregates in preparations of human serum albumin, Infusionstherapie 16, 160–164, 1989. 549. Blümel, J., Schmidt, I., Wilkommen, H., and Löwer, J., Inactivation of parovirus B19 during pasteurization of human serum albumin, Transfusion 42, 1011–1018, 2002. 550. Kreil, T.F., Unger, U., Orth, S.M., et al., N1Ni inluenza virus and the safety of plasma products, Transfusion 47, 452–459, 2007. 551. Anraku, M., Kuono, Y., Kai, Y., et al., The role of N-acetyl-methionate as a new stabilizer for albumin products, Int. J. Pharm. 329, 19–24, 2007. 552. Lin, J.J., Meyder, J.D., Carpenter, J.F., and Manning, M.C., Aggregation of human serum albumin during a thermal viral inactivation step, Int. J. Biol. Macromol. 42, 91–96, 2009. 553. Jeong, E.K., Sung, H.M., and Kim, I.S., Inactivation and removal of inluenza A virus H1N1 during the manufacture of plasma derivatives, Biologicals 38, 652–657, 2010. 554. Yu, M.W. and Finlayson, J.S., Stabilization of human albumin by caprylate and acetyltryptophanate, Vox Sang. 47, 28–40, 1984. 555. Shrake, A., Finlayson, J.S., and Ross, P.D., Thermal stability of human albumin measured by differential scanning calorimetry. I. Effects of caprylate and N-acetyltryptophanate, Vox Sang. 47, 7–18, 1984. 556. Cohen, P., Effects of heating on human serum albumin, in Proceedings of the Workshop on Albumin, eds. J.T. Sgouris and A. René, DHEW Publication No. (NIH) 76-925, Superintendent of Documents, Washington, DC, 1975.

164

Biotechnology of Plasma Proteins

557. Cohn, E.J., Oncley, J.L., Strong, L.E., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. I. The characterization of the protein fractions of human plasma, J. Clin. Invest. 23, 417–432, 1944. 558. Blood Programs in World War II, ed. D.B. Kendrick, The Bovine and Human Albumin Programs, Chapter XII, Ofice of Medical History, U.S. Army Medical Department, Superintendent of Documents, Washington, DC, 1964. 559. Scatchard, G., Gibson, S.T., Woodruff, L.M., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. IV. A study on the thermal stability of human serum albumin, J. Clin. Invest. 23, 445–453, 1944. 560. Scatchard, G., Strong, L.E., Hughes, W.L., Jr., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XXVI. The properties of solutions of human serum albumin of low salt content, J. Clin. Invest. 24, 671–679, 1945. 561. Ballou, G.D., Boyer, P.D., Luck, J.M., and Lum, F.G., Chemical, clinical, and immunological studies on the products of human plasma fractionation. V. The inluence of non-polar anions on the thermal stability of serum albumin, J. Clin. Invest. 23, 454–457, 1944. 562. Boyer, P.D., Lum, F.G., Ballou, G.A., et al., The combination of fatty acids and related compounds with serum albumin; stabilization against heat denaturation, J. Biol. Chem. 162, 181–198, 1946. 563. Boyer, P.D., Ballou, G.A., and Luck, J.M., The combination of fatty acids and related compounds with serum albumin: Stabilization against urea and guanidine denaturation, J. Biol. Chem. 162, 199–209, 1946. 564. Luck, J.M., Mandelate as a stabilizer of serum albumin, J. Phys. Colloid Chem. 51, 229–239, 1947. 565. Duggan, E.L. and Luck, J.M., The combination of organic anions with serum albumin; stabilization against urea denaturation, J. Biol. Chem. 172, 205–220, 1948. 566. Teresi, J.D. and Luck, J.M., The combination of organic anions with serum albumin. VIII. Fatty acid salts, J. Biol. Chem. 194, 823–834, 1952. 567. Hink, J.H., Jr. and Johnson, F.F., Studies on the stabilization of human serum albumin: The effect of pH, the stabilizers, and the albumin, J. Am. Pharm. Assoc. 40, 517–520, 1951. 568. Edsall, J.T., Stabilization of serum albumin to heat, and inactivation of the hepatitis virus, Vox Sang. 46, 338–340, 1984. 569. MacKay, M.E. and Martin, N.H., The stabilization of puriied human albumin to heat, Biochem. J. 65, 284–288, 1957. 570. Girard, M., Mousseau, N., Bietiot, H., and Whitehouse, L.W., Selected examples of physicochemical methods used for the analysis of biopharmaceuticals on the Canadian market, Pharmaceut. Sci. 3, 9–14, 1997. 571. Campbell, C., Shaw, R., Garinkle, B., and Gray, A., Gel permeation chromatography as a stability-indicating assay for human serum albumin, Dev. Biol. Stand. 44, 95–98, 1979. 572. Johnston, A., Uren, E., Johnstone, D., and Wu, J., Low pH, caprylate incubation as a second viral inactivation step in the manufacture of albumin. Parametric and validation studies, Biologicals 31, 213–221, 2003. 573. Miller, I., Gemeiner, M., Göstl, G., et al., Application of 2-D DIGE to survey the quality of biological medicines, Proteomics 11, 2120–2123, 2011. 574. Kobayashi, K., Nakamura, N., Sumi, A., et al., The development of recombinant human serum albumin, Ther. Apher. 2, 257–262, 1998. 575. Flensburg, J. and Belew, M., Characterization of recombinant human serum albumin using matrix-assisted laser desorption ionization time-of-light mass spectrometry, J. Chromatogr. A 1009, 111–117, 2003.

Albumin

165

576. Matsushita, S., Isima, Y., Chuang, V.T., et al., Functional analysis of recombinant human serum albumin domains for pharmaceutical applications, Pharm. Res. 21, 1924–1932, 2004. 577. Langer, K., Anhorn, M.G., Steinhauser, L., et al., Human serum albumin (HSA) nanoparticles: Reproducibility of preparation process and kinetics of enzymatic degradation, Int. J. Pharm. 347, 109–117, 2008. 578. Ang, W.H., Daldini, E., Julilerat-Jeanneret, L., and Dyson, P.J., Strategy to tether organometallic ruthenium-arene anticancer compounds to recombinant human serum albumin, Inorg. Chem. 46, 9048–9050, 2007. 579. Chuang, V.T. and Otagiri, M., Recombinant human serum albumin, Drugs Today (Barc.) 43, 547–561, 2007. 580. Han, Y., Jin, B.S., Lee, S.B., et al., Effects of sugar additives on protein stability of recombinant serum albumin during lyophilization and storage, Arch. Pharm. Res. 30, 1124–1131, 2007. 581. Ohnishi, K., Kawaguchi, A., Nakajima, S., et al., A comparative pharmacokinetic study of recombinant human serum albumin with plasma-derived human serum in patients with liver cirrhosis, J. Clin. Pharmacol. 48, 203–208, 2008. 582. Kasahara, A., Kita, K., and Tomita, E., Repeated administration of recombinant human serum albumin caused no serious allergic reactions in patients with liver cirrhosis: A multicenter clinical study, J. Gastroenterol. 43, 464–472, 2008. 583. Tsuraga, K., Oki, E., Yashiro, T., et al., Use of recombinant human serum albumin in pediatric patients with nephrotic syndrome, Pediatr. Nephrol. 24, 2275–2276, 2009. 584. Yamazoe, H. and Tanabe, T., Drug-carrying albumin ilm for blood-contacting biomaterials, J. Biomater. Sci. Polym. Ed. 21, 647–657, 2010. 585. Migliaccio, G., Sanchez, M., Masiello, F., et al., Humanized culture medium for clinical expansion of human erythroblasts, Cell Transplant. 19, 453–469, 2010. 586. Perkins, A.C. and Frier, M., Experimental biodistribution studies of 99mTc-recombinant human serum albumin (rHSA): A new generation of radiopharmaceutical, Eur. J. Nucl. Med. 21, 1231–1233, 1994. 587. Hunt, A.P., Frier, M., Johnson, R.A., et al., Preparation of Tc-99m-macroaggregated albumin from recombinant human albumin for lung perfusion imaging, Eur. J. Pharm. Biopharm. 62, 26–31, 2006. 588. Hoppmann, S., Miao, Z., Liu, S., et al., Radiolabeled afibody–albumin bioconjugates for HER2-positive cancer targeting, Bioconjug. Chem. 22, 413–421, 2011. 589. Nakatani, T., Sakamoto, Y., Ando, H., and Kobayashi, K., Effects of luid resuscitation with recombinant human serum albumin solution on maintaining hepatic energy metabolism in hemorrhagic shock rabbits, Res. Exp. Med. (Berl.) 196, 317–325, 1996. 590. Sumi, A., Okuyama, K., Kobayashi, K., et al., Puriication of recombinant human serum albumin. Eficient puriication using STREAMLINE, Bioseparation 9, 195–200, 1999. 591. Curling, J.M., Berglöf, J., Lindquist, L.-O., and Eriksson, S., A chromatographic procedure for the puriication of human plasma albumin, Vox Sang. 33, 97–107, 1977. 592. Björling, H., I. Plasma fractionation methods used in Sweden, Vox Sang. 23, 18–25, 1972. 593. Vassileva, R., Jakab, M., and Hasko, F., Application of ion-exchange chromatography for the production of human-albumin, J. Chromatogr. 216, 279–284, 1981. 594. Adcock, W.L., MacGregor, A., Davies, J.R., et al., Chromatographic removal and heat inactivation of hepatitis A virus during manufacture of human albumin, Biotechnol. Appl. Biochem. 28, 85–94, 1998. 595. Adcock, W.L., MacGregor, A., Davies, J.R., et al., Chromatographic removal and heat inactivation of hepatitis B virus during the manufacture of human albumin, Biotechnol. Appl. Biochem. 28, 169–178, 1998.

166

Biotechnology of Plasma Proteins

596. Tanaka, K., Shigueoka, E.M., Sawatani, E., et al., Puriication of human albumin by the combination of the method of Cohn with liquid chromatography, Braz. J. Med. Biol. Res. 31, 1383–1388, 1998. 597. Wolf, M., Kronenberg, H., Dodds, A., et al., A safety study of Albumex® 5, a human albumin solution produced by ion exchange chromatography, Vox Sang. 70, 198–202, 1996. 598. Che, Y., Wilson, F.J., Bertolini, J., et al., Impact of manufacturing improvements on clinical safety of albumin: Australian pharmacovigilance data for 1988–2005, Crit. Care Resusc. 8, 334–338, 2006. 599. Pitiot, O., Folley, L., and Vijayalakshmi, M.A., Protein adsorption on histidylaminohexyl-Sepharose 4B 1. Study of the mechanistic aspects of adsorption for the separation of human serum albumin from its non-enzymatically glycated isoforms (advanced glycation end products), J. Chromatogr. B 758, 161–172, 2001. 600. Crushelli, L., Clerico, A., Penna, G., et al., Colorimetric versus measurement of glycated and non-glycated serum albumin after afinity chromatography, Acta Diabetol. Lat. 27, 349–356, 1990. 601. Shmanai, V., Gontarev, S., Frey, S.K., and Schweigert, F.J., Modiication of aluminum chips for LDI mass spectrometry of proteins, J. Mass Spectrom. 42, 1504–1513, 2007. 602. Kremer, J.M.H., A simple method for isolating laboratory scale quantities of human serum albumin, Vox Sang. 42, 223–224, 1982. 603. Fisher, M.J.E., Bos, O.J.M., van der Linden, R.F., et al., Steroid-binding to human serum-albumin and fragments thereof—Role of protein conformation and fatty acid content, Biochem. Pharmacol. 45, 2411–2416, 1993. 604. Mannuzza, F.J. and Montalto, J.G., Is bovine albumin too complex to be just a comodity?, Bioprocess Int. 8, 40–43, February 2010. 605. Weisberg, H.F., Osmotic pressure of the serum proteins, Ann. Clin. Lab. Sci. 8, 155–164, 1978. 606. Starling, E.H., On the absorption of luids from the connective tissue spaces, J. Physiol. 19, 312–326, 1896. 607. Arroyo, V., Review article: Albumin in the treatment of liver diseases—New features of a classical treatment, Aliment. Pharmacol. Ther. 16(Suppl. 5), 1–5, 2002. 608. Wilkinson, P.C. and McKay, I.C., The chemotactic activity of native and denatured serum albumin, Int. Arch. Allergy 41, 237–247, 1971. 609. Körmöczi, G.F., Wölfel, U.M., Rosenkranz, A.R., et al., Serum proteins modiied by neutrophil-derived oxidants as mediators of neutrophil stimulation, J. Immunol. 167, 451–460, 2001. 610. Polzer, S., van Yperen, M., Kirst, M., et al., Neutralization of X4- and R5-tropic HIV-1 NL4-3 variants by HOCl-modiied serum albumins, BMC Res. Notes 3, 155, 2010. 611. Mera, K., Anraku, M., Kitamura, K., et al., The structure and function of oxidized albumin in hemodialysis patients: Its role in elevated oxidative stress via neutrophil burst, Biochem. Biophys. Res. Commun. 334, 1322–1326, 2005. 612. Jaisson, S., Delevallée-Forte, C., Toure, F., et al., Carbamylated albumin is a potent inhibitor of polymorophonuclear neutrophil respiratory burst, FEBS Lett. 581, 1509– 1513, 2007. 613. Lang, J.D., Jr., Figueroa, M., Chumley, P., et al., Albumin and hydroxylethyl starch modulate oxidative injury to vascular endothelium, Anesthesiology 100, 51–58, 2004. 614. Powers, K.A., Kapus, A., Khadaroo, R.G., et al., 25% Albumin modulates adhesive interactions between neutrophils and the endothelium following shock/resuscitation, Surgery 132, 391–398, 2002. 615. Collison, K.S., Parhar, R.S., Saleh, S.S., et al., RAGE-mediated neutrophil dysfunction is evoked by advanced glycation end products (AGEs), J. Leukoc. Biol. 71, 433–444, 2002.

Albumin

167

616. Mero, K., Takeo, K., Izumi, M., et al., Effect of reactive-aldehydes on the modiication and dysfunction of human serum albumin, J. Pharm. Sci. 99, 1614–1625, 2010. 617. Don, B.R. and Kaysen, G., Serum albumin: Relationship to inlammation and nutrition, Semin. Dial. 17, 432–437, 2004. 618. Wilkinson, P.C. and McKay, I.C., Recognition in leucocyte chemotaxis. Studies with structurally modiied proteins, Antibiot. Chemother. 19, 421–441, 1974. 619. Wilkinson, P.C. and McKay, I.C., The molecular requirements for chemotactic attraction of leucocytes by proteins. Studies of proteins with synthetic side groups, Eur. J. Immunol. 2, 570–577, 1972. 620. Wilkinson, P.C., Recognition and response in mononuclear and granular phagocytes, Clin. Exp. Immunol. 25, 355–366, 1975. 621. Tove, S.B., The esterolytic activity of serum albumin, Biochim. Biophys. Acta 57, 230– 235, 1962. 622. Casida, J.E. and Augustinsson, K.B., Reaction of plasma albumin with 1-napthyl-Nmethylcarbamate and certain other esters, Biochim. Biophys. Acta 36, 411–426, 1959. 623. Chapuis, N., Brühlmann, C., Reist, M., et al., The esterase-like activity of serum albumin may be due to cholinesterase contamination, Pharm. Res. 18, 1435–1439, 2001. 624. Kunkel, H.G. and Ward, S.M., Plasma esterase activity in patients with liver disease and nephrotic system, J. Exp. Med. 86, 325–336, 1947. 625. Powell, H.M. and Jamieson, W.A., Merthiolate as germicide, Am. J. Hyg. 13, 296–300, 1931. 626. Jamieson, W.A. and Powell, H.M., Merthiolate as a preservative for biological products, Am. J. Epidemiol. 14, 218–224, 1931. 627. Janeway, C.A., Gibson, S.T., Woodruff, L.M., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. VII Concentrated human serum albumin, J. Clin. Invest. 23, 465–490, 1944. 628. U.S. Department of Commerce Biography of Scientiic and Industry Reports. 629. Hartley, B.S. and Kilby, B.A., The reaction of p-nitrophenyl esters with chymotrypsin and insulin, Biochem. J. 56, 288–297, 1954. 630. Kézdy, F.J. and Beneer, M.L., The kinetics of the α-chymotrypsin-catalyzed hydrolysis of p-nitrophenyl acetate, Biochemistry 1, 1097–1106, 1962. 631. Lundblad, R.L., Observatons on the hydrolysis of p-nitrophenyl acylates by puriied bovine thrombin, Thromb. Diath. Haemorrh. 30, 248–254, 1973. 632. Park, J.H., Meriwether, B.P., Clodfelder, P., and Cunningham, L.W., The hydrolysis of p-nitrophenyl acetate catalyzed by 3-phosphoglyceraldehyde dehydrogenase, J. Biol. Chem. 236, 136–141, 1961. 633. De Caro, J., Rouim, P., and Rovery, M., Hydrolysis of p-nitrophenyl acetate by the peptide chain fragment (336–449) of porcine pancreatic lipase, Eur. J. Biochem. 158, 601–607, 1986. 634. Petz, D. and Schneider, F., Kinetic analysis of the catalytic properties of peptides in ester hydrolysis, Z. Naturforsch. 31, 675–678, 1976. 635. Thangthaeng, N., Sumien, N., Foster, M.J., et al., Nongradient blue native gel analysis of serum proteins and in gel detection of serum esterase activity, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 879, 386–394, 2010. 636. Steinbuch, M., Granier, C., Tavernier, D., and Faure, A., Side products of routine plasma fractionation. I. Serum cholinesterase, Vox Sang. 36, 142–149, 1979. 637. Li, B., Sedalek, M., Manoharan, I., et al., Butyrylcholinesterase, paraoxanase, and albumin esterase, but not carboxylesterase, are present in human plasma, Biochem. Pharmacol. 70, 1673–1684, 2005. 638. Kolarich, D., Weber, A., Pabst, M., et al., Glycoproteomic characterization of butyrylcholinesterase from human plasma, Proteomics 8, 254–263, 2008.

168

Biotechnology of Plasma Proteins

639. Alberti, J., Martinet, A., Sentellas, S., and Salvà, M., Identiication of the human enzymes responsible for the enzymatic hydrolysis of aclidinium bromide, Drug Metab. Dispos. 38, 1202–1210, 2010. 640. Saxena, A., Luo, C., and Doctor, B.P., Developing procedures for the large-scale puriication of human serum butyrylcholinesterase, Protein Exp. Purif. 61, 191–196, 2008. 641. Roche, M., Dufour, C., Loonis, M., et al., Olive phenols eficiently inhibit the oxidation of serum albumin-bound linoleic acid and butyrylcholinesterase, Biochim. Biophys. Acta 1790, 240–248, 2009. 642. Tildon, J.T. and Ogilivie, J.W., The esterase activity of bovine mercaptalbumin. The reaction of the protein with p-nitrophenyl acetate, J. Biol. Chem. 247, 1265–1271, 1972. 643. Means, G.E. and Bender, M.L., Acylation of human serum albumin by p-nitrophenyl acetate, Biochemistry 14, 4989–4994, 1975. 644. Ikeda, K., Kurono, Y., Ozeki, Y., and Yotsuyanagi, T., Effect of drug bindings on esterase activity of human serum albumin. Dissociation constants of the complexes between the protein and drugs such as N-arylanthranilic acids, coumarin derivatives and prostaglandins, Chem. Pharm. Bull. 27, 80–87, 1979. 645. Kurono, Y., Mari, T., Yotsuyanagi, T., and Ikeda, K., Esterase-like activity of human serum albumin: Structure–activity relationships for the reactions with phenyl acetates and p-nitrophenyl esters, Chem. Pharm. Bull. 27, 2781–2786, 1979. 646. Kurono, Y., Kondo, T., and Ikeda, K., Esterase-like activity of human serum albumin: Enantioselectivity in the burst phase of reaction with p-nitrophenyl α-methoxyphenyl acetate, Arch. Biochem. Biophys. 227, 339–341, 1983. 647. Kurono, Y., Yamada, H., Hata, N., et al., Esterase-like activity of human serum albumin. IV. Reactions with substituted aspirins and 5-nitrosalicyl esters, Chem. Pharm. Bull. 32, 3715–3719, 1984. 648. Yoshida, K., Kurono, Y., Mori, Y., and Ikeda, K., Esterase-like activity of human serum albumin. V. Reaction with 2,4-dinitrophenyl diethyl phosphate, Chem. Pharm. Bull. 33, 4995–5001, 1985. 649. Kurono, Y., Furikawa, A., Tsuji, T., and Ikeda, K., Esterase-like activity of human serum albumin. VI. Reaction with p-nitrophenyl glycinate, Chem. Pharm. Bull. 36, 4068–4074, 1988. 650. Kurono, Y., Kushida, I., Tanaka, H., and Ikeda, K., Esterase-like activity of human serum albumin. VIII. Reaction with amino acid p-nitrophenyl esters, Chem. Pharm. Bull. 40, 2169–2172, 1992. 651. Murachi, T., A general reaction of diisopropylphosphoroluoridate with proteins without direct effect of enzymic activities, Biochim. Biophys. Acta 71, 239–241, 1963. 652. Dubois-Presle, N., Lapicque, F., Maurice, M.H., et al., Stereoselective esterase activity of human serum albumin toward ketoprofen glucuronide, Mol. Pharmacol. 47, 647– 653, 1995. 653. Watanabe, H., Tanase, S., Nakajou, K., et al., Role of arg-410 and tyr-411 in human serum albumin for ligand binding and esterase-like activity, Biochem. J. 349, 813–819, 2000. 654. Suji, G., Khedkar, S.A., Singh, S.K., et al., Binding of lipoic acid induces conformational change and appearance of a new binding site in methylglyoxal modiied serum albumin, Protein J. 27, 205–214, 2008. 655. Li, B., Schopfer, L.M., Hinrichs, S.H., et al., Matrix-assisted laser desorption/ionization time-of-light mass spectrometry assay for organophosphorous toxicants bound to human albumin at Tyr411, Anal. Biochem. 361, 263–272, 2007. 656. Ding, S.J., Carr, J., Carlson, J.E., et al., Five tyrosine and two serines in human albumin are labeled by the organophosphorous agent FP-biotin, Chem. Res. Toxicol. 21, 1787–1794, 2008.

Albumin

169

657. Sakurai, Y., Ma, S.F., Watanbe, H., et al., Esterase-like activity of serum albumin: Characterization of its structural chemistry using p-nitrophenyl esters as substrates, Pharm. Res. 21, 285–292, 2004. 658. Lockridge, O., Xue, W., Gaydess, A., et al., Pseudo-esterase activity of human albumin: Slow turnover on tyrosine 411 and stable acetylation of 82 residues including 59 lysines, J. Biol. Chem. 283, 22582–22590, 2008. 659. Schopfer, L.M., Champion, M.M., Tamblyn, N., et al., Characteristic mass spectral fragments of the organophosphorous agent FP-biotin and FP-biotinylated peptides from trypsin and bovine albumin (Tyr410), Anal. Biochem. 345, 122–132, 2005. 660. Fujino, T., Kojima, M., Beppu, M., et al., Identiication of the cleavage sites of oxidized protein that are susceptible to oxidized protein hydrolase (OPH) in the primary and tertiary structures of the protein, J. Biochem. 127, 1087–1093, 2000. 661. Rigg, B.M. and Baird, C.W., Association of intravenous albumin with alkaline phosphatase activity, J. Clin. Pathol. 18, 441–442, 1965. 662. Elsbach, P. and Pettis, P., Phospholipase activity associated with serum albumin, Biochim. Biophys. Acta 296, 89–93, 1973. 663. Singleton, C.L. and Killian, G.J., A study of phospholipase in albumin and its role in inducing the acrosome reaction of guinea pig spermatozoa in vitro, J. Androl. 4, 150– 156, 1983. 664. Cha, M.-K. and Kim, I.-H., Glutathione-linked thiol peroxidase activity of human serum albumin: A possible antioxidant role of serum albumin, Biochem. Biophys. Res. Commun. 222, 619–625, 1996. 665. Cha, M.-K. and Kim, I.-H., Disulide between Cys392 and Cys438 of human serum albumin is redox-active which is responsible for the thioredoxin-supported lipid peroxidase activity, Arch. Biochem. Biophys. 445, 19–25, 2006. 666. Yang, J., Petersen, C.E., Ha., C.-E., and Bhagavan, N.V., Structured insight into human serum albumin-mediated prostaglandin catalysis, Protein Sci. 11, 19–25, 2006. 667. Sogorb, M.A., Carrerra, V., Benabent, M., and Vilanova, E., Rabbit serum albumin hydrolyzes the carbamate carbaryl, Chem. Res. Toxicol. 15, 520–526, 2002. 668. Sogorb, M.A., Alvarez-Escalante, C., Carrera, V., and Vilanova, E., An in vitro approach for demonstrating the critical role of serum albumin in the detoxiication of the carbamate carbaryl at in vivo toxicologically relevant concentrations, Arch. Toxicol. 81, 113–119, 2007. 669. Sogorb, M.A., Garcia-Arguelles, S., Carrera, V., and Vilanova, E., Serum albumin is as eficient as paraoxonase in the detoxiication of paraoxon at toxicologically relevant concentration, Chem. Res. Toxicol. 21, 1524–1529, 2008. 670. Cordova, J., Ryan, J.D., Boonyaratanakornkit, B.B., and Clark, D.S., Esterase activity of bovine serum albumin up to 160°C: A new benchmark for biocatalysis, Enzyme Microb. Technol. 42, 278–283, 2008. 671. Gerasimova, Y.V., Erchenko, I.A., Shakirov, M.M., et al., Interaction of human serum albumin and its clinically relevant modiication with oligoribonucleotides, Bioorg. Med. Chem. Lett. 18, 4511–4514, 2008. 672. Gerasimova, Y.V., Knoore, D.D., Shakirov, M.M., et al., Human serum albumin as a catalyst of RNA cleavage: N-Homocysteinylation and N-phosphorylation by oligonucleotide afinity reagent alter the reactivity of the protein, Bioorg. Med. Chem. Lett. 18, 5396–5398, 2008. 673. Gerasimova, Y.V., Bobik. T.V., Ponomarenko, N.A., et al., RNA-hydrolyzing activity of human serum albumin and its recombinant analogue, Bioorg. Med. Chem. Lett. 20, 1427–1431, 2010. 674. Jones, R.G.A., Liu, Y., Halls, C., et al., Release of proteolytic activity following reduction in therapeutic human serum albumin containing products: Detection with a new neoepitope endopeptidase immunoassay, J. Pharmaceut. Biomed. Anal. 54, 74–80, 2011.

170

Biotechnology of Plasma Proteins

675. Janeway, C.A., Other uses of plasma fractions with particular reference to serum albumin, Ann. Intern. Med. 26, 368–376, 1947. 676. Janeway, C.A., Berenberg, W., and Hutchins, G., Indications and uses of blood, blood derivatives and blood substitutes, Med. Clin. North Am. 29, 1069–1094, 1945. 677. Sapmaz, I., Manduz, S., Sanri, U.S., et al., Inluence of albumin concentration in priming solution on blood viscosity under hypothermic conditions, Cardiovasc. J. Afr. 20, 168–169, 2009. 678. Kozek-Langenecker, S.A., Inluence of luid therapy on the hemostatic system of intensive care patients, Best Pract. Res. Clin. Anaesthesiol. 23, 225–236, 2009. 679. Arroyo, V., Human serum albumin: Not just a plasma volume expander, Hepatology 50, 355–357, 2009. 680. Conn, H.O., The use, misuse and abuse of albumin infusions, in Albumin Structure, Function and Uses, eds. V.M. Rosenoer, M. Oratz, and M.A. Rothschild, Pergammon Press, Oxford, UK, 1977. 681. Delaney, A.P., Dan, A., McCaffrey, J., and Finfer, S., The role of albumin as a resuscitation luid for patients with sepsis: A systematic review and meta-analysis, Crit. Care Med. 39, 386–391, 2011. 682. Bunn, F., Trivedi, D., and Ashraf, S., Colloid solutions for luid resuscitation, Cochrane Database Syst. Rev. 3, CD001319, 2011. 683. Perel, P. and Roberts, I., Colloids versus crystalloids for luid resuscitation in critically ill patients, Cochrane Database Syst. Rev. 3, CD000567, 2011. 684. Evans, T.W., Review article: Albumin as a drug—Biological effects of albumin unrelated to oncotic pressure, Aliment. Pharmacol. Ther. 16(Suppl. 5), 6–11, 2002. 685. Quinlin, G.J., Martin, G.S., and Evans, T.W., Albumin: Biochemical properties and therapeutic properties, Hepatology 41, 1211–1219, 2005. 686. Cutler, E.E. and Lögdberg, L.E., Albumin, in Blood Banking and Transfusion Medicine. Basic Principles and Practice, 2nd edn., eds. C.D. Hilyer, L.E. Silberstein, P.M. Ness, K.C. Andrews, and J.D. Roback, Chapter 21, pp. 278–287, Churchill-Livingstone/ Elsevier, Philadelphia, 2007. 687. Stange, J., Mitzner, S.R., Klammt, S., et al., Liver support by extracorporeal blood puriication: A clinical observation, Liver Transplant. 6, 603–613, 2000. 688. Mitzner, S.R., Stange, J., Klammt, S., et al., Albumin dialysis MARS: Knowledge from 10 years of clinical investigation, ASAIO J. 55, 498–502, 2009. 689. Patzer II, J.F., Lopez, R.C., Zhu, Y., et al., Bioartiicial liver assist devices in support of patients with liver failure. Hepatobiliary Pancreat. Dis. Int. 1, 18–25, 2002. 690. Riordan, S.M., Kurtovic, J., and Williams, R., Fulminant hepatic failure, in Schiff’s Diseases of the Liver, 10th edn., eds. E.R. Schiff, M.R. Sorrell, and W.C. Maddrey, Chapter 21, pp. 601–636, Lippincott Williams & Wilkins, Philadelphia, 2007. 691. Bañares, R. and Catalina, M.-V., Extracorporeal artiicial liver support systems, in Clinical Gastroenterology: Chronic Liver Failure, eds. P. Ginès, P.S. Kamath, and V. Arroyo, pp. 501–520, Humana/Springer, New York, 2011. 692. Memon, I. and Klein, C.L., Impact of hepatorenal syndrome and liver transplantation, Curr. Opin. Organ Transplant. 16, 301–305, 2011. 693. Plotz, P.H., Berk, P.D., Scharschmidt, B.F., et al., Removing substances from blood by afinity chromatography. I. Removing bilirubin and other albumin-bound substances from plasma and blood with albumin-conjugated beads, J. Clin. Invest. 53, 778–785, 1974. 694. Nakae, H., Eguchi, Y., Saotome, T., et al., Multicenter study of plasma diailtration in patients with acute liver failure, Ther. Apher. Dial. 14, 444–450, 2010. 695. Kaplan, A.A. and Epstein, M., Extracorporeal blood puriication in the management of patients with hepatic failure, Semin. Nephrol. 17, 576–582, 1997.

Albumin

171

696. Sen, P.K., Bhalerao, R.A., Parulkar, G.P., et al., Use of isolated perfused cadaveric liver in the management of hepatic failure, Surgery 59, 774–781, 1966. 697. Chalstray, L.J. and Parbhoo, S.P., Circuitry and technique of extracorporeal porcine liver perfusion for the treatment of hepatic coma, Br. J. Surg. 58, 522–524, 1971. 698. Olumide, F., Eliashiv, A., Kralios, N., et al., Hepatic support with hepatocyte suspension in permeable membrane dialyzer, Surgery 82, 599–606, 1977. 699. Rozga, J., Podesta, L., LePage, E., et al., A bioartiicial liver to treat severe acute liver failure, Ann. Surg. 219, 538–544, 1994. 700. Chen, S.C., Hewitt, W.R., Watanabe, F.D., et al., Clinical experience with a porcine hepatocyte-based liver support system, Int. J. Artif. Organs 19, 664–669, 1996. 701. Schrem, H., Kleine, M., Borlak, J., and Klempnauer, J., Physiological incompatibilities of porcine hepatocytes for clinical liver support, Liver Transplant. 12, 1832–1840, 2006. 702. Miki, T., Ring, A., and Gerlach, J., Hepatic differentiation of human embryonic stem cells is promoted by three-dimensional dynamic perfusion culture conditions, Tissue Eng. C Meth. 17, 557–568, 2011. 703. Etteldorf, J.N., Dobbins, W.T., and Summitt, R.L., Intermittent peritoneal dialysis using 5 per cent albumin in the treatment of salicylate intoxication in children, J. Pediatr. 58, 226–236, 1961. 704. Grollman, A.P. and Odell, G.B., Removal of bilirubin by albumin binding during intermittent peritoneal dialysis, N. Engl. J. Med. 267, 279–282, 1962. 705. Falkenhagen, D., Schmitt, E., Schneider, P., et al., Plasma sorption: An alternative treatment for intoxication, in Plasmapheresis: Therapeutic Application and New Techniques, eds. Y. Nosé, J.W. Smith, P.S. Malchesky, and R.S. Krakauer, pp. 251–256, Raven Press, New York, 1983. 706. Falkenhagen, D., Strubl, W., Vogt, G., et al., Fractionation plasma separation and adsorption system: A novel system for blood puriication to remove albumin bound substances, Artif. Organs 23, 81–86, 1989. 707. Sangster, B., Van Heijst, N.R., and Sixma, J.J., The inluence of haemoperfusion on hemostasis and cellular constituents of the blood in the treatment of intoxication. A comparative study of three types of columns (Haemocel, Amberlite XAD-4, Gambro Adsorba 300 C), Arch. Toxicol. 47, 269–278, 1981. 708. Hughes, R.D., Ton, H.Y., and Langley, P.G., et al., The use of in vitro perfusion circuited to evaluate the blood compatibility of albumin-coated Amberlite XAD-7 resin, Int. J. Artif. Organs 1, 129–134, 1979. 709. Raja, R.M., Resin hemoperfusion for drug intoxication, Int. J. Artif. Organs 9, 319–322, 1986. 710. Stange, J., Ramlow, W., Mitzner, S.R., et al., Dialysis against a recycled albumin solution enables the removal of albumin-bound toxins, Artif. Organs 17, 809–813, 1993. 711. Mitzner, S.R., Stange, U., Klammt, S., et al., Improvement of hepatorenal syndrome with extracorporeal albumin MARS: Results of a prospective, randomized, controlled clinical trials, Liver Transplant. 6, 277–286, 2000. 712. Stange, J., Mitzner, S.R., Klammt, S., et al., Liver support by extracorporeal blood puriication: A clinical observation, Liver Transplant. 6, 603–613, 2000. 713. Detlefsen, W.D., Blood and casein adhesives for bonding woods, in Adhesives from Renewable Resources, eds. R.N. Hemingway, A.H. Conner, and S.J. Branham, Chapter 31, pp. 445–452, American Chemical Society, Washington, DC, 1989. 714. Yang, I., Kuo, M., Myers, D.J., and Pu, A., Comparison of protein-based adhesive resins for wood composites, J. Wood Sci. 52, 503–508, 2006. 715. Hides, G., Kastin, A., Mullerad, M., et al., Sutureless nephron-sparing surgery: Use of albumin glutaraldehyde tissue adhesive (Bioglue), Urology 67, 697–700, 2006. 716. Passage, J., Jalali, H., Tam, R.K., et al., Bioglue surgical adhesive—An appraisal of its indications in cardiac surgery, Ann. Thorac. Surg. 74, 432–437, 2002.

172

Biotechnology of Plasma Proteins

717. Zehr, K.G., Use of bovine albumin-glutaraldehyde glue in cardiovascular surgery, Ann. Thorac. Surg. 84, 1048–1052, 2007. 718. de la Portilla, F., Rada, R., Vega, J., et al., Long-term results change conclusions on BioGlue in the treatment of high transspincteric anal istulas, Dis. Colon Rectum 53, 1220–1221, 2010. 719. Epstein, N.E., Dural repair with four spinal sealants: Focused review of the manufacturers’ inserts and the current literature, Spine J. 10, 1065–1068, 2010. 720. Gaberel, T., Borgey, F., Thibon, P., et al., Surgical site infection associated with the use of bovine serum albumine-glutaraldehyde surgical adhesive (BioGlue) in cranial surgery: A case-control study, Acta Neurochir. (Wien) 153, 156–162, 2011. 721. Chevalier, B., Reant, P., Lafite, S., and Barandon, L., Spontaneous istualization of a caseous calciication of the mitral annulus: An exceptional cause of stroke, Eur. J. Cardiothorac. Surg. 39, e184–e185, 2011. 722. Khan, H., Chaubey, S., and Desai, J., Early failure of coronary artery bypass grafts: An albumin cross-linked glutaraldehyde (BioGlue) related complication, J. Card. Surg. 26, 264–266, 2011. 723. Liu, Y.H., Wu, Y.C., Hsieh, M.J., and Ko, P.J., Effective closure of a tracheal incision by BioGlue in natural oriice transluminal endoscopic surgery performed for thoracic exploration, J. Thorac. Cardiovasc. Surg. 142(2), 458–460, 2011. 724. Babin-Ebell, J., Bougioukakis, P., Urbanski, P., et al., Foreign material reaction to BioGlue® as a possible cause of cardiac tamponade, Thorac. Cardiovasc. Surg. 58, 489– 491, 2010. 725. Poppas, D.P., Schlossberg, S.M., Richmond, I.L., et al., Laser welding in urethral surgery: Improved results with a protein solder, J. Urol. 139, 415–417, 1988. 726. Oz, M.C., Johnson, J.P., Paragi, S., et al., Tissue soldering by use of indocyanine green dye-enhanced ibrinogen with the near infrared diode laser, J. Vasc. Surg. 11, 718–725, 1990. 727. Mozami, N., Oz., M.C., Bass, L.S., and Treat, M.R., Reinforcement of colonic anastomoses with a laser and dye-enhanced ibrinogen, Arch. Surg. 125, 1452–1454, 1990. 728. Oxford English Dictionary, Oxford University Press, Oxford, UK, 2006; http://www. oed.com. 729. Bass, L.S. and Treat, M.R., Laser tissue welding: A comprehensive review of current and future clinical applications, in Laser Surgery and Medicine, ed. A. Puliaito, WileyLiss, New York, 1996. 730. Putnam, F.W., Protein denaturation, in The Proteins, eds. H. Neurath and K. Bailey, Vol. 1, Part G, Academic Press, New York, 1953. 731. Lumry, R. and Eyring, H., Conformational changes of proteins, J. Phys. Chem. 58, 110– 120, 1954. 732. Misawa, S. and Kumagai, I., Refolding of therapeutic proteins produced in Escherichia coli as inclusion bodies, Biopolymers 51, 297–307, 1999. 733. Singh, S.M. and Panda, A.K., Solubilization and refolding of bacterial inclusion body proteins, J. Biosci. Bioeng. 99, 303–310, 2005. 734. Cromwell, M.E.M., Hilario, E., and Jacobsen, F., Protein aggregation and bioprocessing, Am. Assoc. Pharm. Sci. J. 8, E572–E579, 2006. 735. Murphy, R.M. and Kendrick, B.S., Protein misfolding and aggregation, Biotechnol. Prog. 23, 548–552, 2007. 736. Glaser, C.B., Busby, T.F., Ingham, K.C., and Childs, A., Thermal denaturation of alpha1protease inhibitor. Stabilization by neutral salts and sugars, Am. Rev. Respir. Dis. 128, 77–81, 1983. 737. Raman, B., Ramakrishana, T., and Rao, C.M., Refolding of denatured and denatured/ reduced lysozyme at high concentrations, J. Biol. Chem. 271, 17067–17072, 1996.

Albumin

173

738. Hammarström, P., Persson, M., and Freskgård, P.O., Structural mapping of an aggregation nucleation site in a molten globule intermediate, J. Biol. Chem. 274, 32897–32903, 1999. 739. Militella, V., Vetri, V., and Leone, M., Conformational changes involved in thermal aggregation processes of bovine serum albumin, Biophys. Chem. 105, 133–141, 2003. 740. Tsai, C.-J., Lin, S.L., Wolfson, H.J., and Nussinov, R., Studies of protein–protein interfaces: A statistical analysis of the hydrophobic effect, Protein Sci. 6, 53–64, 1997. 741. Kundu, B. and Guptasarma, P., Hydrophobic dye inhibits aggregation of molten carbonic anhydrase during thermal unfolding and refolding, Proteins 37, 321–324, 1999. 742. Kundu, B. and Guptasarma, P., Use of a hydrophobic dye to indirectly probe the structural organization and conformational plasticity of molecules in amorphous aggregates of carbonic anhydrase, Biochem. Biophys. Res. Commun. 293, 572–577, 2002. 743. Cheung, J.K., Raverker, P.S., and Truskett, T.M., Analytical model for studying how environmental factors inluences protein conformational stability in solution, J. Chem. Phys. 125, 234903, 2006. 744. Rezaei-Ghaleh, N., Ramshini, H., Ebrahim-Habibi, A., et al., Thermal aggregation of α-chymotrypsin: Role of hydrophobic and electrostatic interactions, Biophys. Chem. 132, 23–32, 2008. 745. McNally, K.M., Sorg, B.S., Chan, E.K., et al., Optimal parameters for laser tissue soldering, Part I: Tensile strength and scanning electron microscopy analysis, Lasers Surg. Med. 24, 319–331, 1999. 746. Berchane, N.S., Andrews, M.J., Kerr, S., et al., On the mechanical properties of bovine serum albumin (BSA) adhesives, J. Mater. Sci. Mater. Med. 19, 1831–1838, 2008. 747. Duly, E.B., Grimason, S., Grimaon, P., et al., Measurement of serum albumin by capillary zone electrophoresis, bromocresol green, bromocresol purple, and immunoassay methods, J. Clin. Pathol. 56, 780–781, 2003. 748. Ott, B., Zuger, B.J., Erni, D., et al., Comparative in vitro study of tissue welding using a 808 nm diode laser and Ho: YAG laser, Lasers Surg. Med. 16, 260–266, 2001. 749. Shrake, A., Finlayson, J.S., and Ross, P.D., Thermal stability of human albumin measured by differential scanning calorimetry. I. Effects of caprylate and N-acetyltryptophanate, Vox Sang. 47, 7–18, 1984. 750. Michnik, A., Thermal stablitiy of bovine serum albumin DSC studies, J. Therm. Anal. Calorim. 71, 509–519, 2003. 751. Shrake, A., Frazier, D., and Schwarz, F.P., Thermal stabilization of human albumin by medium and short chain n-alkyl fatty acid anions, Biopolymers 81, 235–248, 2006. 752. Shrake, A. and Ross, P.D., Origins and consequences of ligand-induced multiphasic thermal protein denaturation, Biopolymers 32, 925–940, 1992. 753. Ren, Z., Zie, H., Lagerquist, K.A., et al., Optimal dye concentration and irradiation for laser-assisted vascular anastomosis, J. Clin. Laser Med. Surg. 22, 81–86, 2004. 754. McNally, K.M., Sorg, B.S., and Welch, A.J., Novel solid protein solder designs for laserassisted tissue repair, Lasers Surg. Med. 27, 147–157, 2000. 755. Kirsch, A.J., Cooper, C.S., Gath, J., et al., Laser tissue soldering for hypospadias repair: Results of a controlled prospective clinical trial, J. Urol. 165, 574–577, 2001. 756. Xie, H., Shaffer, B.S., Prahl, S.A., and Gregory, K.W., Intraluminal albumin stent assisted laser welding for urethral anastomosis, Lasers Surg. Med. 31, 225–229, 2002. 757. Xie, H., Bendre, S.C., Burke, A.P., et al., Laser-assisted vascular and end to end anastomosis of elastic heterograft to carotid artery with an albumin stent: A preliminary in vivo study, Lasers Surg. Med. 35, 201–205, 2004. 758. Maitz, P.K.M., Trickett, R.J., Dekker, P., et al., Sutureless microvascular anastomoses by a biodegradable laser-activated solid protein solder, Plast. Reconstr. Surg. 104, 1726– 1731, 1999.

174

Biotechnology of Plasma Proteins

759. Wright, B., Vicaretti, M., Schwaiger, N., et al., Laser-assisted end-to-end Bioweld® anastomosis in an ovine model, Lasers Surg. Med. 39, 667–673, 2007. 760. Lauto, A., Foster, L.J.R., Ferris, L., et al., Albumin-genipin solder for laser tissue repair, Lasers Surg. Med. 35, 140–145, 2004. 761. Pabittei, D.R., Heger, M., Beek, J.F., et al., Optimization of suture-free laser-assisted vessel repair by solder-doped electrospun poly (ɛ-caprolactone) scaffold, Ann. Biomed. Eng. 39, 223–234, 2011. 762. Seres, D.S., Surrogate nutrition markers, malnutrition, and adequacy of nutrition support, Nutr. Clin. Pract. 20, 308–313, 2005. 763. Friedman, A.N. and Fadem, S.Z., Reassessment of albumin as a nutritional marker in kidney disease, J. Am. Soc. Nephrol. 21, 223–230, 2010. 764. Sharouni, E., Georgiadou, P., and Voudris, V., Ischemia modiied-albumin changes— Review and clinical implications, Clin. Chem. Lab. Med. 49, 177–184, 2011. 765. Sinha, M.K., Gaze, D.C., Tippins, J.R., et al., Ischemia modiied albumin is a sensitive marker of myocardial ischemia after percutaneous coronary intervention, Circulation 107, 2403–2405, 2003. 766. Bar-Or, D. and Solomons, C., Test for the rapid evaluation of ischemic state, U.S. Patent 5,227,307, July 13, 1993. 767. Rao, M.S.N. and Lal, H., Binding of the cobaltous by native and modiied bovine serum albumin, J. Am. Chem. Soc. 76, 4867–4868, 1954. 768. Rao, M.S.N. and Lal, H., Metal protein interactions in buffer solutions. Part III. Interaction of CuII, ZnII, CdII, CoII, (and NiII) with native and modiied bovine serum albumins, J. Am. Chem. Soc. 80, 3226–3235, 1958. 769. Nandedkar, A.K.N., Basu, P.K., and Friedberg, F., Co++ binding by plasma proteins, Bioinorg. Chem. 2, 149–157, 1972. 770. Nandedkar, A.K.N., Hong, M.S., and Friedberg, F., Co2+ binding by plasma albumin, Biochem. Med. 9, 177–183, 1974. 771. Lakusta, H. and Sarkar, B., Equilibrium studies of zinc (II) and cobalt (II) binding to tripeptide analogues of the amino terminus of human serum albumin, J. Inorg. Biochem. 11, 303–315, 1979. 772. Trueba, M., Vallejo, A., Zatón, Z., and Abad, C., Preferential solvation of bovine serum albumin in cobalt nitrate solutions, Inorg. Chim. Acta 107, 133–137, 1985. 773. Sadler, P.J., Tucker, A., and Viles, J.H., Involvement of a lysine residue in the N-terminal Ni2+ and Cu2+ binding site of serum albumins. Comparison with Co2+, Cd2+, and Al3+. Eur. J. Biochem. 220, 193–200, 1994. 774. Gaze, D.C., Ischemia modiied albumin: A novel biomarker for the detection of cardiac ischemia, Drug Metab. Pharmacokinet. 24, 333–341, 2009. 775. Roy, D., Quiles, J., Gaze, D.C., et al., Role of reactive oxygen species on the formation of the novel diagnostic marker ischemia modiied albumin, Heart 92, 113–114, 2006. 776. Bar-Or, D., Rael, L.T., Bar-Or, R., et al., The cobalt-albumin binding assay: Insights into its mode of action, Clin. Chim. Acta 387, 120–127, 2008. 777. Roy, D. and Kaski, J.C., The ischemia-modiied albumin in relation to pacemaker and deibrillator implantation: The mechanism of IMA formation and the role of reactive oxygen species, Pacing Clin. Electrophysiol. 31, 643–644, 2008. 778. Hjortshøj, S., Dethlefsen, C., Kristensen, S.R., and Ravkilde, J., Kinetics of ischaemia modiied albumin during ongoing severe myocardial ischaemia, Clin. Chim. Acta 403, 114–120, 2009. 779. Roy, D., Quiles, J., Aldama, G., et al., Ischemia modiied albumin for the assessment of patients presenting to the emergency department with acute chest pain but normal or non-diagnostic 12-lead electrocardiograms and negative troponin T, Int. J. Cardiol. 97, 297–301, 2004. 780. Collinson, P.O., Biomarkers in angina, Scand. J. Clin. Lab. Invest. Suppl. 240, 86–92, 2005.

Albumin

175

781. Sharma, R., Gae, D.C., Pellerin, D., et al., Evaluation of ischemia-modiied albumin as a marker of myocardial ischemia in end-stage renal disease, Clin. Sci. (London) 113, 25–32, 2007. 782. Lee, Y.W., Kim, H.J., Cho, Y.H., et al., Application of albumin-adjusted ischemia modiied albumin index as an early screening marker for acute coronary syndrome, Clin. Chim. Acta 384, 24–27, 2007. 783. Borderie, D., Allanore, Y., Meune, C., et al., High ischemia-modiied albumin concentration relects oxidative stress but not myocardial involvement in systemic sclerosis, Clin. Chem. 50, 2190–2193, 2004. 784. van der Zee, P.M., Verberne, H.J., van Straalen, J.P., et al., Ischemia-modiied albumin measurements in symptom-limited exercise myocardial perfusion scintigraphy relect serum albumin concentrations, Clin. Chem. 51, 1744–1746, 2005. 785. Refaai, M.A., Wright, R.W., Parvin, C.A., et al., Ischemia-modiied albumin increases after skeletal muscle ischemia during arthroscopic knee surgery, Clin. Chim. Acta 366, 264–268, 2006. 786. Lippi, G., Salvagno, G.L., Montagnana, M., et al., Inluence of physical exercise and relationship with biochemical variables of NT-pro-brain natriuretic peptide and ischemia modiied albumin, Clin. Chim. Acta 367, 175–180, 2006. 787. Falkensammer, J., Stojakovic, T., Huber, K., et al., Serum levels of ischemia-modiied albumin in healthy volunteers after exercise-induced calf-muscle ischemia, Clin. Chem. Lab. Med. 45, 535–540, 2007. 788. Gunduz, A., Turedi, S., Mentes, A., et al., Ischemia-modiied albumin in the diagnosis of acute mesenteric ischemia: A preliminary study, Am. J. Emerg. Med. 26, 202–205, 2008. 789. Talwalker, S.S., Bar Homme, M., Miller, J.J., and Elin, R.J., Ischemia modiied albumin, a marker of acute ischemic events, a pilot study, Ann. Clin. Lab. Sci. 38, 132–137, 2008. 790. Ukinc, K., Eminagaoglu, S., Ersoz, H.O., et al., A novel indicator of widespread endothelial damage and ischemia in diabetic patients: Ischemia-modiied albumin, Endocrine 36, 425–432, 2009. 791. Turk, A., Nuhaglu, I., Mentese, A., et al., The relationship between diabetic retinopathy and serum levels of ischemia-modiied albumin and malonaldehyde, Retina 31, 602– 608, 2011. 792. Bruschi, M., Mussante, L., Candiano, G., et al., Transitions of serum albumin in patients with glomerulosclerosis ‘in vivo’ characterization by electrophoretic titration curves, Electrophoresis 27, 2960–2969, 2006. 793. Musante, L., Bruschi, M., Candiano, G., et al., Characterization of oxidation end product of plasma albumin ‘in vivo’. Biochem. Biophys. Res. Commun. 349, 668–673, 2006. 794. Bruschi, M., Musante, L., Ghiggeri, G.M., et al., Comparative study of thermal stability of healthy and focal segmental glomerulosclerosis plasma albumin, J. Therm. Anal. Calorim. 87, 27–31, 2007. 795. Musante, L., Candiano, G., Petretto, A., et al., Active focal segmental glomerulosclerosis is associated with massive oxidation of plasma albumin, J. Am. Soc. Nephrol. 18, 799–810, 2007. 796. Bauman, A., Rothschild, M.A., Yallow, R.S., and Berson, S.A., Distribution and metabolism of I131 labeled human serum albumin in congestive heart failure with and without proteinuria, J. Clin. Invest. 34, 1359–1365, 1955. 797. Hallaba, E. and Drouet, J., Iodine-131 labeling of human serum albumin by the iodine monochloride and chloramine-T methods, Int. J. Appl. Radiat. Isot. 22, 46–49, 1971. 798. Peters, T.H., Plasma albumin, in The Plasma Proteins, ed. F. Putnam, Vol. 1, Academic Press, New York, 1975. 799. Johnson, A.M., Amino acids, peptides, and proteins, in Teitz Textbook of Clinical Chemistry and Molecular Diagnostics, 4th edn., eds. C.A. Burtis, E.R. Ashwood, and D.E. Bruns, Chapter 20, pp. 533–595, Saunders/Elsevier, St. Louis, MO, 2006.

176

Biotechnology of Plasma Proteins

800. Anraku, M., Kragh-Hansen, U., Kawai, K., et al., Validation of the chloramine-T induced oxidation of human serum albumin as a model for oxidative damage in vitro, Pharm. Res. 20, 684–692, 2003. 801. Gamsjäger, T., Brenner, L., Sitzwohl, C., and Weinstabl, C., Half-lives of albumin and cholinesterase patients, Clin. Chem. Lab. Med. 46, 1140–1142, 2008. 802. Ding, R., Frej, E., Fordanesh, M., et al., Pharmacokinetics of 5-aminoluoresceinalbumin, a novel luorescence marker of brain tumors during surgery, J. Clin. Pharmacol. 51, 672–678, 2011. 803. Matsusbita, S., Chuang, V.T.C., Kamazawa, M., et al., Recombinant human serum albumin dimer has high blood circulation activity and low vascular permeability in comparison with native human serum albumin, Pharm. Res. 23, 882–891, 2006. 804. McCurdy, T.R., Gataiance, S., Eltringham-Smith, L.J., and Shefield, W.P., A covalently linked recombinant albumin dimer is more rapidly cleared in vivo than are wild-type and mutant C34A albumin, J. Lab. Clin. Med. 143, 115–124, 2004. 805. Cirillo, M., Evaluation of glomerular iltration rate and of albuminuria/proteinuria, J. Nephrol. 23, 125–132, 2010. 806. Birn, H. and Christensen, E.I., Renal albumin absorption in physiology and pathology, Kidney Int. 69, 440–449, 2006. 807. Pollock, C.A. and Poronnik, P., Albumin transport and processing by the proximal tubule: Physiology and pathophysiology, Curr. Opin. Nephrol. Hypertens. 16, 359–364, 2007. 808. Comper, W.D., Haraldsson, B., and Deen, W.M., Resolved: Normal glomeruli ilter nephrotic levels of albumin, J. Am. Soc. Nephrol. 19, 427–432, 2008. 809. Clavant, S.P. and Comper, W.D., Urinary clearance of albumin is critically determined by its tertiary structure, J. Lab. Clin. Med. 142, 372–384, 2003. 810. Wilder, R.L., Yuen, C.C., Subbarao, B., et al., Tritium (3H) radiolabeling of protein A and antibody to high speciic activity: Application to cell surface radioimmunoassays, J. Immunol. Meth. 28, 255–266, 1979. 811. Dottavio-Martin, D. and Ravel, J.M., Radiolabeling of proteins by reductive alkylation with [14C] formaldehyde and sodium cyanoborohydride, Anal. Biochem. 87, 562–565, 1978. 812. Rennke, H.G. and Venkatachalan, M.A., Glomerular permeability of macromolecules. Effect of molecular coniguration on the fractional clearance of uncharged dextran and neutral horseradish peroxidase in the rat, J. Clin. Invest. 63, 713–717, 1979. 813. Asgeirnsson, D., Venturoli, D., Fries, E., et al., Glomerular sieving of three neutral polysaccharides, polyethylene oxide and bikunin in rat. Effect of molecular size and conformation, Acta Physiol. 191, 237–246, 2007. 814. Lundström, K.E., Blom, A., Johnsson, E., et al., High glomerular permeability of bikunin despite similarity in change and hydrodynamic size to serum albumin, Kidney Int. 51, 1053–1058, 1997. 815. The Nephrotic Syndrome, eds. J.S. Cameron and R.J. Glassrock, Marcel Dekker, New York, 1988. 816. Hrick, D.E. and Smith, M.D., Proteinuria and the Nephrotic Syndrome, Year Book Medical Publishers, Chicago, IL, 1986. 817. Haraldsson, B., Nyström, J., and Deen, W.M., Properties of the glomerular barrier and mechanisms of proteinuria, Physiol. Rev. 88, 451–487, 2008. 818. Kaysen, G.A., Plasma composition in the nephrotic syndrome, Am. J. Nephrol. 13, 347– 359, 1993. 819. Nishimura, M., Shimada, J., Ito, K., et al., Acute arterial thrombosis with antithrombin III deiciency in nephrotic syndrome: Report of a case, Surg. Today 30, 663–666, 2000. 820. Charlesworth, J.A., Gracey, D.M., and Pussell, B.A., Adult nephrotic syndrome: Nonspeciic strategies for treatment, Nephrology (Carlton) 13, 45–50, 2008.

Albumin

177

821. Anderson, C.L., Chaudhury, C., Kim, J., et al., Perspective—FcRn transports albumin: Relevance to immunology and medicine, Trends Immunol. 27, 343–348, 2006. 822. Gurbaxani, B., Mathematical modeling as accounting: Predicting the fate of serum proteins and therapeutic monoclonal antibodies, Clin. Immunol. 122, 121–124, 2007. 823. Anderson, J.T. and Sandlie, I., The versatile MHC class I-related FcRn protects IgG and albumin from degradation: Implications for development of new diagnostics and therapeutics, Drug. Metab. Pharmacokinet. 24, 318–332, 2009. 824. Kim, J., Hayton, W.L., Robinson, J.M., and Anderson, C.L., Kinetics of FcRnmediated recycling of IgG and albumin in human: Pathophysiology and therapeutic implications using a simpliied mechanism-based model, Clin. Immunol. 122, 146– 155, 2007. 825. Baker, K., Qiao, S.W., Kuo, T., et al., Immune and non-immune functions of the (not so) neonatal Fc receptor, FcRn, Semin. Immunopathol. 31, 223–236, 2009. 826. Molineux, G., The design and development of pegilgratim (PEG-rmetHuG-CSF, Neulasta), Curr. Pharm. Des. 10, 1235–1244, 2004. 827. Gao, Z., Bai, G., Chen, J., et al., Development, characterization, and evaluation of a fusion protein of a novel glucagon-like peptide-1 (GLP-1) analog and human serum albumin in Pichia pastoris, Biosci. Biotechnol. Biochem. 73, 688–694, 2009. 828. Xu, J., Okada, S., Tan, L., et al., Human growth hormone expressed in tobacco cells as an arabinoglactan–protein fusion glycoprotein has a prolonged serum life, Transgenic Res. 19, 849–867, 2010. 829. Longsworth, L.G., Shedlovsky, T., and Macinnes, D.A., Electrophoretic patterns of normal and pathological human blood serum and plasma, J. Exp. Med. 70, 399–413, 1939. 830. Martin, N.H. and Morris, R., The albumin/globulin ratio: A technical study, J. Clin. Pathol. 2, 64–66, 1949. 831. Lerner, A.B. and Barnum, C.P., A new rapid method of the determination of serum albumin and globulin by ultraviolet absorption, Arch. Biochem. 11, 505–514, 1946. 832. Sandor, G., Serum Proteins in Health and Disease, Physiological and pathological changes in serum albumin levels, Chapter 8, pp. 623–634, Williams & Wilkins, Baltimore, MD, 1966. 833. Alberghina, D., Giannetto, C., Vazzana, I., et al., Reference intervals for total protein concentration, serum protein fractions, and albumin/globulin ratios in clinically healthy dairy cows, J. Vet. Diagn. Invest. 23, 111–114, 2011. 834. Raj, M.A., Reddy, A.G., Reddy, A.R., and Adilaxmamma, K., Effect of dietary vanaspati alone and in combination with stressors on sero-biochemical proile and immunity in white leghorn layers, Toxicol. Int. 18, 31–34, 2011. 835. Mehrabi, Z., Firouzhakhsh, F., and Jafarpour, A., Effects of dietary supplementation of symbiotic on growth performance, serum biochemical parameters and carcass composition in rainbow trout (Oncorhynchus mykiss) ingerlings, J. Anim. Physiol. Anim. Nutr. (Berl) 96(3), 474–481, 2011. 836. Johnson, A.M., Amino acids, peptides, and proteins, in Tietz Clinical Chemistry and Molecular Diagnostics, 4th edn., eds. C.A. Burtis, E.R. Ashwood, and D.E. Bruns, Chapter 20, pp. 533–595, Saunders/Elsevier, St. Louis, MO, 2006. 837. Wallace, J.F., Pugia, M.J., Lott, J.A., et al., Multisite evaluation of a new dipstick for albumin, protein, and creatinine, J. Clin. Lab. Anal. 15, 231–235, 2001. 838. Croal, B.L., Mutch, W.J., Clark, B.M., et al., The clinical application of a urine albumin: Creatinine ratio point of care device, Clin. Chim. Acta 307, 15–21, 2001. 839. Matteucci, E. and Giampietro, O., Point-of-care testing in diabetes care, Mine Rev. Med. Chem. 11, 178–184, 2011. 840. Doumas, B.T. and Peters, T., Jr., Serum and urine albumin: A progress report on their measurement and clinical signiicance, Clin. Chim. Acta 258, 3–20, 1997.

178

Biotechnology of Plasma Proteins

841. Bates, R.G. and Paabo, M., Measurement of pH, in Handbook of Biochemistry and Molecular Biology, 4th edn., eds. R.L. Lundblad and F. MacDonald, pp. 709–713, CRC Press, Boca Raton, FL, 2010. 842. Klotz, I.M. and Walker, F.M., Spectral changes in some dye ions and their relation to the protein error in indicators, J. Phys. Colloid Chem. 51, 666–680, 1947. 843. Doumas, B.T., Watson, W.A., and Biggs, H.G., Albumin standards and the measurement of serum albumin with bromocresol green, Clin. Chim. Acta 31, 87–96, 1971. 844. Mehrotra, R., Duong, U., Jiwakanon, S., et al., Serum albumin as a predictor of mortality in peritoneal dialysis: Comparisons with hemodialysis, Am. J. Kidney Dis. 58, 418–428, 2011. 845. Snozek, C.L., Saenger, A.K., Greipp, P.R., et al., Comparison of bromocresol green and agarose protein electrophoresis for quantitation of serum albumin in multiple myeloma, Clin. Chem. 53, 1099–1103, 2007. 846. Meng, Q.H. and Krahn, J., Lithium heparinised blood-collection tubes give falsely low albumin results with an automated bromocresol green method in haemodialysis patients, Clin. Chem. Lab. Med. 46, 396–400, 2008. 847. Suzuki, Y., Inluence of pH for the determination of serum albumin by a dye-binding method in the presence of a detergent, Anal. Sci. 24, 1061–1064, 2008. 848. Klotz, I.M., Ligand–receptor complexes: Origin and development of the concept, J. Biol. Chem. 279, 1–12, 2004. 849. Carroll, B., Use of dyestuffs for determining the activity of proteolytic enzymes, Science 111, 387–388, 1950. 850. Tang, L.X., Rowell, F.J., and Cumming, R.H., A rapid homogeneous assay for subtilisin, Anal. Lett. 29, 2085–2095, 1996. 851. Lee, S.-H., Miller, J.S., Moon, J.J., and West, J.L., Proteolytically degradable hydrogels with a luorogenic substrate for studies of cellular proteolytic activity and migration, Biotechnol. Prog. 21, 1736–1741, 2005. 852. Sapan, C.V., Lundblad, R.L., and Price, N.C., Colorimetric protein assay techniques, Biotechnol. Appl. Biochem. 29, 99–108, 1999. 853. Farrance, I., Dennis, P.M., Gibson, P.J., and Biegler, B., A comparative study of commercial human and bovine albumin preparations, Ann. Clin. Biochem. 15, 31–36, 1978. 854. Lundblad, R.L. and Price, N.C., Determination of protein concentration in the manufacture and characterization of biopharmaceuticals with a note on validation process, Bioprocess Int. January 2004, 1–7, 2004. 855. Jenzano, J.W., Hogan, S.L., Noyes, C.M., et al., Comparison of ive techniques for the determination of protein content in mixed human saliva, Anal. Biochem. 159, 370–376, 1986. 856. Löf, A.L., Gustafsson, G., Novak, V., et al., Determination of total protein in highly puriied factor IX concentrates, Vox Sang. 63, 172–177, 1992. 857. Yang, H., Zhang, Y., and Poeschi, U., Quantiication of nitrotyrosine in nitrated proteins, Anal. Bioanal. Chem. 387, 879–886, 2010. 858. Mortstedt, H., Jeppsson, M.C., Ferrari, G., et al., Strategy for the identiication and detection of multiple oxidative modiications within protein applied on persulfateoxidized hemoglobin and human serum albumin, Rapid Commun. Mass Spectrom. 25, 327–340, 2011. 859. Nayak, R. and Knapp, D.R., Matrix-free LDI mass spectrometry platform using patterned nanostructured gold ilm, Anal. Chem. 82, 7772–7778, 2010. 860. Esteban-Fernández, D., Scheler, C., and Linscheid, M., Absolute protein quantitation by LC-ICP-MS using MeCat peptide labeling, Anal. Bioanal. Chem. 401, 657–666, 2011. 861. Wu, F., Sun, D., Wang, N., et al., Comparison of surfactant-assisted shotgun methods using acid-labile surfactants and sodium dodecyl sulfate for membrane proteome analysis, Anal. Chim. Acta 698, 36–43, 2011.

Albumin

179

862. Shin, S., Yang, H.J., and Kim, J., Effects of temperature on ultrasound-assisted tryptic protein digestion, Anal. Biochem. 414, 125–130, 2011. 863. Tanaka, N., Nagasaka, K., and Komatsu, Y., Selected reaction monitoring by linear iontrap mass spectrometry can effectively be applicable to simultaneous quantiication of multiple peptides, Biol. Pharm. Bull. 34, 135–141, 2011. 864. Edsall, G. and Wyman, L., Human serum albumin as a stabilizing agent for Schick toxin, Am. J. Public Health Nations Health 34, 365–367, 1943. 865. Levine, L., Ipsen, J., and McComb, J.A., Adult immunization—Preparation and evaluation of a combined luid tetanus and diptheria toxoid for adult use, Am. J. Hyg. 73, 20–35, 1961. 866. Zhi, J., Teller, S.B., Satoh, H., et al., Inluence of human serum albumin content in formulations on the bioequivalency of interferon α-2a given by subcutaneous injection in healthy male volunteers, J. Clin. Pharmacol. 35, 281–284, 1995. 867. Hawe, A. and Friess, W., Physiochemical characterization of the freezing behavior of mannitol–human serum albumin formulation, AAPS Pharm Sci. Tech. 7, 94, 2006. 868. Hawe, A. and Friess, W., Stabilization of a hydrophobic recombinant cytokine by human serum albumin, J. Pharmaceut. Sci. 96, 2987–2999, 2007. 869. Warren, D.J., Nordlund, M.S., and Paus, E., Formulation of immunoassay calibrators in pasteurized albumin can signiicantly enhance their durability, J. Immunol. Meth. 35, 145–147, 2010. 870. Weinberg, J.M., Lipotoxicity, Kidney Int. 70, 1560–1566, 2006. 871. Solbu, M.D., Jenssen, T.G., Eriksen, B.O., and Toft, I., Changes in insulin sensitivity, renal function, and markers of endothelial dysfunction in hypertension—The impact of microalbuminuria: A 13-year follow up study, Metabolism 58, 408–415, 2009. 872. Odhiambo, A., Perlman, D.H., Huang, H., et al., Identiication of oxidative posttranslational modiication of serum albumin in patients with idiopathic pulmonary arterial hypertension and pulmonary hypertension of sickle cell anemia, Rapid Commun. Mass Spectrom. 21, 2195–2203, 2007. 873. Matsuda, R., Anguizola, J., Joseph, K.S., and Hage, D.S., High-performance afinity chromatography and the analysis of drug interactions with modiied proteins: Binding of gliclazide with glycated human serum albumin, Anal. Bioanal. Chem. 401, 2811–2819, 2011. 874. Liggins, J. and Furth, A.J., Role of protein-bound carbonyl groups in the formation of advanced glycation endproducts, Biochim. Biophys. Acta 1361, 123–130, 1997. 875. Yatscoff, R.W., Tevaarwerk, G.J.M., and MacDonald, J.C., Quantiication of nonenzymatically glycated albumin and total serum protein by afinity chromatography, Clin. Chem. 303, 446–449, 1984. 876. Rohovec, J., Machmeyer, T., Alme, S., and Peters, J.A., The structure of the sugar residue in glycated human serum albumin and its molecular recognition by phenylboronate, Chem. Eur. J. 9, 2193–2199, 2003. 877. Watkins, N.G., Neglia-Fisher, C.I., Dyer, D.G., et al., Effect of phosphate on the kinetics and speciicity of glycation of proteins, J. Biol. Chem. 262, 7207–7212, 1987. 878. Stamler, J.S., Jaraki, O., Osborne, J., et al., Nitric oxide circulates in mammalian plasma primarily as an S-nitroso adduct of serum albumin, Proc. Natl. Acad. Sci. USA 89, 7674– 7677, 1992. 879. Yang, Y. and Loscalzo, J., S-Nitrosoprotein formation and localization in endothelial cells, Proc. Natl. Acad. Sci. USA 102, 117–122, 2005. 880. Arteel, G.E., Briviba, K., and Sigs, H., Protection against peroxynitrite, FEBS Lett. 445, 226–230, 1999. 881. Dahm, C.C., Moore, K., and Murphy, M.P., Persistent S-nitrosation of complex I and other mitochondrial membrane proteins by S-nitrosothiols but not nitric oxide or peroxynitrite. Implications for the interaction of nitric oxide ith mitochondria, J. Biol. Chem. 281, 10056–10065, 2006.

180

Biotechnology of Plasma Proteins

882. Tochinskii, I.M. and Metzer, D.M., in Sulfur in Proteins, The chemical properties of SH groups. Sulfhydryl Reagents, Chapter 1, pp. 3–71, Oxford/Pergammon, New York, NY, 1981. 883. Xu, G. and Chance, M.R., Radiolytic modiication of sulfur-containing amino acid residues in model peptides: Fundamental studies for protein footprinting, Anal. Chem. 77, 2437–2449, 2005. 884. Kettenhofen, N.J. and Wood, M.J., Formation, reactivity, and detection of protein sulfenic acids, Chem. Res. Toxicol. 23, 1633–1646, 2010. 885. Rehder, D.S. and Borges, C.R., Cysteine sulfenic acid as an intermediate in disulide bond formation and nonenzymatic protein folding, Biochemistry 49, 7748–7755, 2010. 886. Shaw, B.F., Schneider, G.E., Bilgicer, B., et al., Lysine acetylation can generate highly charged enzymes with increased resistance toward irreversible inactivation, Protein Sci. 17, 1446–1455, 2008. 887. Glocker, M.O., Borchers, C., Fiedler, W., et al., Molecular characterization of surface topology in protein tertiary structures by amino-acylation and mass spectrometric peptide mapping, Bioconjug. Chem. 5, 583–590, 1994. 888. Means, G.E., Reductive alkylation of amino groups, Meth. Enzymol. 47, 469–478, 1977. 889. Fenaille, F., Guy, P.A., and Tabet, J.-C., Study of protein modiication by 4-hydroxy2-nonenal and other short chain aldehydes analyzed by electrospray ionization tandem mass spectrometry, J. Am. Soc. Mass Spectrom. 14, 215–226, 2003. 890. Bucala, R. and Cerami, A., Characterization of antisera to the addition product formed by the nonenzymatic reaction of 16α-hydroxyestrone with albumin, Mol. Immunol. 20, 1289–1292, 1983. 891. Lohmann, W., Hayen, H., and Karst, U., Covalent protein modiication by reactive drug metabolites using online electrochemistry/liquid chromatography/mass spectrometry, Anal. Chem. 80, 9714–9719, 2008. 892. Shinar, E., Navok, T., and Chevion, M., The analogous mechanisms for enzymatic inactivation induced by ascorbate and superoxide in the presence of copper, J. Biol. Chem. 258, 14778–14783, 1983. 893. Zhu, B.Z., Antholine, W.E., and Frei, B., Thiourea protects against copper-induced oxidative damage by formation of a redox-inactive thiourea–copper complex, Free Radic. Biol. Med. 32, 1333–1338, 2002. 894. Liu, M., Hou, J., Huang, L., et al., Site-speciic proteomics approach for study of protein S-nitrosylation, Anal. Chem. 82, 7160–7168, 2010. 895. Landino, L.M., Koumas, M.T., Mason, C.E., and Alston, J.A., Ascorbic acid reduction of microtubule protein disulides and its relevance to protein S-nitrosylation assays, Biochem. Biophys. Res. Commun. 340, 347–352, 2006. 896. Giustarini, D., Dalle-Donne, I., Colombo, R., et al., Is ascorbate able to reduce disulide bridges? A cautionary note, Nitric Oxide 19, 252–258, 2008. 897. Gebicki, J.M., Nauser, T., Domazou, A., et al., Reduction of protein radicals by GSH and ascorbate: Potential biological signiicance, Amino Acids 39, 1131–1137, 2010. 898. Abello, N., Kerstjens, H.A., Postma, D.S., and Bischoff, R., Protein tyrosine nitration: Selectivity, physicochemical and biological consequences, denitration, and proteomics methods for the identiication of tyrosine-nitrated proteins, J. Proeome Res. 8, 3222– 3238, 2009. 899. Ogaswara, Y., Mukai, Y., Togawa, T., et al., Determination of plasma thiol bound to albumin using afinity chromatography and high-performance liquid chromatography with luorescence detection: Ratio of cysteinyl albumin as a possible biomarker of oxidative stress, J. Chromatogr. B 845, 157–163, 2007. 900. Sengupta, S., Webbe, C., Majors, A.K., et al., Relative roles of albumin and ceruloplasmin in the formation of homocystine, homocysteine–cysteine-mixed disulide and cystine in circulation, J. Biol. Chem. 276, 46896–46904, 2001.

Albumin

181

901. Li, Y., Yan, X.-P., Chen, C., et al., Human serum albumin-mercurial species interactions, J. Proteome Res. 6, 2277–2286, 2007. 902. Kirkman, C.R., Albertini, R.J., Sweeney, L.M., and Gagas, M.C., 1,3-Butanedione I. Review of metabolism and the implications to human health and assessment, Crit. Rev. Toxicol. 40, 1–11, 2010. 903. Natsch, A., Gfeller, H., Kuhn, F., et al., Chemical basis for the extreme skin sensitization potency of (E)-4-(ethoxymethylene)-2-phenyloxazol-5(4H)-one, Chem. Res. Toxicol. 23, 1913–1920, 2010. 904. Narazaki, R. and Otagiri, M., Covalent binding of a bucillamine derivative with albumin in sera from healthy subjects and patients with various diseases, Pharm. Res. 14, 351–354, 1997.

5

Plasma Immunoglobulins

It is necessary to have clarity on the deinitions when discussing immunoglobulins and their complex interactions in humoral immunology and cellular immunology. A list of deinitions as understood within the current work is given in Table 5.1. It is also useful to have a brief orientation to the several immunoglobulins found in human plasma and some of their characteristics (Table 5.2). Immunoglobulins comprise some one-third of the total plasma protein, with immunoglobulin G (IgG) as the major component.1 The readers are directed to several early reviews for a discussion on the history of gamma (γ)-globulins and the development of the current nomenclature.2–4 Figure 5.1 shows a schematic drawing of IgG, emphasizing more on the function than on the chemical structure. The IgG molecule can be divided into two important functional regions.5 The variable regions of the light chain and the heavy chain at the aminoterminal form a structure, which is recognized as the antigenic determinant or epitope; this structure is called the paratope. At the carboxyl terminal, the Fc domain binds various receptors that serve for transport across the epithelial surface or are present on the effector cells that are responsible for phagocytosis or antibody-dependent cell-mediated cytotoxicity. Limited proteolysis has been useful in the study of the structure and the function of immunoglobulins and, as described later, was used on early immune serum globulin (ISG) preparations to reduce adverse reactions. Papain or trypsin cleaves the IgG molecule above the hinge region, resulting in two Fab fragments, each of which contains the paratope that can bind the antigenic determinant; thus, Fab is monovalent. Pepsin cleaves the IgG below the hinge region, preserving the disulide bonds between the two heavy chains and thus yielding the bivalent F(ab)2′. These derivatives lack the heavy chain and therefore cannot interact with effector cells; they can neutralize pathogens (bacteriostatic) but cannot mobilize the cytotoxic mechanisms to destroy pathogens (bacteriocidal). Fab and F(ab)2′ fragments are useful for imaging tumors and for delivering chemotherapeutic agents to tumor sites. The F(ab)2′ material has been successfully used as a therapeutic product (Gamma-Venin) for 40 years. The F(ab)2′ product is also known as 5S intravenous immunoglobulin (IVIG) as a measure of its sedimentation constant and has a shorter half-life (approximately 2 days).6 The major commercial immune globulin is γ-globulin, better known as immunoglobulin G (IgG), which is available as an intramuscular immune globulin (IMG), as an IVIG, and, more recently, as a subcutaneous product (SCIG). Only small amounts of that product may be given by intramuscular injection, and this technique is limited to high-titer immune globulins.7,8 There has been only limited development of the members of immunoglobulin classes, other than IgG, as therapeutic products. There has been some interest in a therapeutic concentrate of immunoglobulin M (IgM).9,10 A humanized monoclonal IgM, panobacumab, has been evaluated for pharmacokinetics and safety in patients with Pseudomonas aeruginosa pneumonia.11

183

184

Biotechnology of Plasma Proteins

TABLE 5.1 Immunology Definitions Term Active Immunization Antibody

Antigen

Antigenic Determinant

Anti-idiotypic Antibody

Autoantibody

Allogeneic Serum Autologous Serum

Bactericidal Bacteriostatic Biotherapy

Definition and Comment

References

The process of establishing immunity by vaccinating with an immunogen. A protein in the body that is either naturally occurring or elaborated in response to an antigenic stimulus that reacts with a speciic antigen. An antibody may be secreted in the blood or lymphatic luid. A substance that binds to an antibody or T-cell receptor. An antigen is frequently considered equivalent to an immunogen. Also known as epitope, it is a speciic structure in an immunogen that binds to a speciic antibody containing a paratope directed against that determinant/epitope. A determinant may be either conformation or linear in nature. A conformational determinant is based on the three-dimensional structure of the immunogen, while a linear determinant is based on a sequence of monomer units within a polymeric immunogen. A conformational determinant may also be known as a discontinuous epitope, while a linear determinant is also known a continuous epitope. A conformational epitope is frequently lost on the denaturation of a protein. An antibody directed against the idiotopes in a speciic antibody. Some interest has been evinced in the use of anti-idiotypic antibodies as vaccines. An antibody directed against the normal constituents of an organism; autoantibodies are responsible for autoimmune diseases. Serum obtained from a dissimilar individual of the same species. Serum obtained from the same individual (per Oxford Dictionary of English Language, 2004), although it is frequently used in place of homologue to indicate serum from the same species. Bacteria or pathogen destroyed (“killed”) frequently by phagocytosis. An agent that prevents bacteria or pathogen from reproducing. A term introduced to distinguish the use of biological substances, such as blood, serum, and tissue extracts, as therapeutics from chemical therapeutics. Currently, it is used to differentiate the use of biologics, such as monoclonal antibodies, from chemotherapy.

[406] [407]

[407]

[412–417]

[408–411]

[418–420]

[421–423] [424–426]

[427] [428–430] [407,431–433]

185

Plasma Immunoglobulins

TABLE 5.1 (Continued) Immunology Definitions Term Camelid

Cellular Immunology

Convalescent Serum Hapten

Heterologous Serum

Homologous Serum Humoral Immunology

Hyperimmune Globulin

Idiotope Idiotype Immunogen

Definition and Comment

References

A term used to describe a unique antibody protein found in the sera of members of the Camelidae family. This antibody lacks the light chain and has been expressed as a recombinant protein. Immunological phenomena based on the functions of cells of lymphoid tissues such as B lymphocytes and T lymphocytes in biological tissues and luids. Specialized cells in cellular immunology include dendritic cells and natural killer cells. It should be noted that the differentiation between cellular immunology and humoral immunology is somewhat artiicial. Convalescent serum is obtained from individuals recovering from speciic infectious diseases. A substance which, by itself, is an immunogen but can react with an antibody. A hapten, such as dinitrophenol, is immunogenic when combined with a protein. Serum usually obtained from different species (but reference was found using heterologous to distinguish from autologous serum). Serum obtained from the same species. Immunological reactions involving proteins, such as antibodies and lectins and other biological macromolecules, occurring in the various physiological luids. Hyperimmune globulins can be regarded as a current example of products developed from convalescent serum. Hyperimmune globulin preparations can be regarded as a puriied immunoglobulin product containing a high amount of antibody directed against a speciic pathogen. Starting plasma can be derived from convalescent individuals, donors screened for speciic immunoglobulin reactivity, or immunized individuals. Hyperimmune globulin preparations may also be achieved by the addition of humanized monoclonal antibodies to an immune globulin preparation. An epitope found (usually) within the variable region of an antibody. The collection of idiotopes found within a speciic antibody molecule. A substance that can react against components of cellular immunity (B cells and T cells) and also react with the products of the responses of cellular immunity. Immunogen structure/design is of considerable interest in vaccine development. It is also known as a complete antigen.

[434–436]

[64]

[437–439] [440,441]

[442,443]

[444] [445]

[142,295,446]

[407,447] [407–409] [412,448–450]

(continued)

186

Biotechnology of Plasma Proteins

TABLE 5.1 (Continued) Immunology Definitions Term

Definition and Comment

References

Immunogenic

The property of being an immunogen.

Immune Globulin

A generic name given to preparations of IgG that are derived from plasma. Such preparations may be intravenous immune globulins (IVIG) or intramuscular immunoglobulins. There was early oral administration of immune globulins.

Immunity

The mechanism(s) by which an organism is enabled to resist disease.

Immunology

The study of immunity.

Immunomodulation

The modiication of the immune response; used most frequently in cancer therapeutics but does have a broader range of applications.

[407,454,455]

Immunotherapy

Immunotherapy is a term originally used to describe the use of active or passive immunization and was later modiied to include any therapeutic approach to the suppression or enhancement of the immune system, including the use of cells, genes, and gene products including proteins and microRNA products.

[407,456–460]

Intramuscular Immune Globulin

A therapeutic preparation of plasma immune globulins (γ-globulins) given by intramuscular injection. Early problems with intravenous use of immune globulins made intramuscular injection the preferred route of administration. While intramuscular injection has been largely supplanted by intravenous methods, intramuscular can be an effective strategy.

[461,462]

Intravenous Immune Globulin (IVIG)

A therapeutic preparation of plasma immune globulin (γ-globulins) administered directly into the venous circulation (parenteral drug). These preparations were not readily available until some 30 years after the irst use of immune globulins.

[138,463–466]

Immunization

The process of establishing immunity.

Monoreactive Antibody

An antibody that reacts with a single antigen/epitope.

Passive Immunization

The process of establishing immunity by the transfer of antibody from the donor to the recipient. In the case of IVIG, there are many donors.

[471–473]

Paratope

The structure within the Fab portion of an immunoglobulin that recognizes a speciic epitope on an immunogen. An antibody that can react with more than one antigen. A polyreactive antibody is not to be confused with a polyclonal antibody/polyclonal antibody product. A monoclonal antibody may be polyreactive.

[474,475]

Polyreactive antibody

[407,451,452] [24,453]

[407] [407]

[467] [238,468–470]

[476–478]

187

Plasma Immunoglobulins

TABLE 5.1 (Continued) Immunology Definitions Term Reagin

Serum

Serum Therapy

a

Definition and Comment

References

Reagin is a term used originally to describe all antibodies and was later restricted to describe antibodies in allergic and hypersensitivity reactions; it has been largely supplanted by the term IgE to describe these antibodies. Reagin is still used to describe a speciic antibody in syphilis. The fluid obtained after the clotting of blood. Serum is not plasma; plasma is obtained from blood most frequently after the collection of whole blood in an anticoagulant such as citrate or heparin. Serum has a slightly lower protein concentration than plasma, and the two materials should not be confused with each other. That said, it is essential to go to the original study to make sure that serum is not confused with plasma. The use of serum as a vehicle for passive immunization. Convalescent serum was preferred for serum therapy.a

[407,479–481]

[485,486]

[482–484]

The term serum therapy has been used more recently to describe the use of autologous serum or umbilical cord serum for the treatment of various corneal problems.487–489

An IgM-enriched immunoglobulin has been suggested to be useful in Gram-negative sepsis.12 Bovine milk has been suggested as a source of immunoglobulin A (IgA).13,14 Immunoglobulin E (IgE) is also known as the reaginic antibody4 and is a therapeutic target,15 although engineered antibodies of IgE class have therapeutic potential.16 There are two therapeutic concepts driving the use of native and recombinant immunoglobulins. The irst is passive immunization, which is the transfer of immunity from a donor to a recipient, and it is considered as an approach to neutralize viral and microbial pathogens. The second is immunomodulation or immunotherapy, where therapeutic immunoglobulins modulate cellular immunity inluencing pathologies, such as inlammatory neuropathies. This separation of therapeutic concepts is based more on the history and is somewhat artiicial, as it proves most dificult to separate the immunomodulation effects from the antipathogen effects in passive immunization.17 Passive immunization, also known earlier as passive acquired immunity, is achieved by the transfer of an immunoglobulin from a donor to a recipient; in the case of IVIG, there are many donors. Active immunization, also known as active acquired immunity, is achieved by vaccination with a suitable immunogen. Passive immunization was also known as serum therapy, as early work used both homologous and heterologous sera. Though there were adverse reactions, such as serum sickness, these therapeutic approaches were quite useful. The readers are directed to several sources

188

Biotechnology of Plasma Proteins

TABLE 5.2 Immunoglobulins Found in Human Plasmaa MWb (kDa)

t1/2 (Days)

(mg/dL)c,d

Comment

IgG

150

24

700–1600e

The most common immunoglobulin in plasma. Major function is protecting against pathogens and facilitating opsonization of viral and bacterial pathogens.f The action can be either bacteriostatic or bacteriocidal or both. IgG in plasma comprises various subclasses: IgG1, IgG2, IgG3, and IgG4. These subclasses have various characteristics. IgG1 is the major subclass with somewhat less IgG2; there are relatively small amounts of IgG3 and IgG4. It comprises 80+% plasma immunoglobulin.

IgA

160

6

70–400

It provides protection against pathogens, primarily viral or bacterial,f with emphasis on mucosal secretion (mucosal immunity). Subclasses IgA1, IgA2. The plasma form is a monomer, while the mucosal secretory product is a dimer or tetramer with an associated J chain of molecular mass of 15 kDa containing one N-linked glycan. More IgA is synthesized per day than IgG, but approximately one-half is catabolized in the liver and the remainder is transported to external secretion.

IgM

900

5

30–360

IgD

190

2.8g

N/A

It provides early protectionf against pathogens; a polymeric form (pentamer; closed ring including ive IgG and a J chain) of IgG, it is considered to be an early immunoglobulin with function linked to IgD; IgM may be the most ancient of the immunoglobulins. As with IgA, there is a secretory form of unknown function. Though the precise function is not known, increasing evidence suggests (1) relationship with IgM and (2) important in immunomodulation. Phylogenetic research suggests that IgD is an early immunoglobulin like IgM.

Immunoglobulin

189

Plasma Immunoglobulins

TABLE 5.2 (Continued) Immunoglobulins Found in Human Plasmaa Immunoglobulin IgE

a b c d e f

g

MWb (kDa)

t1/2 (Days)

(mg/dL)c,d

175

2.4g

N/A

Comment Associated with mast cells, this is responsible for allergic responses such as hypersensitivity and anaphylaxis.

The material in this table has been adapted from several sources including.490–497 Approximate values. Concentration in plasma (reference interval). Adult values given. Further data on IgG subclass values can be obtained from.498 Resistance is frequently used to describe the innate ability of an organism to neutralize (resist) a pathogen or toxin produced in a disease (adapted from499). The above resistance refers to the ability of an immunoglobulin to neutralize (bacteriostatic) or kill (bacteriocidal) pathogens or toxins. From Salonen et al.500

that review the early work on passive immunization.18–21 A puriied immune globulin, ISG,22 was developed as part of the Cohn fractionation process for human plasma during World War II.23 ISG was considered a by-product of the fractionation process, as the goal was to produce albumin.23 There were some early clinical studies using this material, most notably on the prophylaxis and prevention of measles.24,25 General Kendrick’s book on blood programs during World War II23 is very interesting and the lessons learned are still as valuable as they were some 50 years ago. Initially, the ISG preparations derived from human plasma were of limited use, as there were signiicant adverse reactions from early intravenous use, which in turn limited the intramuscular administration of ISG. There was also limited understanding of the market for polyvalent ISGs, as primary immune deiciency was described only in 1952.26–28 Early studies on the stability of ISG prepared by the Cohn fractionation process demonstrated fragmentation on storage.29,30 This fragmentation, which resulted in the reduction of anticomplement activity, was attributed to the presence of plasmin in the ISG preparation; the anticomplement activity had been suggested to be responsible for the adverse reactions associated with the parenteral use of the ISG.31 Barandun and coworkers31 reviewed the various approaches developed to reduce the anticomplement activity of ISG. The use of pepsin or plasmin resulted in derivatives with reduced or no anticomplement activity while retaining the ability to form complexes with antigens. An immunoglobulin fraction treated with pepsin did have a markedly reduced half-life (10–14 hours)32 and loss of Fc function (see Figure 5.1), while an immunoglobulin preparation treated with plasmin had a longer half-life (18–20 days)32 with a variable retention of Fc function.33 Exposure to low pH (pH 4.0) had a variable effect on the anticomplement activity but did retain full Fc function. Milt Mozen and colleagues at Cutter in Berkeley34 subjected an ISG to limited reduction with dithiothreitol followed by carboxyamidomethylation with the

190

Biotechnology of Plasma Proteins

Fab fragment Fab 2 Hinge Fc

+ Fc fragments

3

1

IgG Mild reduction F(ab)′2

SH SH Fab′

FIGURE 5.1 A schematic presentation of IgG and its derivatives. The IgG molecule is a homodimer of a heterodimer; only a single chain is shown to represent the heterodimer unit. The IgG molecule is divided into two major functional domains: the Fab region, which contains the paratope that recognizes the antigen (epitope), and the Fc domain, which binds to speciic receptors (Fc receptors) on target cells such as macrophages.5,519 The primary site of glycosylation of IgG is in the Fc domain (1); only a single site is shown in this igure, but glycosylation occurs on the heavy chain in both of the heterodimers. Glycosylation can occur in the Fab region but is considered unusual. It is also possible to have glycosylation only on one heavy chain in an Fc fragment. Papain (2) cleaves IgG above the “hinge” region to yield Fab fragment, which is a monomer of a heterodimer fragment, which is monovalent in binding antigen.520 Pepsin (3) cleaves IgG below the “hinge” region to yield the F(ab)′2 fragment, which is a dimer of the heterodimer Fab; the Fab′ monomer can be obtained from F(ab)′2 by mild reduction.521 Cleavage of IgG similar to that observed with pepsis can be obtained with matrix metalloproteinases and may be important for IgG function in the extravascular space.522 This illustration does not consider the issue of immunoglobulin subtypes.523

reduction of anticomplement activity and retention of Fc function. The problem was resolved through the development and implementation of changes in the manufacturing strategies,27,35 resulting in the current IVIG products.36,37 IVIG is deined as a sterile preparation of immunoglobulins prepared from human plasma. The 2004 edition of the British Pharmacopoeia38 deines human normal immunoglobulin as “a liquid or freeze-dried preparation containing immunoglobulins, mostly IgG … prepared from pooled material from not fewer than 1000 donors.” The use of a larger pool (10,000–40,000 donors) is suggested by other workers.39 The larger the pool, the more diverse is the antibody population. The current IVIG products40 are approved for use in the treatment of primary immune deiciencies, prevention of cytomegalovirus infection after bone marrow transplantation, prevention of hepatitis B infection after liver transplantation,41–45 and treatment of selected secondary immune deiciencies.46,47 However, the off-label use of IVIG is extensive.47–49 Darabi and coworkers50 reviewed the use of IVIG at Massachusetts

Plasma Immunoglobulins

191

General Hospital for the calendar year 2004. The major use was for chronic neuropathy treatment, with considerably less use for primary immune deiciency (primary hypogammaglobulinemia) treatment. This poses a problem, as supporting documentation for such off-label use is not always available.49–55 Studies with careful clinical end points do show the effectiveness of IVIG in chronic inlammatory demyelinating polyradiculoneuropathy (CIDP),56 and there is ongoing work on the development of molecular markers (biomarkers) to measure therapeutic responses.57 Unsworth and Wallington58 discuss the risk of IVIG use, suggesting restricted use to those applications where there is evidence-based effectiveness. The off-label use of IVIG is further complicated by possible differences between various commercial preparations of IVIG.59–63 The use of IVIG in immunomodulation is discussed later, although it is dificult to separate the humoral activity of IVIG from its activity in cellular immunology.40,64 The ability of antibodies to neutralize and destroy pathogens, such as bacteria and viruses (humoral activity), is the better understood function of IVIG—the application in infectious diseases. However, even the application of IVIG in infectious diseases is quite complicated beyond bacteriostatic and bactericidal activities.40 There is a solid technology that does make it possible to identify the antibodies in an IVIG preparation that react with speciic pathogens as well as establish the integrity of the Fc domain necessary for phagocytosis. IVIG products from different manufacturers (and hence potentially different manufacturing processes) exhibit differences in activity against some pathogens.65–70 These differences appear to be based on the fractionation process rather than the geographical source, as Matejtschuk and coworkers70 evaluated an IVIG product (Vigam) prepared either from plasma sourced in the United States or plasma sourced in the United Kingdom; the difference then was based on the geographical source rather than on the manufacturing process. The products were evaluated for composition (e.g., aggregates, monomer, dimer, IgG class, etc.) and reactivity against various viral and bacterial pathogens. There were modest differences between the U.S. product and the U.K. product; however, as the authors note, with large enough donor pools, there should be no differences in eficacy on the basis of the geographical source of the plasma. That is not to say that there is no potential for a difference in the quality of the IgG, resulting from the geographical source, and it is possible to make advantageous use of such a difference in the development of a therapeutic product. In the case of H1N1 virus, the use of plasma (convalescent plasma) obtained from the pandemic sites permitted the manufacture of a hyperimmune product.71–73 Changes in donor population, vaccination policies, and a decrease in a speciic disease prevalence can affect the quality attributes of the donor plasma pool, which in turn will affect the product attributes.74,75 Additional discussion on the use of IVIG in passive immunization is presented later. The manufacture of IVIG by necessity (large number of donors) starts with a large volume of plasma (500–2500 L, depending on the capacity of the manufacturing facility), and the crude starting material for actual manufacturing (e.g., the Cohn Fraction II + III) is obtained from the starting plasma after a prior separation step, such as cryoprecipitation.76,77 The cryoprecipitation step is essential and is equivalent to the original Cohn I step.37 An alcohol precipitation step is then used to obtain a protein fraction equivalent to Cohn II + Cohn III (see Chapter 2). The supernatant

192

Biotechnology of Plasma Proteins

fraction from this step is taken on for the preparation of Cohn IV (source of antithrombin, α1-antitrypsin, factor IX) and Cohn V (source of albumin and plasma protein fraction). The crude immunoglobulin (II + III paste) can be either taken on directly for the manufacture of IVIG or stored frozen for subsequent use. At least four physical and/or chemical environmental transitions to the IgG molecule have occurred, prior to the speciic IVIG-manufacturing steps: irst, divalent cations are removed by an anticoagulant; second, the plasma is frozen; third, the plasma is allowed to slowly thaw into the cryoprecipitate slurry; and fourth, the plasma is precipitated with alcohol (20%–25% by volume, approximately 4 M) at pH 5.2 (−5°C). If the precipitate is frozen subsequent to processing, there is a ifth challenge to the protein. This is followed by solubilization of the paste and various fractionation steps, leading to the equivalent of an active pharmaceutical ingredient (API), which is then formulated to yield a inal drug product, which might be a liquid or a lyophilized solid.78–80 For a protein, this is the equivalent of being a lineman/ linebacker in American football for a number of years; as this is the author’s experience, he can attest to the fact that it is mostly functional but never quite the same as the original. The effect of alcohol on plasma proteins is discussed in Chapter 7, with speciic reference to the formation of latent forms of protein protease inhibitors. A consideration of the literature reveals little information on the effect of alcohol and/or freeze/ thaw cycles on immunoglobulin structure/function. Successive freeze/thaw cycles do promote aggregation.81–85 Recent studies by St-Amour and coworkers86 suggest that the conditions of alcohol fractionation (30% ethanol) can “activate” the cryptic autoreactive IgG present in the starting plasma; low pH (20,000 kDa (40 monomer units).431 There is a distribution of vWF multimers in plasma over this molecular weight range, which can be visualized with agarose cell electrohoresis.432–435 The synthesis of vWF is observed in endothelial cells and megakaryocytes; it is stored in the Weibel–Palade bodies in the endothelial cells and in α-granules in the platelets and is found in plasma and in the extracellular matrix.436 There are some small structural differences in platelet vWF and plasma vWF,437,438 but such differences do not appear to affect the function of either vWF in primary hemostasis.438 vWF is processed by a metalloprotein, ADAMTS-13 (a disintegrin and metalloproteinase with thrombospondin-like domains-13).439–442 Very large vWF multimers are found in the deiciency of ADAMTS-13, resulting in thrombotic thrombocytopenic purpura.443–448 The potential of a recombinant ADAMTS-13 for the treatment of thrombotic thrombocytopenic purpura is being considered.448–451 The incidence rate of thrombotic thrombocytopenic purpura has been determined;452 such information will help determine the feasibility of product development. von Willebrand’s disease (vWD), the absence of normal vWF function, was described by Erik von Willebrand early in the last century, but it was a complicated journey from the early clinical observations to the deinition of syndrome.453 vWD is heterogeneous on presentation, with bleeding ranging from mild to severe and difference in multimer distributions.410,454 Type I is a partial deiciency with modest clinical issues, and Type III is a total deiciency with severe consequences. Type II vWF is characterized by a loss of high-molecular-weight multimers. The short note by Marques and Fritsma454 is an excellent review of everything that most people ever need to know about vWD. The identiication and diagnosis of

Factor VIII and von Willebrand Factor

255

vWD have been a challenge for many years.455–459 Additional complication is provided by potential artifacts resulting from sample processing. Böhm and coworkers460 have observed a cold-induced loss of vWF in citrated whole blood and suggested that the loss of high-molecular-weight multimers may be the relection of vWF binding to glycoprotein Ib on the platelets. vWD is the most common of the various hemorrhagic diatheses, with an incidence ranging between 0.1% and 2%.454,461 The most severe form of vWD, type III, is quite rare. It is possible to use some of the low-purity factor VIII concentrates and cryoprecipitate to treat vWD, but a plasma product is available 462–466 and a recombinant product is in development.467–469 Given the relatively small number of type III vWD patients and the availability of established plasma-derived therapeutic products, it may be dificult to establish a business strategy for the recombinant product. It is possible to treat vWD with desmopressin (DDAVP; 1-desamino-8-D-arginine vasopressin),470–472 and it is also useful for diagnosis.473

ECONOMIC ISSUES IMPACTING THE HEMOPHILIA BUSINESS The factor VIII “business” is a mature business, and with an increasing number of providers with essentially functionally similar recombinant products, it can be suggested that the market will move more toward a commodity rather than toward a specialty product. With an increased number of providers, it is unlikely that the availability/pricing behavior suggested for recombinant factor VIII in 1994474 is possible now. Here, it was suggested that providers of recombinant products were not interested in supplying the lower-cost plasma-derived products for HIV-infected individuals even when this cohort might not beneit from the recombinant product. Evaluation of the factor VIII business is of critical importance in determining the investment in the development of new therapeutic products. This is of particular importance in the current medical landscape. Regardless of geography and/or national health policy, there is a limit to the amount of resources available to support health care.475 I commend Iserson’s paper475 that was published in 1992; it is obvious that we have not learned much in the past 20 years, as we face the same problems of resource allocation even today. The care for hemophilia must it into a larger program for health-care delivery and be subject to the same evolving trends represented by evidence-based medicine476 for decisions on therapeutic approaches. Hemophilia is an expensive disease both in inancial477 and human resources.478,479 The issue of the cost of factor VIII products477 has been a matter of concern for at least 40 years480,481 and interest in this issue has only increased with the advent of more sophisticated products. Between 1972/73 when Green480 and Lazerson481 published their observations and 1995, ultrahigh-purity and recombinant factor VIII concentrates (Table 6.1) became available at a much higher cost. Nauenberg and Sullivan474 reported that the annual cost of factor VIII therapy increased from $10,000 to $60,000 in 1991. It is argued (accurately) that these materials provided a much better QoL for the hemophiliac.482 In 1995, Tuddenham and Laffan483 asked the question as to whether the more advanced therapeutic products (which at that time contained

256

Biotechnology of Plasma Proteins

albumin as an excipient*) provided suficient advantage to justify increased cost. It is noted that since that time there is a more advanced factor VIII product, which was manufactured and formulated in the absence of exogenous albumin.†,484 The development of factor VIII therapeutics beyond the ultrapure plasma-derived products is driven by safety considerations based on potential viral contamination.483 There has been no documented transmission of HIV by a factor VIII concentrate for at least 20 years,‡,482,485 and current methods for screening blood donations have markedly reduced the risk of known pathogen transmission.486 While recombinant products would presumably provide zero-risk of pathogen transmission,482 it is dificult to establish the true cost-effectiveness of the ultrahigh-purity/recombinant products.487 Hay and coworkers487 do emphasize the importance of “personalized medicine” in treatment decisions. The issue of safety is confounded by other issues such as inhibitor development, which is discussed next. The economics of hemophilia care are somewhat complicated by discussions on the value of prophylaxis versus on-demand,488,489 with most recent data supporting the cost-effectiveness of prophylaxis.490 The paper by Brown and Aledort488 was a meeting report discussing methods for assessing the impact of various treatment regimens in hemophilia and the use of QoL metrics to measure effectiveness. Incorporation of QoL and pharmacoeconomic considerations will be required in the design of future clinical trials. While viral safety is the most visible issue, development of inhibitors is also of importance in assessing product safety.491 Gringeri491 discussed the continuing question as to whether there is increased inhibitor formation with recombinant factor VIII products. Immunogenicity is a problem with most protein biopharmaceuticals and can result from formulation.492–496 Patients view viral safety as the most important product attribute, but inhibitor development is also a strong consideration.497 An earlier study,498 which involved patients, providers, and pharmacists, provided a more complex picture that varied with the extent of the disease (moderate vs. severe). Recombinant factor VIII is the product of choice in developed countries§ with the potential of plasma-derived products available in other geographies.499–503 It is not clear that all geographies will produce the three major plasma products—albumin, factor VIII, and IVIG—and it is also clear that there are restraints on full self-suficiency.504 This short section on economics and factor VIII has not mentioned gene therapy. Pasturized plasma-derived human albumin is considered to be a safe product continually being evaluated (Chapter 4).544,545 † Notwithstanding Veder’s question as to whether factor VIII is a protein,116 factor VIII, plasma-derived or recombinant, is a protein, and a therapeutic containing factor VIII contains protein. ‡ The risk of hepatitis B transmission via blood transfusion in the United States is estimated to be approximately 1 in 300,000, and for both hepatitis C and HIV, transmission risk is greater than 1 in 1 million.486 Notwithstanding these impressive numbers, there is a perception that suficient risk still exists (risk is discussed in Chapter 2) and there is substantial ongoing work to approach zero risk.546–549 § Financial pressures on allocation of health-care resources do have an impact on products used in a given geography, but there is pressure to provide the recombinant products.535 Geographies where there is interest in plasma-derived therapeutic products other than factor VIII such as IVIG likely wish to have national self-suficiency, and it may be dificult to rationalize the use of recombinant factor VIII. For a general discussion.536 However, this was some time ago, and it is clear from the work in India, Brazil, and Iran that the developing world is moving into the plasma business (see Chapter 2). *

Factor VIII and von Willebrand Factor

257

This is, in part, a relection of the author’s skepticism and the dificulties in achieving a reasonable business model.55 However, to be fair, it is clear that full achievement of gene therapy would provide a solution to the problem of treating hemophilia on a global basis.505

CONCLUSIONS AND FUTURE DIRECTIONS* • A continued increase in the number of suppliers has the potential to describe factor VIII as a commodity product such as albumin. The same argument cannot be made for IVIG, relecting the large number of existing and potential indications. • It is unlikely that a modiication of factor VIII, for example, with poly(ethylene) glycol, is likely to provide a useful product. • It is unlikely that a factor VIII mimetic can be developed that would be competitive with existing factor VIII products. • Without a quantum leap in tissue-speciic vector delivery, it is unlikely that gene therapy will be successful. • Genotyping of previously untreated hemophilia A patients506–508 is critical to solving the factor VIII inhibitor problems. • Advances in the formulation for factor VIII products to function outside the “cold chain” are essential for use in many developing geographies. • Emphasis should be placed on drug delivery methods and devices to achieve the full promise of prophylaxis. While considerably more work is required, it is possible that the use of prophylaxis recommended for factor VIII inhibitor patients509 may reduce inhibitor development.510 Prophylaxis is demonstrated to be more effective than on-demand therapy.490 • Oral delivery of potentially hemostatic agents requires further investigation. The results obtained in the 1930s158,162 are solid and should not be disregarded simply on the basis of less-sophisticated analytical technologies.

REFERENCES 1. Haberichter, S.L., Jacobi, P., and Montgomery, R.R., Critical independent regions in VWF propeptide and mature VWF that enable VWF storage, Blood 101, 1384–1391, 2003. 2. Dong, J.-F., Moake, J.L., Nolasco, L., et al., ADAMTS-13 rapidly cleaves newly secreted ultralarge von Willebrand factor multimers on the endothelial surface under lowing conditions, Blood 100, 4033–4039, 2002. *

This discussion is concerned only with factor VIII. The deiciency of factor IX, hemophilia B, is discussed in Chapter 8. The hemophilia B business is not as large as hemophilia A as there are far fewer deicient individuals. Nevertheless, there are a number of “players” in the hemophilia B business and more players are considering entering the business. Factor IX is less complex than factor VIII, and gene therapy for hemophilia B has been somewhat more successful than that for hemophilia A. vWF deiciency is also a much smaller business opportunity than hemophilia A, and a number of therapeutic opportunities exist for a less-demanding deiciency.

258

Biotechnology of Plasma Proteins

3. Vesely, S.K., George, J.N., Lämmle, B., et al., ADAMTS13 activity in thrombotic thrombocytopenic purpura-hemolytic uremic syndrome: Relation to presenting features and clinical outcomes in a prospective cohort of 142 patients, Blood 102, 60–66, 2003. 4. Burnouf, T., Caron, C., Burkhardt, T., and Goudemand, E.J., Content and functional activity of von Willebrand factor in aphersis plasma, Vox Sang. 87, 27–33, 2004. 5. Ruggeri, Z.M., Von Willbrand factor, Curr. Opin. Hematol. 10, 142–149, 2003. 6. Weiss, H.J., Sussman, I.I., and Hoyer, L.W., Stabilization of factor VIII in plasma by the von Willebrand factor. Studies on posttransfusion and dissociated factor VIII and in patients with von Willebrand’s disease, J. Clin. Invest. 60, 390–404, 1977. 7. Wise, R.J., Dorner, A.J., Krane, M., et al., The role of von Willebrand factor multimers and propeptide cleavage in binding and stabilization of factor VIII, J. Biol. Chem. 266, 21948–21955, 1991. 8. Vlot, A.J., Koppelman, S.J., van den Berg, M.H., et al., The afinity and stoichiometry of binding of human factor VIII to von Willebrand factor, Blood 85, 3150–3157, 1995. 9. Bird, A., Isarangkura, P., Almagro, D., et al., Factor concentrates for haemophilia in the developing world, Haemophilia 4, 481–485, 1998. 10. O’Mahony, B. and Black, C., Expanding hemophilia care in developing countries, Semin. Thromb. Hemost. 31, 561–568, 2005. 11. Rogoff, E.G., Guirguis, H.S., Lipton, R.A., et al., The upward spiral of drug costs: A time series analysis of drugs used in the treatment of hemophilia, Thromb. Haemost. 88, 545–553, 2002. 12. van den Berg, H.M. and Fischer, K., Prophylaxis for severe hemophilia: Experience from Europe and the United States, Semin. Thromb. Hemost. 29, 49–54, 2003. 13. Carlsson, K.S., Höjgård, S., Lethagen, S., et al., Willingness to pay for on-demand and prophylactic treatment for severe haemophilia in Sweden, Haemophilia 10, 527–541, 2004. 14. Powell, J.S., Recombinant factor VIII in the management of hemophilia A: Current use and future promise, Ther. Clin. Risk Manag. 5, 391–402, 2009. 15. Franchini, M., Tagliaferri, A., and Mannucci, P.M., The management of hemophilia in elderly patients, Clin. Interv. Aging 2, 361–368, 2007. 16. Gianotten, W.L. and Heijnen, L., Haemophilia, aging and sexuality, Haemophilia 15, 55–62, 2009. 17. Mannucci, P.M., Schutgens, R.E., Santagostino, E., and Mauser-Bunschotgen, E.P., How I treat age-related morbidities in elderly persons with hemophilia, Blood 114, 5256–5263, 2009. 18. Green, K., Treatment strategies for adolescents with hemophilia: Opportunities to enhance development, Adolesc. Med. 10, 369–376, 1999. 19. Health Resources and Services Administration, HHS, Ricky Ray hemophilia relief program. Adoption of interim rule as inal rule with amendments, Fed. Regist. 66(226), 58667–58672, 2001. 20. Evatt, B.L., The natural evolution of haemophilia care: Developing and sustaining comprehensive care globally, Haemophilia 12(Suppl. 3), 13–21, 2006. 21. Klausner, A., ‘Adjustment’ in the blood fraction market, Nat. Biotechnol. 3, 119–125, 1985. 22. Hoffman, T., Frantantoni, J., and Murano, G., Clinical use of biologicals produced in continuous cell lines, Develop. Biol. Standard. (Basal) 70, 211–214, 1989. 23. Abrams, P., Analyzying biotech’s past, present, and future, Nat. Biotechnol. 11, 450– 451, 1993. 24. Aggarwal, S., What’s fueling the biotech engine—2009–2010, Nat. Biotechnol. 28, 1165–1171, 2010. 25. Radar, R.A., (Re)deining biopharmaceutical, Nat. Biotechnol. 26, 741–751, 2008.

Factor VIII and von Willebrand Factor

259

26. Werner, R.G., Biobusiness in the pharmaceutical industry, Arzneim. Forsch. 37, 1086– 1093, 1987. 27. Gitschier, J., Wood, W.I., Goralka, T.M., et al., Characterization of the human factor VIII gene, Nature 312, 326–330, 1984. 28. Eaton, D.L., Hass, P.E., Riddle, L., et al., Characterization of recombinant human factor VIII, J. Biol. Chem. 262, 3285–3290, 1987. 29. Toole, J.J., Knopf, J.L., Wozney, J.M., et al., Molecular cloning of a cDNA encoding human antihaemophilic factor, Nature 312, 342–347, 1984. 30. Kaufman, R.J., Wasley, L.C., and Dorner, A.J., Synthesis, processing, and secretion of recombinant human factor VIII expressed in mammalian cells, J. Biol. Chem. 263, 6352–6362, 1988. 31. Jiang, H., Wu, S.L., Karger, B.L., and Hancock, W.S., Characterization of the glycosylation occupancy and the active site in the follow-on protein therapeutic: TNK-tissue plasminogen activator, Anal. Chem. 82, 6154–6162, 2010. 32. Schiestl, M., Stangler, T., Torella, C., et al., Acceptable changes in quality attributes of glycosylated biopharmaceuticals, Nat. Biotechnol. 29, 310–312, 2011. 33. Hironaka, T., Furukawa, K., Esmon, P.C., et al., Comparative study of the sugar chains of factor VIII puriied from human plasma and from the culture media of recombinant baby hamster kidney cells, J. Biol. Chem. 267, 8012–8020, 1992. 34. Kumar, H.P.M., Hague, C., Haley, T., et al., Elucidation of N-linked oligosaccharide structures of recombinant human factor VIII using luorophore-assisted carbohydrate electrophoresis, Biotechnol. Appl. Biochem. 24, 207–216, 1996. 35. Sandberg, H., Almstedt, A., Brandt, J., et al., Structural and functional characteristics of the B-domain-deleted recombinant factor VIII protein, r-VIII SQ, Thromb. Haemost. 85, 93–100, 2001. 36. Kelly, B., Jankowski, M., and Booth, J., An improved manufacturing process for Xyntha/ ReFacto AF, Haemophilia 16, 717–725, 2010. 37. Kingdon, H.S. and Lundblad, R.L., An adventure in biotechnology: The development of haemophilia A therapeutics—From whole-blood transfusion to recombinant DNA to gene therapy, Biotechnol. Appl. Biochem. 35, 141–148, 2002. 38. Lee, C.A., The natural history of HIV disease in haemophilia, Blood Rev. 12, 135–144, 1998. 39. Josefson, D., Haemophilia patients launch action against Bayer over contaminated blood products, BMJ 326, 1286, 2003. 40. Alban, S., The ‘precautionary principle’ as a guide for future drug development, Eur. J. Clin. Invest. 35(Suppl. 1), 33–44, 2005. 41. Colombo, M., Mannucci, P.M., Carnelli, V., et al., Transmission of non-A, non-B hepatitis by heat-treated factor VIII concentrate, Lancet 2(8445), 1–5, 1985. 42. Orringer, D.P., Koury, M.J., Blatt, P.M., and Roberts, H.R., Hemolysis caused by factor VIII concentrates, Arch. Int. Med. 136, 1018–1020, 1976. 43. Tuddenham, E.G.D., Lane, R.S., Rotblat, F., et al., Response to infusions of polyelectrolyte fractionated human factor VIII concentrates in human haemophilia A and von Willebrand’s disease, Brit. J. Haematol. 52, 259–267, 1982. 44. Morini, M., Rafenelli, D., Filimberti, E., and Cinotti, S., Protein content and factor VIII complex in untreated, treated and monoclonal factor VIII concentrates, Thromb. Res. 56, 169–178, 1989. 45. Farrugia, A., Evolving perspectives in product safety for haemophilia, Haemophilia 8, 236–243, 2002. 46. AuBuchon, J.P., Birkmeyer, J.D., and Busch, M.P., Safety of the blood supply in the United States: Opportunities and controversies, Ann. Intern. Med. 127, 904–909, 1997. 47. Pereira, A., Cost-effectiveness analysis and the selection of blood products, Curr. Opin. Hematol. 7, 420–425, 2000.

260

Biotechnology of Plasma Proteins

48. Gringeri, A., Bypassing agent regimes and costs for prophylaxis in patients with inhibitors, Haemophilia 15, 1336–1337, 2009. 49. Odeyemi, I.A. and Dane, A.M., Optimising immune tolerance induction strategies in the management of haemophilia patients with inhibitors: A cost-minimization analysis, Curr. Med. Res. Opin. 25, 239–250, 2009. 50. Pokras, S.M., Petrilla, A.A., Weatherall, J., and Lee, W.C., The economics of inpatient on-demand treatment for haemophilia with high-responding inhibitors: A US retrospective data analysis, Haemophilia 18, 248–260, 2012. 51. Viiala, N.O., Larsen, S.R., and Rasko, J.E., Gene therapy for hemophilia: Clinical trials and technical tribulations, Semin. Thromb. Hemost. 25, 81–92, 2009. 52. Mátrai, J., Chuah, M.K.L., and VandenDriessche, T., Preclinical and clinical progress in hemophilia gene therapy, Curr. Opin. Hematol. 17, 387–392, 2010. 53. Perrus, I., Chuah, M., and VandenDriessche, T., Gene therapy strategies for hemophilia: Beneits versus risks, J. Gene Med. 12, 797–809, 2010. 54. High, K.A., Gene therapy for hemophilia: A long and winding road, J. Thromb. Haemost. 9(Suppl. s1), 2–11, 2011. 55. Philippidis, A., Developing a balanced business model for gene therapy, Human Gene Ther. 22, 645–646, 2011. 56. Högy, B., Keinecke, H.O., and Borte, M., Pharmacoeconomic evaluation of immunoglobulin treatment in patients with antibody deiciencies from the perspective of the German statuatory health insurance, Eur. J. Health Econ. 6, 24–29, 2005. 57. Beauté, J., Levy, P., Millet, V., et al., Economic evaluation of immunoglobulin replacement in patients with primary antibody deiciencies, Clin. Exp. Immunol. 160, 240–245, 2010. 58. Lewitt, P.A., Rezai, A.R., Leehey, M.A., et al., AAV2-GAD gene therapy for advanced Parkinson’s disease: A double blind, sham-surgery controlled randomized trial, Lancet Neurol. 10, 309–319, 2011. 59. Lundblad, R.L. and Davie, E.W., The activation of antihemophilic factor (Factor 8) by activated Christmas factor (activated Factor 9), Biochemistry 3, 1720–1725, 1964. 60. Varadi, K. and Hemker, H.C., Kinetics of the formation of the factor X activating enzyme of the blood coagulation system, Thromb. Res. 8, 303–317, 1976. 61. Blosteinb, M.D, Furie, B.C., Rajotte, I., and Furie, B., The Gla domain of factor IXa binds to factor VIIIa in the tenase complex, J. Biol. Chem. 278, 31297–31302, 2003. 62. Yuan, Q.P., Walke, E.N., and Sheehan, J.P., The factor IXa heparin-binding exosite is a cofactor interactive site: Mechanism for antithombin-independent inhibition of intrinsic tenase by heparin, Biochemistry 44, 3615–3625, 2005. 63. Perdekamp, M.T., Rubenstein, D.A., Jesty, J., and Hultin, M.B., Platelet factor V supports hemostasis in a patient with an acquired factor V inhibitor, as shown by prothrombinase and tenase assays, Blood Coagul. Fibrinolysis 17, 593–597, 2006. 64. Tuddenham, E.G., Ways to bypass a blocked tenase complex, Thromb. Haemost. 95, 1–2, 2006. 65. Takeyama, M., Nogami, K., Saenko, E.L., et al., Protein S down-regulates factor Xase activity independent of activated protein C: Speciic binding of factor VIIIa to protein S inhibits interactions with factor IXa, Brit. J. Haematol. 143, 409–420, 2008. 66. Subbaiah, P.V., Bajwa, S.S., Smith, C.M., and Hanahan, D.J., Interactions of the components of the prothrombinase complex, Biochim. Biophys. Acta 444, 131–146, 1976. 67. Barhoover, M.A., Orban, T., Bukys, M.A., and Kalafatis, M., Cooperative regulation of the activity of factor Xa within prothrombinase by discrete amino acid regions from factor Va heavy chain, Biochemistry 47, 12835–12843, 2008. 68. Bradford, H.N., Micucci, J.A., and Krishnaswamy, S., Regulated cleavage of prothrombin by prothrombinase: Repositioning a cleavage site reveals the unique kinetic behavior of the action of prothrombinase on its compound substrate, J. Biol. Chem. 285, 328–338, 2010.

Factor VIII and von Willebrand Factor

261

69. Undas, A., Siudak, E., Brummel-Ziedins, K., et al., Prothrombinase formation at the site of microvascular injury and aspirin resistance: The effect of simvastatin, Thromb. Res. 125, 283–285, 2010. 70. Lee, C.J., Wu, S., Eun, C., and Pedersen, L.G., A revisit to the one form kinetic model of prothrombinase, Biophys. Chem. 149, 28–33, 2010. 71. van Diegjien, G., Tans, G., Rosing, J., and Hemker, H.C., The role of phospholipid and factor VIIIa in the activation of bovine factor X, J. Biol. Chem. 256, 3433–3442, 1981. 72. Johnson, D.J., Langdown, J., and Huntington, J.A., Molecular basis of factor IXa recognition by heparin-activated antithrombin revealed by a 1.7 Å structure of the ternary complex, Proc. Natl. Acad. Sci. USA 107, 645–650, 2010. 73. Neurath, H., Limited proteolysis and zymogen activation, in Proteases and Biological Control, eds. E. Reich, D.B. Rifkin, and E. Shaw, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1975. 74. Agyei-Owusu, K. and Leeper, F.J., Thiamin diphosphate in biological chemistry: Analogues of thiamin diphosphate in studies of enzymes and riboswitches, FEBS J. 276, 2905–2916, 2009. 75. Mandl, J., Szarka, A., and Bánhegyi, G., Vitamin C: Update on physiology and pharmacology, Brit. J. Pharmacol. 157, 1097–1110, 2009. 76. Marí, M., Morales, A., Colell, A., et al., Mitochondrial glutathione, a key survival antioxidant, Antioxid. Redox. Signal. 11, 2685–2700, 2009. 77. Singh, R. and Mozzarelli, A., Cofactor chemogenomics, Meth. Mol. Biol. 575, 93–122, 2009. 78. Chang, J., Jin, J., Lollar, P., et al., Changing residue 338 in human factor IX from arginine to alanine causes an increase in catalytic activity, J. Biol. Chem. 273, 12089–12094, 1998. 79. Stürzbecher, J., Kopetzki, E., Bode, W., and Hopfner, K.-P., Dramatic enhancement of the catalytic activity of coagulation factor IXa by alcohols, FEBS Lett. 412, 295–300, 1997. 80. Lundblad, R.L. and Roberts, H.R., The acceleration by polylysine of the activation of factor X by factor IXa, Thromb. Res. 25, 319–329, 1982. 81. Fay, P.J. and Koshibu, K., The A2 subunit of factor VIIIa modulates the active site of factor IXa, J. Biol. Chem. 272, 19049–19054, 1998. 82. Fay, P.J., The A1 and A2 subunits of factor VIIIa synergistically stimulate factor IXa catalytic activity, J. Biol. Chem. 274, 15401–15406, 1999. 83. Kolkman, J.A. and Mertens, K., Insertion loop 256–268 in coagulation factor IX restricts enzymatic activity in the absence but not the presence of factor VIII, Biochemistry 39, 7398–7405, 2000. 84. Scheilinger, F., Dockal, M., Rosing, J. and Kerschbaumer, R.J., Enhancement of the enzymatic activity of activated coagulation factor IX by anti-factor IX antibodies, J. Thromb. Haemost. 6, 315–322, 2008. 85. Zögg, T. and Brandstetter, H., Structural basis of the cofactor- and substrate-assisted activation of human coagulation factor IXa, Structure 17, 1669–1678, 2009. 86. McMahan, S.A. and Burgess, R.R., Mapping protease suscetibility sites on the Escherichia coli transcription factor δ70, Biochemistry 38, 12424–12431, 1999. 87. Sajnani, G., Pastrana, M.A., Dynin, I., et al., Scapie prion protein structural constraints obtained by limited proteolysis and mass spectrometry, J. Mol. Biol. 382, 88–98, 2008. 88. Kazanov, M.D., Igarashi, Y., Eroshkin, A.M., et al., Structural determinants of limited proteolysis, J. Proteome Res. 10, 3642–3651, 2011. 89. Sohn, J., Grant, R.A., and Sauer, R.T., Allostery is an intrinsic property of the protease domain of DegS. Implications for enzyme function and evolution, J. Biol. Chem. 285, 34039–34047, 2010.

262

Biotechnology of Plasma Proteins

90. Corey, D.R., Willett, W.S., Coombs, G.S., and Craik, C.S., Trypsin speciicity increased through substrate-assisted catalysis, Biochemistry 34, 11521–11527, 1995. 91. Dall’Acqua, W. and Carter, P., Substrate-assisted catalysis: Molecular basis and biological signiicance, Protein Sci. 9, 1–9, 2000. 92. Bihoreau, N., Sauger, A., Yon, J.M., and Van de Pol, H., Isolation and characterization of different activated forms of factor VIII, the human antihemophilic factor, Eur. J. Biochem. 185, 111–118, 1989. 93. Aronson, D.L. and Chang, P., Activated factor VIII in therapeutic preparations: Analysis by ultracentrifugation, Vox Sang. 71. 142–149, 1996. 94. D’Amici, G.M., Timperio, A.M., Gevi, F., et al., Recombinant clotting factor VIII concentrates: Heterogeneity and high-purity evaluation, Electrophoresis 31, 2730–2739, 2010. 95. Kaufman, R.J., Wasley, L.C., and Dorner, A.J., Synthesis, processing, and secretion of recombinant human factor VIII expressed in mammalian cells, J. Biol. Chem. 263, 6352–6362, 1988. 96. Pittman, D.D., Tomkinson, K.N., and Kaufman, R.J., Post-translational requirements for functional factor V and factor VIII secretion in mammalian cells, J. Biol. Chem. 269, 17329–17337, 1994. 97. Ganz, P.R., Tackaberry, E.S., Palmer, D.S., and Rock, G., Human factor VIII from heparinized plasma Puriication and characterization of a single-chain form, Eur. J. Biochem. 170, 521–528, 1988. 98. Rock, G. and Palmer, D.S., Accumulative effect of DDAVP and heparin in increasing plasma factor VII levels, Vox Sang. 41, 56–60, 1981. 99. Palmer, D.S., Rosborough, D., Perkins, H., et al., Characterization of factors affecting the stability of frozen heparinized plasma, Vox Sang. 65, 258–270, 1993. 100. Krachmalnicoff, A. and Thomas, D.P., The stability of factor VIII in heparinized plasma, Thromb. Haemost. 49, 224–227, 1983. 101. Cumming, A., Wensley, R.T., Winkelman, L., and Lane, R.S., A simple plasma anticoagulantexchange method to increase the recovery of factor VIII in therapeutic concentrates, Vox Sang. 58, 264–269, 1990. 102. Weinstein, M.J., Fulcher, C.A., Chute, L.E., and Zimmerman, T.S., Apparent molecular weight of puriied human factor VIII procoagulant protein compared with puriied and plasma factor VIII procoagulant protein antigen, Blood 62, 1114–1117, 1983. 103. Andersson, L.-O., Forsman, N., Huang, K., et al., Isolation and characerization of human factor VIII: Molecular forms in commercial factor VIII concentrate, cryoprecipitate, and plasma, Proc. Natl. Acad. Sci. USA 83, 2979–2983, 1986. 104. Kemball-Cook, G., Bevan, S.A., and Barrowcliffe, T.W., Factor VIII heavy chain polypeptides in plasma and concentrates, Brit. J. Haematol. 76, 80–87, 1990. 105. Kemball-Cook, G., Edwards, S.J., and Barrowcliffe, T.W., Proteolysis of factor VIII heavy chain polypeptides in plasma and concentrates, Brit. J. Haematol. 78, 222–228, 1991. 106. Jankowski, M.A., Patel, H., Rouse, J.C., et al., Deining ‘full-length’ recombinant factor VIII: A comparative structural analysis, Haemophilia 13, 30–37, 2007. 107. Xu, W., Chen, J., Yamasaki, G., et al., Lectin binding assays for in-process monitoring of sialylation in protein production, Mol. Biotechnol. 45, 248–256, 2010. 108. Michnick, D.A., Pittman, D.D., Wise, R.J., and Kaufman, R.J., Identiication of individual tyrosine sulfation sites with factor VIII required for optimal activity and eficient thrombin cleavage, J. Biol. Chem. 269, 20095–20102, 1994. 109. Seibert, C. and Sakmar, T.P., Toward a framework for sulfoproteomics: Synthesis and characerization of sulfotyrosine-containing peptides, Biopolymers 90, 459–470, 2008. 110. Fay, P.J., Chavin, S.I., Malone, J.E., et al., The effect of carbohydrate depletion on procoagulant activity and in vivo survival of highly puriied human factor VIII, Biochim. Biophys. Acta 800, 152–158, 1984.

Factor VIII and von Willebrand Factor

263

111. Kosloski, M.P., Miclea, R.D., and Balu-Iyer, S., Role of glycosylation in conformational stability, activity, macromolecular interaction and immunogenicity of recombinant human factor VIII, AAPS J. 11, 424–431, 2009. 112. Aly, A.M., Higuchi, M., Kasper, C.K., et al., Hemophilia-A due to mutations that create new N-glycosylation sites, Proc. Natl. Acad. Sci. USA 89, 4933–4937, 1992. 113. Bovenschen, N., Rijken, D.C., Havekes, L.M., et al., The B domain of coagulation factor VIII interacts with the asialoglycoprotein receptor, J. Thromb. Haemost. 3, 1257–1265, 2005. 114. Ingram, G.I., The history of haemophilia, J. Clin. Pathol. 29, 469–479, 1976. 115. Bloom, A.L., Inherited disorders of blood coagulation, in Haemostasis and Thrombosis, eds. A.L. Bloom and D.P. Thomas, Chapter 20, pp. 321–370, Churchhill Livingstone, Edinburgh, Scotland, 1981. 116. Veder, H.A., Is the antihaemophilic globulin a protein?, Nature 209, 202, 1966. 117. Lundblad, R.L., unpublished observations, 1989. 118. Veder, H.A., Further puriication of the antihaemophilic factor (AHF), Thromb. Diath. Haemorrh. 16, 738–751, 1966. 119. Hershgold, E.J., Davison, A.M., and Janszen, M.E., Isolation and some chemical properties of human factor VIII (antihemophilic factor), J. Lab. Clin. Med. 77, 185–205, 1971. 120. Lundblad, R.L. and Bradshaw, R.A., Addressing product improvement using chemical modiication in biopharmaceutical manufacture. A case study in blood coagulation factor VIII, Bioprocess International, September, 2006. 121. Butennas, S. and Mann, K.G., Blood coagulation, Biochemistry (Mosc.) 67, 3–12, 2002. 122. Lacroix-Desmazes, S., Nararrete, A.M., André, S., et al., Dynamics of factor VIII interactions determine its immunological fate in hemophilia, Blood 112, 240–249, 2008. 123. Terraube, V., O’Donnell, J.S., Jenkins, P.V., et al., Factor VIII and von Willebrand factor interaction: Biological, clinical and therapeutic importance, Haemophilia 16, 3–13, 2010. 124. Morini, M., Mannucci, P.M., Tenconi, P.M., et al., Pharmacokinetics of monoclonallypuriied and recombinant factor VIII in patients with severe von Willebrand disease, Thromb. Haemost. 70, 270–272, 1993. 125. Deitcher, S.R., Tuller, J., and Johnson, J.A., Intranasal DDAVP induced increases in plasma von Willebrand factor alter the pharmacokinetics of high-purity factor VIII concentrates in severe haemophilia A patients, Haemophilia 5, 88–95, 1999. 126. Björkman, S. and Berntorp, E., Pharmacokinetics of coagulation factors: Clinical relevance for patients with haemophilia, Clin. Pharmacokinet. 40, 815–832, 2001. 127. Fischer, K., Pendu, R., van Schooten, C.J., et al., Models for prediction of factor VIII half-life in severe haemophiliacs: Distinct approaches for blood group O and non-O patients, PLoS One 4, e6745, 2009. 128. Furlan, M., Von Willebrand factor: Molecular size and functional activity, Ann. Hematol. 72, 341–348, 1996. 129. Budde, U. and Schneppenheim, R., Von Willebrand factor and von Willebrand disease, Rev. Clin. Exp. Hematol. 5, 335–368, 2001. 130. Fricke, W.A. and Wong Yu, M.-Y., Characterization of von Willebrand factor in Factor VIII concentrates, Am. J. Hematol. 31, 41–45, 1989. 131. Vlot, A.J., Koppelman, S.J., Meijers, J.C., et al., Kinetics of factor VIII-von Willebrand factor association, Blood 87, 1809–1816, 1996. 132. Lin, Y., Yang, X., and Chevrier, M.C., Relationships between factor VIII: Ag and factor VIII in recombinant and plasma-derived factor VIII concentrates, Haemophilia 10, 459–469, 2004. 133. Aronson, D.L. and Chang, P., Ultracentrifugal analysis of factor VIII and von Willebrand factor in therapeutic preparations, Vox Sang. 69, 8–13, 1995.

264

Biotechnology of Plasma Proteins

134. Haberichter, S.L., Shi, Q., and Montgomery, R.R., Regulated release of vWF and FV IIII and the biologic implications, Pediatr. Blood Cancer 46, 547–553, 2006. 135. van den Biggelaar, M., Meijer, A.B., Voorberg, J., and Mertens, K., Intracellular cotraficking of factor VIII and von Willebrand factor type 2N variants to storage granules, Blood 113, 102–109, 2009. 136. Saenko, E.L., Loster, K., Josic, D., and Sarafanov, A.G., Effect of von Willebrand factor and its proteolytic fragments on kinetics of interaction between the light and heavy chains of human factor VIII, Thromb. Res. 96, 343–354, 1999. 137. Li, X. and Gabriel, D.A., The physical exchange of factor VIII (FVIII) between von Willebrand factor and activated platelets and the effect of FVIII B-domain on platelet binding, Biochemistry 36, 10760–10767, 1997. 138. Ahmad, S.S., Scandura, J.M., and Walsh, P.N., Structural and functional characterization of platelet receptor-mediated factor VIII binding, J. Biol. Chem. 275, 13071–13081, 2000. 139. Ahmad, S.S., London, F.S., and Walsh, P.N., Binding studies of the enzyme (factor IXa) with the cofactor (factor VIIIa) in the assembly of the factor-X activating complex on the activated platelet surface, J. Thromb. Haemost. 1, 2348–2355, 2003. 140. Novakovic, V.A., Cullinan, D.B., Wakabayashi, H., et al., Membrane-binding properties of the factor VIII C2 domain, Biochem. J. 435, 187–196, 2011. 141. Meems, H., Meijer, A.B., Cullinan, D.B., et al., Factor VIII C1 domain residues Lys2092 and Phe 2093 contribute to membrane binding and cofactor activity, BIood 114, 3938– 3946, 2009. 142. Obergfell, A., Sturm, A., Speer, C.P., et al., Factor VIII is a positive regulator of platelet function, Platelets 17, 448–453, 2006. 143. Hsu, T.-C., Pratt, K.P., and Thompson, A.R., The factor VIII C1 domain contributes to platelet binding, Blood 111, 200–208, 2008. 144. Pabinger, I. and Cihan, A., Biomarkers and venous thromboembolism, Arterioscler. Thromb. Vasc. Biol. 29, 332–336, 2009. 145. Kempton, C.L., Hoffman, M., Roberts, H.R., and Monroe, D.M., Platelet heterogeneity: Variation in coagulation complexes on platelet subpopulations, Arterioscler. Thromb. Vasc. Biol. 25, 861–866, 2005. 146. Panteleev, M.A., Ananyeva, N.M., Greco, N.J., et al., Two subpopulations of thrombinactivated platelets differ in their binding of the components of the intrinsic factor X-activating complex, J. Thromb. Haemost. 3, 2545–2553, 2005. 147. Rosendaal, F.R., Smit, C., and Briet, E., Hemophilia treatment in historical perspective: A review of medical and social developments, Ann. Hematol. 62, 5–15, 1991. 148. Young, J.H., James Blundell (1790–1878), experimental physiologists and obstetrician, Med. Hist. 8, 159–169, 1964. 149. Hajdu, S.I., Blood transfusion from antiquity to the discovery of the Rh factor, Ann. Clin. Lab. Sci. 33, 471–473, 2003. 150. Bause, G.S., The Blundell gravitator, Anesthesiology 110, 1416, 2009. 151. Lane, S., Haemorrhagic diathesis: The successful transfusion of blood, Lancet 35, 185– 188, 1840. 152. Hicks, J.B., Cases of transfusion with some remarks on a new method of performing the operation, Guys Hospital Reports, Series 3, 14, 1–14, 1869. 153. Duncan, J., On re-infusion of blood on primary and other amputations, Brit. Med. J. 1, 192–193, 1886. 154. Cotterill, J.M., Severe injury from dynamite, transfusion of blood, four times, recovery, Brit. Med. J. II, 630, 1886. 155. Mollison, P.L., The introduction of citrate as an anticoagulant for transfusion and of glucose as red cell preservative, Brit. J. Haematol. 108, 13–18, 2000. 156. Diamond, L.K., History of blood banking in the United States, J. Am. Med. Assoc. 193, 40–44, 1965.

Factor VIII and von Willebrand Factor

265

157. Eibl, M.M., History of immunoglobulin replacement, Immunol. Allergy Clin. North Am. 28, 737–764, 2008. 158. Eley, R.C., Green, A.A., and McKhann, C.F., The use of a blood coagulant extract from the human placenta in the treatment of hemophilia, J. Pediatr. 8, 135–147, 1936. 159. Burrows, W., Jordan-Burrows Textbook of Bacteriology, 15th edn., W.B. Saunders, Philadelphia, 1949. 160. Mortiz, F., Petersen, C.A., and Mill, C.F., A new method for accurately determining the clotting time of the blood, Arch. Int. Med. 32, 188–191, 1923. 161. Biggs, R. and Macfarlane, R.G., Human Blood Coagulation and Its Disorders, C.C. Thomas, Springield, IL, 1953. 162. Bendien, W.M. and van Crevald, S., Investigations on hemophilia, J. Dis. Child. 54, 713–725, 1937. 163. Petersen, L.C., Elm, T., Ezban, M., et al., Plasma elimination kinetics for factor VII are independent of its activation to factor VIIa and complex formation with plasma inhibitors, Thromb. Haemost. 101, 818–826, 2009. 164. Patek, A.J. and Stetson, R.P., Hemophilia: I. The abnormal coagulation time of the blood and its relation to the blood platelets, J. Clin. Invest. 15, 531–542, 1936. 165. Hedner, J., Factor VIIa and its potential therapeutic use in bleeding—Associated pathologies, Thromb. Haemost. 100, 557–562, 2008. 166. Overlack, A., Stumpe, K.O., Kolloch, R., et al., Antihypertensive effect of orally administered glandular kallikrein in essential hypertension. Results of a double blind study, Hypertension 3, I18–I21, 1981. 167. Addis, T., The effect of intravenous injections of fresh human serum and use of phosphated blood on the coagulation time of the blood in heriditary hemophila, Proc. Soc. Exp. Biol. Med. 14, 19–23, 1916. 168. Tullis, J.L., Blood Cells and Plasma Proteins; Their State in Nature, Academic Press, New York, 1953. 169. Cohn, E.J., Oncley, J.L., Strong, L.E., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. I: The Characterization of the protein fractions of human plasma, J. Clin. Invest. 23, 417–432, 1944. 170. Williams, J.W., Petermann, M.L., Colovos, G.C., et al., Chemical, clinical, and immunological studies on the products of human plasma fractionation. II: Electrophoretic and ultracentrifugal studies of solutions of human serum albumin and immune serum globulins, J. Clin. Invest. 23, 433–436, 1944. 171. Brand, E., Kassell, B., and Saidel, L.J., Chemical, clinical and immunological studies on the products of human plasma fractionation. III: Amino acid compositions of plasma proteins, J. Clin. Invest. 23, 437–444, 1944. 172. Ballou, G.A., Boyer, P.D., Luck, J.M., and Lum, F.G., Chemical, clinical, and immunological studies on the products of human plasma fractionation. V: The inluence of non-polar anions on the thermal stability of serum albumin, J. Clin. Invest. 12, 454–457, 1944. 173. Scatchard, G., Barchelder, A.C., and Brown, A., Chemical, clinical, and immunological studies on the products of human plasma fractionation. VI: The osmotic presssure of plasma and of serum albumin, J. Clin. Invest. 23, 458–464, 1944. 174. Enders, J.F., Chemical, clinical, and immunological studies on the products of human plasma fractionation. X: The concentrations of certain antibodies in globulin fractions derived from human blood plasma, J. Clin. Invest. 23, 510–530, 1944. 175. Stokes, J., Maris, E.P., and Gellis, S.S., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XI: The use of concentrated normal human serum gamm globulin (human immune serum globulin) in the prophylaxis and treatment of measles, J. Clin. Invest. 23, 531–540. 176. Ordman, C.W., Jennings, C.G., and Janeway, C.A., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XII: The use of concentrated

266

177. 178. 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. 194. 195. 196. 197.

Biotechnology of Plasma Proteins normal human serum gamma globulin (human immune serum globulin) in the prevention and attenuation of measles, J. Clin. Invest. 23, 541–549, 1944. Edsall, J.T., Ferry, R.M., and Armstrong, S.H., Chemical, clinical, and immunological studies on the products of human plasma fractionation. XV: The proteins concerned in the blood coagulation mechanism, J. Clin. Invest. 23, 557–565, 1944. Neurath, H., Dees, J.S., and Fox, H., The preparation and properties of human ibrinogen solutions, J. Urol. 49, 497–502, 1943. Chargaff, E. and Bendich, A., On the coagulation of ibrinogen, J. Biol. Chem. 149, 93–109, 1943. Neurath, H., The golden years of protein science, Protein Sci. 4, 1939–1943, 1995. Burnouf, T., Modern plasma fractionation, Transfus. Med. Rev. 21, 101–107, 2007. Mikaelsson, M., Nilsson, I.M., Vilhardt, H., and Wiechel, B., Factor VIII concentrate prepared from blood donors stimulated by intranasal administration of a vasopressin analogue, Transfusion 22, 229–233, 1982. Nilsson, I.M., Blombäck, M., Jorpes, E., et al., Von Willebrand’s disease and its correction with human plasma fraction 1–0, Acta Med. Scand. 159, 179–188, 1957. Pitney, W.R., Treatment of haemophilia A with fraction 1–0, Bibl. Haematol. 23,1314, 1965. Steinbuch, M., Precipitation methods in plasma fractionation, Vox Sang. 23, 92–106, 1972. Nilsson, I.M., Management of haemophilia in Sweden, Thromb. Haemost. 35, 510–521, 1976. Pool, J.G., Cryoprecipitate in the treatment of hemophilia, Calif. Med. 113, 66–67, 1970. Spotnitz, W.D., Mintz, P.D., Avery, N., et al., Fibrin glue from stored human plasma. An inexpensive and eficient method for local blood bank preparation, Am. Surg. 53, 460–462, 1987. Grifith, M.J., Biochemical characterization of the method M AHF process developed to reduce the risk of virus transmission, in Biotechnology and the Promise of Pure Factor VIII, ed. H.R. Roberts, Baxter Publishing, Brussels, Belgium, 1989. Schreiber, A.B., Hrinda, M.E., Newman, J., et al., Removal of viral contaminants by monoclonal antibody puriication of plasma proteins, Curr. Stud. Hematol. Blood Transfus. 56, 146–153, 1989. Johnson, A.J., Mathews, R.W., and Fulton, A.J., Approaches to plasma fractionation for improved recovery and the development of potentially useful clinical factors, Clin. Haematol. 13, 3–15, 1984. Pipe, S.W., Recombinant clotting factors, Thromb. Haemost. 99, 840–850, 2008. Grillberger, L., Kreil, T.R., Nasr, S., and Reiter, M., Emerging trends in plasma-free manufacturing of recombinant protein therapeutics expressed in mammalian cells, Biotechnol. J. 4, 186–201, 2009. Mauser-Bunschoten, E.P., Posthouwer, D., Fischer, K., and van den Berg, H.M., Safety and eficacy of a plasma-derived monoclonal factor VIII concentrate during 10 years of follow-up, Haemophilia 13, 697–700, 2007. Batlle, J., Villar, A., Liras, A., et al., Consensus opinion for the selection and use of therapeutic products for the treatment of haemophilia in Spain, Blood Coagul. Fibrinolysis 19, 333–340, 2008. Konigs, C. and von Hentig, N., A review of current literature on second-generation, sucrose-formulated, full-length recombinant factor VIII, Drugs Today 45, 549–561, 2009. Powell, J.S., Liposomal approach towards the development of a longer-acting factor VIII, Haemophilia 13(Suppl. 2), 23–28, 2007.

Factor VIII and von Willebrand Factor

267

198. Andersson, L.-O., Forsman, N., Huang, K., et al., Isolation and characterization of human factor VIII: Molecular forms in commercial factor VIII concentrate, cryoprecipitate, and plasma, Proc. Natl. Acad. Sci. USA 83, 2979–2983, 1986. 199. Lundblad, R.L., Kingdon, H.S., Mann, K.G., and White, G.C., Issues with the assay of factor VIII activity in plasma and factor VIII concentrates, Thromb. Haemost. 84, 942–948, 2000. 200. Lambert, T., Recht, M., Valentino, L.A., et al., Reformulated Beneix: Eficacy and safety in previously treated patients with moderately severe to severe hemophilia B, Haemophilia 13, 233–243, 2007. 201. Björkman, S., A commentary on the differences in pharmacokinetics between recombinant and plasma-derived factor IX and their implications for dosing, Haemophilia 17, 179–184, 2011. 202. McMullen, B.A., Fujikawa, K., Davie, E.W., et al., Locations of disulide bonds and free cysteines in the heavy and light chains of recombinant human factor VIII (antihemophilic factor A), Protein Sci. 4, 740–746, 1995. 203. King, B.R., Goos, P.W., Paterson, M.A., et al., Changes in altitude cause unintended insulin delivery from insulin pumps: Mechanisms and implications, Diabetes Care 34, 1932–1933, 2011. 204. Choudhary, P., Shin, J., Wang, Y., et al., Insulin pump therapy with automated insulin suspension in response to hypoglycermia: Reduction in noctural hypoglycemia in those at greatest risk, Diabetes Care 34, 2023–2025, 2011. 205. Kordonouri, O., Hartmann, R., and Danne, T., Treatment of type diabetes in children and adolescents using modern insulin pumps, Diabetes Res. Clin. Pract. 93 (Suppl. 1), S118–S124, 2011. 206. Frias, J.P., Bode, B.W., Bailey, T.S., et al., A 16-week open-label, multicenter pilot study assessing insulin pump therapy in patients with type 2 diabetes suboptimally controlled with multiple daily injections, J. Diabetes Sci. Technol. 5, 887–893, 2011. 207. Pfützner, A., Musholt, P.B., Malmgren-Hansen, B., et al., Analysis of the environmental impact of insulin infusion sets based on loss of resources with waste, J. Diabetes Sci. Technol. 5, 843–847, 2011. 208. Feichtner, F., Mader, J.K., Schaller, R., et al., A stepwise approach toward closed-loop blood glucose control for intensive care unit patients: Results from a feasibility study in type 1 diabetic subjects using vascular micro dialysis with infrared spectrometry and a model predictive control algorithm, J. Diabet. Sci. Technol. 5, 1901–1905, 2011. 209. Plasse, T., Ohnuma, T., Bruckner, H., et al., Portable inlusion pumps in ambulatory cancer chemotherapy, Cancer 50, 27–31, 1982. 210. Green, E., White, R., Janes, K., et al., Courage, collaboration, complexity and chemotherapy safety: The view from the sharp end, Can. Oncol. Nurs. J. 21, 81–90, 2011. 211. Schmid, E.F., Ashkenazy, R., Merson, J., and Smith, R.A., Will biomedical innovation change the future of healthcare?, Drug Discov. Today 14(21–22), 1037–1044, 2009. 212. Ragni, M.V., Moore, C.G., Bias, V., et al., Challenges of rare disease research: Limited patients and competing priorities, Haemophilia (in press). 213. Moghissi, K., Bond, M.G., Sambrook, R.J., et al., Treatment of endotracheal or endobronchial obstruction by non-small cell lung cancer: Lack of patients in an MRC randomized trial leaves key questions unanswered, Clin. Oncol. 11, 179–183, 1999. 214. Kimmelman, J., The ethics of human gene transfer, Nat. Rev. Genet. 9, 239–244, 2008. 215. Liras, A., Induced human pluripotent stem cells and advanced therapies: Future perspectives for the treatment of haemophilia, Thromb. Res. 128, 8–13, 2011. 216. Xu, D., Alipio, Z., Fink, L.M., et al., Phenotypic correction of murine hemophilia A using an iPS cell-based therapy, Proc. Natl. Acad. Sci. USA 106, 808–813, 2009.

268

Biotechnology of Plasma Proteins

217. Alexeev, V. and Yoon, K., Stable and inheritable changes in genotype and phenotype of albino melanocytes induced by an RNA–DNA oligonucleotide, Nat. Biotechnol. 16, 1343–1346, 1998. 218. Bartlett, R.J., Long-lasting gene repair, Nat. Biotechnol. 16, 1312–1313, 1998. 219. Richardson, P.D., Kran, B.T., and Steer, C.J., Targeted gene correction strategies, Curr. Opin. Mol. Therapeutics 3, 327–337, 2001. 220. Li, H., Haurigot, V., Doyon, Y., et al., In vivo genome editing restores haemostasis in a mouse model of haemophilia, Nature 475, 217–221, 2011. 221. Chao, H., Mansield, S.G., Bartel, R.C., et al., Phenotype correction of hemophilia A mice by spliceosome-mediated RNA trans-splicing, Nat. Med. 9, 1015–1019, 2003. 222. Chao, H. and Walsh, C.E., RNA repair for haemophilia A, Expert Rev. Mol. Med. 8, 1–8, 2006. 223. Waddington, S.N., Buckley, S.M., David, A.L., et al., Fetal gene transfer, Curr. Opin. Mol. Ther. 9, 432–438, 2007. 224. Ponder, K.P., Immunology of neonatal gene transfer, Curr. Gene Ther. 7, 403–410, 2007. 225. Davey, M.G. and Flake, A.W., Genetic therapy for the fetus: A once in a lifetime opportunity, Hum. Gene Ther. 22, 383–385, 2011. 226. Strong, C., Regulatory and ethical issues for Phase I in utero gene transfer studies, Hum. Gene Ther. 22, 1323–1330, 2011. 227. von der Leyen, H.E., Mann, M.J., and Dzau, V.J., Gene inhibition and gene augmentation for the treatment of vascular proliferative disorders, Semin. Interv. Cardiol. 1, 209–214, 1996. 228. Forbes, S.J. and Hodgson, H.J., Review article: Gene therapy in gastroenterology and hepatology, Aliment. Pharmacol. Ther. 11, 823–836, 1997. 229. Mátrai, J., Chuah, M.K.L., and VandenDriessche, T., Preclinical and clinical progress in hemophilia gene therapy, Curr. Opin. Hematol. 17, 387–392, 2010. 230. Petrus, I., Chauh, M., and VandenDriessche, T., Gene therapy for hemophilia: Beneits versus risks, J. Gene Med. 12, 797–809, 2010. 231. Miao, C.H., Immunomodulation for inhibitors in hemophilia A: The importance role of Treg cells, Expert Rev. Hematol. 3, 469–483, 2010. 232. Wang, L., Louboutin, J.P., Bell, P., et al., Muscle-directed gene therapy for hemophilia B with more eficient and less immunogenic AAV vectors, J. Thromb. Haemost. 9, 2009–2019, 2011. 233. Chuah, M.K., VandenDriessche, T., and Morgan, R.A., Development and analysis of retroviral expressing human factor VIII as a potential gene therapy for hemophillia A, Hum. Gene Ther. 6, 1363–1377, 1995. 234. Shi, Q., Wilcox, D.A., Fahs, S.A., et al., Expression of human factor VIII under control of the platelet-speciic αIIb promoter in megakaryocytic cell line as well as storage together with vWF, Mol. Genet. Metab. 79, 25–33, 2003. 235. Yarovoi, H.V., Kufrin, D., Eslin, D.E., et al., Factor VIII ectopically expressed in platelets: Eficacy in hemophilia A treatment, Blood 102, 4006–4013, 2003. 236. Wilcox, D.A., Shi, Q., Nurden, P., et al., Induction of megakaryocytes to synthesis and store a releasable pool of human factor VIII, J. Thromb. Haemost. 1, 2477–2489, 2003. 237. Shi, Q. and Montgomery, R.R., Platelets as delivery systems for disease treatments, Adv. Drug. Deliv. Rev. 62, 1196–1203, 2010. 238. Rosenberg, J.B., Foster, P.A., Kaufman, R.J., et al., Intracellular traficking of factor VIII to von Willebrand storage granules, J. Clin. Invest. 101, 613–624, 1998. 239. Rosenberg, J.B., Greengard, J.S., and Montgomery, R.R., Genetic induction of a releasable pool of factor VIII in human endothelial cells, Arterioscler. Thromb. Vasc. Biol. 20, 2689–2695, 2000. 240. Haberichter, S.L., Shi, Q., and Montgomery, R.R., Regulated release of VWF and FVIII and the biologic implications, Pediatr. Blood Cancer 46, 547–553, 2006.

Factor VIII and von Willebrand Factor

269

241. Grifith, M., Ultrapure plasma factor VIII producted by anti-F VIII c immunoafinity chromatography and solvent/detergent viral inactivation. Characterization of the Method M process and Hemophil M antihemophilic factor(human), Ann. Hematol. 63, 131–137, 1991. 242. Aronson, D.L. and Chang, P., Ultracentrifugal analysis of factor VIII and von Willebrand factor in therapeutic preparations, Vox Sang. 69, 8–13, 1995. 243. Graham, J.B., Biochemical genetic speculations provoked by considering the enigma of von Willibrand’s disease, Thromb. Diath. Haemorrh. 9(Suppl. 11), 119–125, 1962. 244. Bifi, R., Pozzi, S., Agazzi, A., et al., Use of totally implantable central venous access ports for high-dose chemotherapy and peripheral blood stem cell transplantation, Ann. Oncol. 15, 296–300, 2004. 245. Titapiwantanakun, R., Moir, C., Pruthi, R.K., et al., Central venous access devices for paediatric patients with haemophilia: A single-institution experience, Haemphilia 15, 168–174, 2009. 246. Fischer, K., Collins, P., Björkman, S., et al., Trends in bleeding patterns during prophylaxis for severe haemophilia: Observations from a series of prospective clinical trials, Haemophilia 17, 433–438, 2011. 247. Gissel, M., Whelihan, M.R., Ferris, L.A., et al., The inluence of prophylactic factor VIII in severe haemophilia A, Haemophilia 18, 193–199, 2012. 248. Manco-Johnson, M.J. and Blanchette, V.S., North American prophylaxis studies for persons with severe haemophilia: Background, rationale and design, Haemophilia 9(Suppl. 1), 44–48, 2003. 249. Howard, T.E., Yanover, C., Mahlangu, J., et al., Hemophilia management: Time to get personal? Haemophilia 17, 721–728, 2011. 250. Matucci, M., Messori, A., Donati-Cori, G., et al., Kinetic evaluation of four factor VIII concentrates by model-independent methods, Scand. J. Haematol. 34, 22–28, 1985. 251. McLeod, A.G., Walker, I.R., Zheng, S., and Hayward, C.P., Loss of factor VIII activity during storage in PVC containers due to adsorption, Haemophilia 6, 89–92, 2000. 252. Henze, W., Kellermann, E., Larson, P., et al., Stability of full-length recombinant FVIII formulated with sucrose during continuous infusion using a mini-pump infusion device, J. Thromb. Haemost. 3, 1530–1531, 2005. 253. Neidhardt, E., Koval, R., Burke, E., and Warne, N., In vitro evaluation of B-domain deleted recombinant factor VIII (ReFacto™) stability during simulated continuous infusion administration, Haemophilia 11, 319–325, 2005. 254. Fernandez, M., Yu, T., Bjornson, E., et al., Stability of ADVATE™, antihemophilic factor (recombinant) plasma/albumin-free method, during simulated continuous infusion, Blood Coagul. Fibrinolysis 17, 165–171, 2006. 255. Revel-Vilk, S., Blanchette, V.S., Schmugge, M., et al., In vitro and in vivo stability of diluted recombinant factor VIII for continuous infusion use in haemophilia A, Haemophilia 16, 72–79, 2010. 256. Sheth, S., Dimichele, D., Lee, M., et al., Heart transplant in a factor VIII-deicient patient with a high-titre inhibitor: Perioperative management using high-dose continuous infusion factor VIII and recombinant factor VIIa, Haemophilia 7, 227–232, 2001. 257. Windyga, J., Rusen, L., Gruppo, R., et al., BBDrFVIII (Moroctocog alfa[AF-CC]) for surgical haemostasis in patients with haemophilia A: Results of a pivotal study, Haemophilia 16, 731–739, 2010. 258. Stachnik, J.M. and Gabay, M.P., Continuous infusion of coagulation factor products, Ann. Pharmacother. 36, 882–891, 2002. 259. Batorova, A. and Martinowitz, U., Continuous infusion of coagulation factors: Current opinion, Curr. Opin. Hematol. 13, 308–315, 2006.

270

Biotechnology of Plasma Proteins

260. Rochat, C., McFadyen, M.L., Schwyzer, R., et al., Continuous infusion of intermediatepurity factor VIII in haemophilia A patients undergoing elective surgery, Haemophilia 5, 181–186, 1999. 261. St. Charles, M.E., Sadri, H., Minshall, M.E., and Tunnis, S.L., Health economic comparison between continuous subcutaneous insulin infusion and multiple daily injections of insulin for the treatment of adult type 1 diabetes in Canada, Clin. Ther. 31, 657–667, 2009. 262. Cummins, E., Royle, P., Snaith, A., et al., Clinical effectiveness and cost-effectiveness of continuous subcutaneous insulin infusion for diabetes: Systematic review and economic evaluation, Health Technol. Assess. 14, 1–181, 2010. 263. Lynch, P.M., Riedel, A.A., Samant, N., et al., Resource utlization with insulin pump therapy for type 2 diabetes mellitus, Am. J. Manag. Care 16, 892–896, 2010. 264. Fatouros, A., Lidén, Y., and Sjöström, B., Recombinant factor VIII SQ—Stability of VIII: C in homogenates from porcine, monkey and human subcutaneous tissue, J. Pharm. Pharmacol. 52, 797–805, 2000. 265. Miekka, S.I., Jameson, T., Singh, M., et al., Novel delivery systems for coagulation proteins, Haemophilia 4, 436–442, 1998. 266. Matsui, H., Shibata, M., Brown, B., et al., Ex vivo gene therapy for hemophilia A that enhances safe delivery and sustained in vivo factor VIII expression from lentivirally engineered endothelial progenitors, Stem Cells 25, 2660–2669, 2007. 267. Peng, A., Gaitonde, P., Kosloski, M.P., et al., Effect of route of administration of human recombinant factor VIII on its immunogenicity in Hemophilia A mice, J. Pharm. Sci. 98, 4480–4484, 2009. 268. Bowman, K., Sarkar, R., Raut, S., and Leong, K.W., Gene transfer to hemophilia A mice via oral delivery of FVIII-chitosan nanoparticles, J. Control Release 132, 252–259, 2008. 269. Brown, L.R., Commercial challenges of protein drug delivery, Expert Opin. Drug Deliv. 2, 29–42, 2005. 270. Barteau, B., Chèvre, R., Letrou-Bonneval, E., et al., Physicochemical parameters of non-viral vectors that govern transfection eficiency, Curr. Gene Ther. 8, 313–323, 2008. 271. Bünning, H., Perabo, L., Coutelle, O., et al., Recent developments in adeno-associated virus vector technology, J. Gene Med. 10, 717–733, 2008. 272. Chopinet, L., Wasunga, L., and Rols, M.P., First explanations for differences in electrotransfection eficiency in vitro and in vivo using spheroid model, Int. J. Pharm. 423, 7–15, 2012. 273. Fay, P.J., Factor VIII structure and function, Int. J. Hematol. 83, 103–108, 2006. 274. Fay, P.J. and Smudzin, T.M., Characterization of the interaction of between the A2 subunit and A1/A3-C1-C2 dimer in human factor VIIIa, J. Biol. Chem. 267, 13246–13250, 1992. 275. Lollar, P. and Parker, E.T., Structural basis for the decreased procoagulant activity of human factor VIII compared to the porcine homolog, J. Biol. Chem. 266, 12481–12486, 1991. 276. Austen, D.E.G., Thiol groups in the blood clotting action of factor VIII, Brit. J. Haematol. 19, 477–484, 1970. 277. Mello Périssé, A.C., Soria, J., Soria, C., and Master, L., Dissociation beween platelet agglomerating activity and factor VIII procoagulant activity of bovine preparations by chemical treatment II. Effect of periodate oxidation, Pathol. BIol. 21(Suppl.), 63–65, 1973. 278. Kaelin, A.C., Sodium periodate modiication of factor VIII procoagulant activity, Brit. J. Haematol. 31, 349–359, 1975. 279. Manning, F., Fágáin, C.O., O’Kennedy, R., and Woodhams, B., Effects of chemical modiiers on recombinant factor VIII activity, Thromb. Res. 80, 247–254, 1995.

Factor VIII and von Willebrand Factor

271

280. Jameel, F., Tschessolov, S., Bjornson, E., et al., Development of freeze-dried biosynthetic factor VIII, I. A case study in the optimization of formulation, Pharm. Dev. Technol. 14, 687–697, 2009. 281. Lundblad, R.L., unpublished observations, 1976. 282. Chiari, M., Ettori, C., Righetti, P.G., et al., Oxidation of cysteine to cysteic acid in proteins by peroxyacids, as monitored by immobilized pH gradients, Electrophoresis 12, 376–377, 1991. 283. Wang, W. and Kelner, D.N., Correlation of rFVIII inactivation with aggregation in solution, Pharm. Res. 20, 693–700, 2003. 284. Vehar, G.A., Keyt, B., Eaton, D., et al., Structure of human factor VIII, Nature 312, 337–342, 1984. 285. Bihoreau, N., Pin, S., de Kersabiec, A.M., et al., Copper-atom identiication in the active and inactive forms of plasma-derived FVIII and recombinant FVIII-delta II, Eur. J. Biochem. 222, 41–48, 1994. 286. Pan, Y., DeFay, T., Gitschier, J., and Cohen, F.E., Proposed structure of the A domains of factor VIII by homology modelling, Nat. Struct. Biol. 2, 740–744, 1995. 287. Wakabayashi, H., Zhou, Q., Nogami, K., et al., pH-dependent association of factor VIII chains: Enhancement of afinity at physiological pH by Cu2+, Biochim. Biophys. Acta 1764, 1094–1101, 2006. 288. Tagliavacca, L., Moon, N., Durham, W.R., and Kaufmann, R.J., Identiication and functional requirement of Cu(I) and its ligands within coagulation factor VIII, J. Biol. Chem. 272, 27428–27434, 1997. 289. Vassiliev, V.B., Kachurin, A.M., Beltramini, M., et al., Copper depletion/repletion of human ceruloplasmin is followed by the changes in its spectral features and functional properties, J. Inorg. Biochem. 65, 167–174, 1997. 290. Gross, E.L., Draheim, J.E., Curtiss, A.S., et al., Thermal denaturation of plastocyanin: The effect of oxidation state, reductants, and anaerobicity, Arch. Biochem. Biophys. 298, 413–419, 1992. 291. Wakabatashi, H., Koszelak, M.E., Mastri, M., et al., Metal ion-independent association of factor VIII subunits and the roles of calcium and copper ions for cofactor activity and inter-subunit afinity, Biochemistry 40, 10293–10300, 2001. 292. MacPherson, L.S. and Murphy, M.E.P., Type-2 copper-containing enzymes, Cell. Mol. Life Sci. 64, 2887–2899, 2007. 293. Shen, B.W., Spiegel, P.C., Chang, C.-H., et al., The tertiary structure and domain organization of coagulation factor VIII, Blood 111, 1240–1247, 2008. 294. Ngo, J.C., Huang, M., Roth, D.A., et al., Crystal structure of human factor VIII: Implications for the formation of the factor IXa-factor VIIIa complex, Structure 16, 597–606, 2008. 295. Glusker, J.P., Structural aspects of metal liganding to functional groups in proteins, Adv. Protein Chem. 42, 1–76, 1991. 296. Rigo, A., Corazza, A., di Paolo, M.L., et al., Interaction of copper with cysteine: Stability of cuprous complexes and catalytic role of cupric ions in aerobic thiol oxidation, J. Inorg. Biochem. 98, 1495–1501, 2004. 297. Cavallini, D., De Marco, C., Dupre, S., and Rotilio, G., The copper catalyzed oxidation of cysteine to cystine, Arch. Biochem. Biophys. 130, 354–361, 1969. 298. Kachur, A.V., Koch, C.J., and Biaglow, J.E., Mechanism of copper-catalyzed autooxidation of cysteine, Free Radic. Res. 31, 23–34, 1999. 299. Takeyama, M., Nogami, K., Okuda, M., et al., Selective factor VIII and V inactivation by imidioacetate ion exchange resin through metal ion adsorption, Brit. J. Haematol. 142, 962–970, 2008. 300. Venkateswarlu, D., Structural investigation of zymogenic and activated forms of human blood coagulation factor VIII: A computational molecular dynamics study, BMC Struct. Biol. 10, 7, 2010.

272

Biotechnology of Plasma Proteins

301. Andruzzi, L., Nakano, M., Nilges, M.J., and Blackburn, N.J., Spectroscopic studies of metal binding and metal selectivity in Bacillus subtilis BSco, a homologue of the yeast mitochondrial protein Sco1p, J. Am. Chem. Soc. 127, 16548–16558, 2005. 302. Kragh-Hansen, U., Chuang, V.T.G., and Otagiri, M., Practical aspects of the ligandbinding and enzymatic properties of human serum albumin, Biol. Pharm. Bull. 25, 695– 704, 2002. 303. Austen, D.E.G., Thiol groups in the blood clotting action of factor VIII, Brit. J. Haematol. 19. 477–484, 1970. 304. Felix, K. and Weser, U., Release of copper from yeast copper thionein after S-alkylation of copper thiolate clusters, Biochem. J. 252, 577–581, 1988. 305. Manning, F., Fágáin, C., O’Kennedy, R., and Woodhams, B., Effects of chemical modiication on recombinant factor VIII activity, Thromb. Res. 80, 247–254, 1995. 306. Derrick, T.S., Kashi, R.S., Durrani, M., et al., Effect of metal cations on the conformation and inactivation of recombinant factor VIII, J. Pharm. Sci. 93, 2549–2557, 2004. 307. Bayele, H.K., Murdock, P.J., and Pasi, K.J., Residual factor VIII-like cofactor activity of thioredoxin and related-oxidoreductases, Biochim. Biophys. Acta 1800, 308–404, 2010. 308. Mei, B., Pan, C., Jiang, H., et al., Rational design of a fully active, long-acting PEGylated factor VIII for hemophilia A treatment, Blood 116, 270–279, 2010. 309. Seyfried, B.K., Siekmann, J., Belgacem, O., et al., MALDI linear TOF mass spectrometry of PEGylated (glyco)proteins, J. Mass Spectrom. 45, 612–617, 2010. 310. Monetti, C., Rottensteiner, H., Gritsch, H., et al., Structural analysis of the recombinant therapeutic product rFVIII and its PEGylated variants using 2-D DIGE, ELectrophoresis 32, 1292–1301, 2011. 311. Goodson, R.J. and Katre, N.V., Site-directed pegylation of recombinant interleukin-2 at its glycosylation site, Biotechnology 8, 343–346, 1990. 312. Rosendahl, M.S., Doherty, D.H., Smith, D.J., et al., A long-acting, highly potent interferon α-2-conjugate created using site-speciic PEGylation, Bioconjug. Chem. 16, 200–207, 2005. 313. Molineux, G., Pegylation: Engineering improved pharmaceuticals for enhanced therapy, Cancer Treat. Rev. 28(Suppl. A), 13–16, 2002. 314. Zamboni, W.C., Pharmacokinetics of pegilgrastim, Pharmacotherapy 23(8 Pt 2), 9S–14S, 2003. 315. Healy, J.M., Lewis, S.D., Kurz, M., et al., Pharmacokinetics and biodistribution of novel aptamer compositions, Pharm. Res. 21, 2234–2246, 2004. 316. Da Pieve, C., Williams, P., Haddleton, D.M., et al., Modiication of thiol functionalized aptamers by conjugation of synthetic polymers, Bioconjug. Chem. 21, 169–174, 2010. 317. Milla, P., Dosio, F., and Cattel, L., PEGylation of proteins and liposomes: A powerful and lexible strategy to improve the drug delivery, Curr. Drug. Metab. 13, 105–119, 2012. 318. Lenting, P.J., van Mourik, J.A., and Mertens, K., The life cycle of coagulation factor VIII in view of its structure and function, Blood 92, 3983–3996, 1998. 319. Bertina, R.M., Reitsma, P.H., Rosendaal, R.F., and Vandenbroucke, J.P., Resistance to activated protein C and factor V Leiden as risk factors for venous thrombosis, Thromb. Haemost. 74, 449–453, 1995. 320. Gary, T., Hafner, F., Froehlich, H., et al., High factor VIII activity, high plasminogen activator inhibitor 1 antigen levels and low factor XII activity contribute to a thrombophilic tendency in elderly venous thromboembolism patients, Acta Haematol. 124, 214–217, 2010. 321. Mulder, R., van Schouwenburg, I.M., Mahmoodi, B.K., et al., Associations between high factor VIII and free protein S levels with traditional arterial thrombotic risk factors and their risk on arterial thromobosis: Results from a retrospective family cohort study, Thromb. Res. 126, e249–e254, 2010. 322. Ota, S., Yamada, N., Ogihara, Y., et al., High plasma level of factor VIII: An important risk factors for venous thromboembolism, Circ. J. 75, 1472–1475, 2011.

Factor VIII and von Willebrand Factor

273

323. Mahmood, I. and Green, M.D., Pharmacokinetic and pharmacodynamic considerations in the development of therapeutic proteins, Clin. Pharmacokinet. 44, 331–347, 2005. 324. Solá, R.J. and Griebenow, K., Glycosylation of therapeutic proteins. An effective strategy to optimize eficacy, BioDrugs 24, 9–21, 2010. 325. Defrees, S., Wang, Z.-G., Xing, R., et al., GlycoPEGylation of recombinant therapeutic proteins produced in Escherichia coli, Glycobiology 16, 833–843, 2006. 326. Salmaso, S., Semenzato, A., Bersani, S., et al., Site-selective protein glycation and PEGylation, Eur. Polym. J. 44, 1378–1389, 2008. 327. Wang, Y.J., Liu, Y.-D., Chen, J., et al., Eficient preparation and PEGylation of recombinant human non-glycosylated erthryopoietin expressed as inclusion body in E. coli, Int. J. Pharm. 386, 156–164, 2010. 328. Henderson, G.E., Isett, K.D., and Gerngross, T.U., Site-speciic modiication of recombinant proteins: A novel platform for modifying glycoproteins expressed in E. coli, Bioconjug. Chem. 22, 903–912, 2011. 329. Foster, P.A., Fulcher, C.A., Houghten, P.A., and Zimmerman, T.S., Synthetic factor VIII peptides with amino acid sequences contained within the C2 domain of factor VIII inhibit factor VIII binding to phosphatidylserine, Blood 75, 1999–2004, 1990. 330. Barrow, E.S. and Graham, J.B., Polyglutamic acid and polyaspartic acids: Synthetic polypeptides with predominantly factor VII-like coagulant activity, Proc. Soc. Exp. Biol. Med. 152, 160–164, 1976. 331. Barrow, E.M. and Graham, J.B., Kidney antihemophilic factor. Partial puriication and some properties, Biochemistry 7, 3917–3925, 1968. 332. Brakman, P., Sjolin, K.E., and Astrup, T., Is there a hemostatic effect of peanuts in hemophiliod diorders? Thromb. Diath. Haemorrh. 8, 442–454, 1962. 333. Verstraete, M. and Ruys, C.A., Double-blind experiments on the effect of a peanut extract on the bleeding incidence in 92 haemophiliacs, Brit. Med. J. 4(5577), 453–456, 1967. 334. Wolman, I.J., Peanuts and hemophilia, Clin. Pediatr. 7, 311, 1968. 335. Emekli-Alturfan, E., Kasikci, E., and Yarat, A. Peanuts improve blood glutathione, HDL-cholesterol level and change tissue factor activity in rats fed a high-cholesterol diet, Eur. J. Nutr. 46, 476–482, 2007. 336. Powell, J.S., Nugent, D.J., Harrison, J.A., et al., Safety and pharmacokinetics of a recombinant factor VIII with pegylated liposomes in severe hemophila A, J. Thromb. Haemost. 6, 277–283, 2008. 337. Martinowitz, U., Lalezari, S., Luboshitz, J., et al., Infusion rates of recombinant FVIII-FS with PEGylated liposomes in haemophilia A, Haemophilia 14, 1122–1124, 2008. 338. Powell, J., The next generation of anti-haemophilia factor, factor VIII. Long-lasting protection from spontaneous bleeding, are we there yet?, Thromb. Haemost. 100, 365–366, 2008. 339. Pan, J., Liu, T., Kim, J.Y., et al., Enhanced eficacy of recombinant FVIII in noncovalent complex with PEGylated liposome in hemophilia A mice, Blood 114, 2802–2811, 2009. 340. Yatuv, R., Robinson, M., Dayan-Tarshish, I., and Baru, M., The use of PEGylated liposomes in the development of drug delivery applications for the treatment of hemophilia, Int. J. Nanomed. 5, 581–591, 2010. 341. Di Minno, G., Cerbone, A.M., Coppola, A., et al., Longer-acting factor VIII to overcome limitations in haemophilia management: The PEGylated liposomes formulation issue, Haemophilia 16(Suppl. 1), 2–6, 2010. 342. Rapaport, S.I., Hjort, P.E., and Patch, M.J., Further evidence that thrombin activation of factor VIII is an essential step in intrinsic clotting, Scand. J. Clin. Lab. Med. 17(Suppl. 84), 88–100, 1965.

274

Biotechnology of Plasma Proteins

343. Pieters, J., Lindhout, T., and Hemker, H.C., In situ-generated thrombin is the only enzyme that effectively activates factor VIII and factor V in thromboplastin-activated plasma, Blood 74, 1021–1024, 1989. 344. McGee, M.P., Li, L.C., and Hensler, M., Functional assembly of intrinsic coagulation proteases on monocytes and platelets. Comparison between cofactor activities induced by thrombin and factor Xa, J. Exp. Med. 176, 27–35, 1992. 345. Donath, M.-J.S.H., Lenting, P.J., van Mourik, J.A., and Mertens, K., The role of cleavage of the light chain at positions Arg1689 or Arg1721 in subunit interaction and activation of human blood coagulation factor VIII, J. Biol. Chem. 270, 3648–3655, 1995. 346. Fay, P.J., Beattie, T.L., Regan, L.M., et al., Model for the factor VIIIa-dependent decay of the intrinsic tenase. Role of subunit dissociation and factor IXa-catalyzed proteolysis, J. Biol. Chem. 271, 6027–6032, 1996. 347. Bovenschen, N., Mertens, K., Hu, L., et al., LDL receptor cooperates with LDL receptorrelated protein in regulating plasma levels of coagulation factor VIII in vivo, Blood 106, 906–912, 2005. 348. Franchini, M. and Montagnana, M., Low-density lipoprotein receptor-related protein 1: New functions for an old molecule, Clin. Chem. Lab. Med. 49, 967–970, 2011. 349. Blostein, M.D., Rigby, A.C., Furie, B.C., et al., Amphipathic helices support function of blood coagulation factor IXa, Biochemistry 39, 12000–12006, 2000. 350. Ganopolsky, J.G., Charbonneau, S., Peng, H.T., et al., Characterization of an ideal amphiphathic peptide as a procoagulant agent, Biochem. J. 412, 545–551, 2008. 351. Bidwell, E., The puriication of antihaemophilic globulin, Brit. J. Haemtol. 1, 35–45, 1955. 352. Bidwell, E., The puriication of antihaemophilic globulin from animal blood, Brit. J. Haemtol. 1, 386–389, 1955. 353. Rizza, C.D., Management of patients with inherited blood coagulation defects, in Hemostasis and Thrombosis, eds. A.L. Bloom and D.P. Thomas, Chapter 21, pp. 371– 388, Churchill Livingstone, Edinburgh, Scotland, 1981. 354. Forbes, C.D., Barr, R.D., McNicol, G.P., and Douglas, A.S., Aggregation of human platelets by commercial preparations of bovine and porcine antihaemophilic globulin, J. Clin. Pathol. 25, 210–217, 1972. 355. Gervasi, G.B., Bartoli, C., Carpita, G., et al., Decrease of bleeding time by a peptide fraction from bovine factor VIII in laboratory animals, Arzneimittelforschung 38, 1268– 1270, 1988. 356. Baldacci, M., Catalani, R., Fascetti, E., and Gervasi, G.B., Polypeptide fraction from bovine factor VIII does not inluence human platelet aggregation and blood coagulation, Pharm. Res. Commun. 20, 7–12, 1988. 357. Conte, A., Palmari, L., and Ronca, G., Absorption and excretion in the experimental animal of a 14C-ethylmaleimide labelled peptide fraction of bovine factor VIII with antihaemorrhagic activity, Arzneimittalforschung 39, 463–466, 1989. 358. Ammannati, P., Siravo, D., Arrarelli, L., et al., Decrease of diffusion of glycosylated albumin in retinal microcirculation by peptide fraction from bovine factor VIII, Arzneimittalforschung 39, 661–664, 1989. 359. Pazzagli, L., Cecchi, C. Cappugi, G., et al., A peptide fraction from factor VIII reduced PKC activity in cultured endothelial cells, Life Sci. 62, 829–837, 1998. 360. Cardillo Piccolino, F., Ghiglione, D., Ceppa, P., et al., Fundus angiography with luorescein-labelled peptide fraction from bovine factor VIII: Correlation with histologic indings, Eur. J. Ophthalmol. 2, 135–143, 1992. 361. Lollar, P., Parker, C.G., and Tracy, R.P., Molecular characterization of commercial porcine factor VIII concentrates, Blood 71, 137–143, 1988. 362. Lollar, P., Parker, E.T., and Fay, P.J., Coagulant properties of hybrid human/porcine factor VIII molecules, J. Biol. Chem. 267, 23652–23657, 1992.

Factor VIII and von Willebrand Factor

275

363. Ciavarella, N., Antoncecchi, S., and Ranieri, P., Eficacy of porcine factor VIII in the management of haemophiliacs with inhibitors, Brit. J. Haematol. 58, 641–648, 1984. 364. Barrow, R.T. and Lollar, P., Neutralization of antifactor VIII inhibitors by recombinant porcine factor VIII, J. Thromb. Haemost. 4, 2223–2229, 2006. 365. Healey, J.F., Parker, E.T., Barrow, R.T., et al., The comparative immunogenicity of human and porcine factor VIII in haemophilia A mice, Thromb. Haemost. 102, 35–41, 2009. 366. Toschi, V., OBI-1, porcine recombinant Factor VIII for the potential treatment of patients with congenital hemophilia A and alloantibodies against human factor VIII, Curr. Opin. Mol. Ther. 12, 617–625, 2010. 367. Roberts, H.R., Scales, M.B., Madison, J.T., et al., A clinical and experimental study of acquired inhibitors to factor 8, Blood 26, 805–818, 1965. 368. Shapiro, S.S., The immunologic character of acquired inhibitors of antihemophilic globulin (factor 8) and the kinetics of their interaction with factor 8, J. Clin. Invest. 46, 147–156, 1967. 369. Shapiro, S.S., Antibodies to blood coagulation factors, Clinics in Hematology 8, 207– 214, 1979. 370. Brozović, M., Acquired disorders of blood coagulation, in Haemostasis and Thrombosis, eds. A.L. Bloom and D.P. Thomas, Chapter 24, pp. 411–438, Churchill Livingstone, Edinburgh, Scotland, 1981. 371. Mathias, M., Liesner, R., Hann, I., and Khair, K., Immune tolerance in children with factor VIII and IX inhibitors: A single centre experience, Haemophilia 11, 340–345, 2005. 372. Di Paola, J., Aledort, L., Britton, H., et al., Application of current knowledge to the management of bleeding events during immune tolerance induction, Haemophilia 12, 591–597, 2006. 373. Kurth, M.A., Dimichele, D., Sexauer, C., et al., Immune tolerance therapy utilizing factor VIII/von Willebrand factor concentrate in haemophilia A patients with high titre factor VIII inhibitors, Haemophilia 14, 50–55, 2008. 374. Mathias, M., Liesner, R., Hann, I., and Khair, K., Immune tolerance in children with factor VIII and IX inhibitors: A single centre experience, Haemophilia 11, 340–345, 2005. 375. Pollack, A. and Lewis, M.J., Letter: Factor-VIII inhibitor bypassing activity, Lancet 2(7975), 43–44, 1976. 376. English, P.J., Sheppard, E.M., and Wensley, R.T., Factor VIII inhibitor bypassing activity, Lancet 2(7978), 207–208, 1976. 377. Aronson, D.L., Conjecture on factor VIII bypassing activity, Prog. Clin. Biol. Res. 72, 103–121, 1981. 378. Vinazzer, H., Comparison between two concentrates with factor VIII inhibitor bypassing activity, Thromb. Res. 26, 21–29, 1982. 379. Davie, E.W., A brief historical review of the waterfall/cascade of blood coagulation, J. Biol. Chem. 278, 50819–50832, 2003. 380. Walsh, C.E., Gene therapy for the hemophilias, Curr. Opin. Pediatr. 14, 12–16, 2002. 381. von Auer, C., Oldenburg, J., von Depka, M., et al., Inhibitor development in patients with hemophilia A after continuous infusion of FVIII concentrates, Ann. N. Y. Acad. Sci. 1051, 498–505, 2005. 382. Reding, M.T., Immunological aspects of inhibitor development, Haemophilia 12 (Suppl. 6), 30–35, 2006. 383. Lei, T.C. and Scott, D.W., Induction of tolerance to factor VIII inhibitors by gene therapy with immunodominant A2 and C2 domains presented by B cells as Ig fusion proteins, Blood 105, 4865–4870, 2005. 384. Xu, L., Mei, M., Ma, X., and Ponder, K.P., High expression reduces an antibody response after neonatal gene therapy with B domain-deleted human factor VIII in mice, J. Thromb. Haemost. 5, 1805–1812, 2007.

276

Biotechnology of Plasma Proteins

385. Finn, J.D., Ozelo, M.C., Sabatino, D.E., et al., Eradication of neutralizing antibodies to factor VIII in canine hemophilia after liver gene therapy, Blood 116, 5842–5848, 2010. 386. Prowse, C.V., Activated prothrombin complex concentrates: Approaches to their preparation, Thromb. Res. 25, 213–218, 1982. 387. Lusher, J.M., Controlled clinical trials with prothrombin complex concentrates, Prog. Clin. Biol. Res. 150, 277–290, 1984. 388. Blatt, P.M., Lundblad, R.L., Kingdon, H.S., et al., Thrombogenic materials in prothrombin complex concentrates, Ann. Intern. Med. 81, 766–770, 1974. 389. White, G.C. 2nd., Roberts, H.R., Kingdon, H.S., and Lundblad, R.L., Prothrombin complex concentrates: Potentially thrombogeneic materials and clues to the mechanism of thrombosis in vivo, Blood 49, 159–170, 1977. 390. Martin-Villar, J., Magallón, M., Ortega, F., and Gago, J., Eficacy of prothrombin complex concentrates in hemophiliacs with antibodies to factor VIII. A retrospective study of 114 hemorrhagic episodes, in Activated Prothrombin Complex Concentrates. Managing Hemophilia with Factor VIII Inhibitors, eds. G. Mariani, M.A. Russo, and F. Mandelli, Praeger, New York, 1982. 391. Kelly, P. and Penner, J.A., Antihemophilic factor inhibitors. Management with prothrombin complex concentrates, J. Am. Med. Assoc. 236, 2061–2064, 1976. 392. Prowse, C.V., Activated prothrombin complex concentrates: Approaches to their preparation, Thromb. Res. 25, 213–218, 1982. 393. Vinazzer, H., Comparison between the two concentrates with factor VIII inhibitor bypassing activity, Thromb. Res. 26, 21–29, 1982. 394. Segligsohn, U., Kasper, C.K., Osterud, B., and Rapaport, S.I., Activated factor VII: Presence in factor IX concentrates and persistance in the circulation after infusion, Blood 53, 828–837, 1979. 395. Rao, L.V. and Rapaport, S.I., Factor VIIa-catalyzed activation of factor X independent of tissue factor: Its possible signiicance for control of hemophilc bleeding by infused factor VIIa, Blood 75, 1069–1073, 1990. 396. Lundblad, R.L., Bergstrom, J., De Vreker, R., et al., Measurement of active coagulation factors in Autoplex-T with colorimetric active site-speciic assay technology, Thromb. Haemost. 80, 811–815, 1998. 397. Kisiel, W., Recollections on the discovery of factor VIIa as a novel therapeutic agent for hemophiliacs with inhibitors, J. Thromb. Haemost. 7, 1053–1056, 2009. 398. Ruggeri, Z.M. (ed.), Von Willebrand Factor and the Mechanisms of Platelet Function, Springer-Verlag, Berlin, Germany, 1999. 399. Groot, E., de Groot, P.G., Fijnheer, R., and Lenting, P.J., The presence of active von Willebrand factor under various pathological conditions, Curr. Opin. Hematol. 14, 284–289, 2007. 400. Reininger, A.J., Function of von Willebrand factor in haemostasis and thrombosis, Haemophilia 15 (Suppl. 5), 11–26, 2008. 401. Lenting, P.J., Pegon, J.N., Groot, E., and De Groot, P.G., Regulation of von Willebrand factor-platelet interactions, Thromb. Haemost. 104, 449–455, 2010. 402. Yakushkin, V.V., Zyuryaey, I.T., Khaspekova, S.G., et al., Glycoprotein IIb–IIIa content and platelet aggregation in healthy volunteers and patients with acute coronary syndrome, Platelets 22, 243–251, 2011. 403. Benett, J.S., Berger, B.W., and Billings, P.C., The structure and function of platelet integrins, J. Thromb. Haemost. 7(Suppl. 1), 200–205, 2009. 404. Clemetson, K.J. and Clemetson, J.M., Platelet Gp1b complex as a target for antithrombotic drug development, Thromb. Haemost. 99, 473–479, 2008. 405. Firbas, C., Siller-Matula, J.M., and Jilma, B., Targeting von Willebrand factor and platelet glycoprotein Ib receptor, Expert Rev. Cardiovasc. Ther. 8, 1689–1701, 2010. 406. Legaz, M.E., Schmer, G., Counts, R.B., et al., Isolation and characterization of human factor VIII (antihemophilic factor), J. Biol. Chem. 248, 3946–3955, 1973.

Factor VIII and von Willebrand Factor

277

407. Shapiro, G.A., Andersen, J.C., Pizzo, S.V., et al., The subunit structure of normal and hemophilic factor VIII, J. Clin. Invest. 52, 2198–2210, 1973. 408. Owen, W.G. and Wagner, R.H., Antihemophilic factor: Separation of an active fragment following dissociation by salts or detergents, Thromb. Diath. Haemorrh. 27, 502–515, 1972. 409. Owen, W.G., Big-piece, little piece or: Yes, factor VIII is a protein, J. Thromb. Haemost. 3, 1905–1909, 2005. 410. Zimmerman, T.S. and Meyer, D., Structure and function of factor VIII/von Willebrand factor, in Thrombosis and Haemostasis, eds. A.L. Bloom and D.P. Thomas, Chapter 8, pp. 111–123, Churchill Livingstone, Edinburgh, Scotland, 1981. 411. Gadisseur, A., Hermans, C., Berneman, Z., et al., Laboratory diagnosis and molecular classiication of von Willebrand disease, Acta Haematol. 121, 71–84, 2009. 412. James, A.H., Manco-Johnson, M.J., Yawn, B.P., et al., Von Willebrand disease: Key points from the 2008 National Heart, Lung, and Blood Institute guidelines, Obstet. Gynecol. 114, 674–678, 2009. 413. Favaloro, E.J., Diagnosis and classiication of von Willebrand disease: A review of the differential utility of various functional von Willebrand factor assay, Blood Coagul. Fibrinolysis 22, 553–564, 2011. 414. Bloom, A.L., Peake, I.R., and Giddings, J.C., Letter: Factor VIII-related antigen or von Willebrand factor?, Lancet 1(7857), 576, 1974. 415. Thomson, C., Forbes, C.D., and Prentice, C.R., Evidence for a qualitative defect in factor-VIII-related antigen in von Willebrand’s disease, Lancet 1(7858), 594–596, 1974. 416. Zimmerman, T.S., Roberts, J., and Edgington, T.S., Factor-VIII-related antigen: Multiple molecular forms in human plasma, Proc. Natl. Acad. Sci. USA 72, 5121–5125, 1975. 417. Weiss, H.J., Abnormalities of factor VIII and platelet aggregation—Use of ristocetin in diagnosing the von Willebrand syndrome, Blood 45, 403–412, 1975. 418. Ness, P.M., Hymas, P.G., and Perkins, H.A., Re-evaluation of plasma from patients previously diagnosed as having von Willebrand’s disease with the factor VIII-related antigen and ristocetin cofactor assay, Am. J. Clin. Pathol. 71, 26–30, 1979. 419. Castaman, G. and Rodeghiero, F., Advances in the diagnosis and management of type 1 von Willebrand disease, Expert Rev. Hematol. 4, 95–106, 2011. 420. Hughes, S.F., Hendricks, B.D., Edwards, D.R., and Middleton, J.F., Tourniquet-applied upper limb orthopaedic surgery results in increased inlammation and changes to leukocyte, coagulation and endothelial markers, PLoS One 5, e11846, 2010. 421. Gluhovschi, C., Gluhovschi, G., Potencz, E., et al., The endothelial cell markers von Willebrand Factor (vWF), CD31 and CD34 are lost in glomerulonephritis and no longer correlate with the morphological indices of glomerular sclerosis, interstitial ibrosis, activity and chronicity, Folia Histochem. Cytobiol. 48, 230–236, 2010. 422. Kiskin, N.I., Hellen, N., Babich, V., et al., Protein mobilities and P-selectin storage in Weibel-Palade bodies, J. Cell. Sci. 123, 2964–2975, 2010. 423. Lampka, M., Grabczewska, Z., Jendryczka-Maćkiewicz, E., et al., Circulating endothelial cells in coronary artery disease, Kardiol. Pol. 68, 1100–1105, 2010. 424. Panagiotopouls, I., Palatinos, G., Michalopoulos, A., et al., Alterations in biomarkers of endothelial function following on-pump coronary artery revascularization, J. Clin. Lab. Anal. 24, 389–398, 2010. 425. Lumachi, F., Zanella, S., Cella, G., et al., Endothelial activation markers soluble E-selectin and von Willebrand factor in primary hyperparathyroidism, In Vivo 25, 279– 282, 2011. 426. Ochoa, C.D., Wu, S., and Stevens, T., New developments in lung endothelial heterogeneity: Von Willebrand factor, P-selectin, and the Weibel-Palade body, Semin. Thromb. Hemost. 36, 301–308, 2010. 427. Rand, J.R., Chu, S.V., and Potter, B.J., In vitro multimerization of human von Willebrand factor from its subunits, Brit. J. Haematol. 67, 433–436, 1987.

278

Biotechnology of Plasma Proteins

428. Purvis, A.R., Gross, J., Dang, L.T., et al., Two cys residues essential for von Willebrand factor multimer assembly in the Golgi, Proc. Natl. Acad. Sci. USA 104, 15647–15652, 2007. 429. Journet, A.M., Saffaripour, S., and Wagner, D.D., Requirement for both D domains of the propolypeptide in von Willebrand factor multimerization and storage, Thromb. Haemost. 70, 1053–1057, 1993. 430. Sadler, J.E., von Willebrand factor assembly and secretion, J. Thromb. Haemost. 7 (Suppl. 1), 24–27, 2009. 431. Sadler, J.E., Biochemistry and genetics of von Willebrand factor, Annu. Rev. Biochem. 67, 395–424, 1998. 432. Slaughter, T.F., Parker, J.K., and Greenberg, C.S., A rapid method for the diagnosis of von Willebrand’s disease subtypes by the clinical laboratory, Arch. Pathol. Lab. Med. 119, 148–152, 1995. 433. Ott. H.W., Griesmacher, A., Schnapka-Koepf, M., et al., Analysis of von Willebrand factor multimers by simultaneous high- and low-resolution vertical SDS-agarose gel electrophoresis and Cy5-labeled antibody high-sensitivity luorescence detection, Am. J. Clin. Pathol. 133, 322–330, 2010. 434. Pruthi, R.K. Daniels, T.M., Heit, J.A., et al., Plasma von Willebrand factor multimer quantitative analysis by in-gel immunostaining and infrared luorescent imaging, Thromb. Res. 126, 543–549, 2010. 435. Ledford-Kraemer, M.R., Analysis of von Willebrand factor structure by mulimer analysis, Am. J. Hematol. 85, 510–514, 2010. 436. Wagner, D.D., Cell biology of von Willebrand factor, Ann. Rev. Cell Biol. 6, 217–246, 1990. 437. Hohenstein, K., Griesmacher, A., Weigel, G., et al., Native multimer analysis of plasma and platelet von Willebrand factor compared to denaturing separation: Implication for the interpretation of satellite bands, Electrophoresis 32, 1684–1691, 2011. 438. McGrath, R.T., McRae, E., Smith, O.P., and O’Donnell, J.S., Platelet von Willebrand factor—Structure, function and biological importance, Brit. J. Haematol. 148, 834–843, 2010. 439. Chung, D.W. and Fujikawa, K., Processing of von Willebrand factor by ADAMTS-13, Biochemistry 41, 11065–11070, 2002. 440. Cal, S., Obaya, A.J., Llamazares, M., et al., Cloning, expression analysis, and structural characterization of seven novel human ADAMTSs, a family of metalloproteinases with disintegrin and thrombospondin-1 domains, Gene 283, 49–62, 2002. 441. Don, J.F., Moake, J.L., Bernardo, A., et al., ADAMTS-13 metalloproteinase interacts with the endothelial cell-derived ultra-large von Willebrand factor, J. Biol. Chem. 278, 29633–29639, 2003. 442. Dong, J.F., Moake, J.L., Nolasco, L., et al., ADAMTS-13 rapidly cleaves newly secreted ultralarge von Willebrand factor multimers on the endothelial surface under lowing conditions, Blood 100, 4033–4039, 2002. 443. Furlan, M. and Lämmle, B., Assays of von Willebrand factor-cleaving protease: A test for diagnosis of familial and acquired thrombotic thrombocytopenic purpura, Semin. Thromb. Hemost. 28, 167–172, 2002. 444. Tsai, H.M., Thrombotic thrombocytopenic purpura: A thrombotic disorder caused by ADAMTS-13 deiciency, Hematol. Oncol. Clin. North Am. 21, 609–632, 2007. 445. Manea, M. and Karpman, D., Molecular basis of ADAMTS-13 dysfunction in thrombotic thrombocytopenic purpura, Pediatr. Nephrol. 24, 447–458, 2009. 446. Peyvandi, F., Palla, R., Lotta, L.A., et al., ADAMTS-13 assays in thrombotic thrombocytopenic purpura, J. Thromb. Haemost. 8, 631–640, 2010. 447. Zhou, Z., Nguyen, T.C., Guchhait, P., and Dong, J.F., Von Willebrand factor, ADAMTS-13 and thrombotic thrombocytopenic purpura, Semin. Thromb. Hemost. 36, 71–81, 2010.

Factor VIII and von Willebrand Factor

279

448. Lancellotti, S. and De Cristofaro, R., Structure and proteolytic properties of ADAMTS-13: A metalloprotease involved in the pathogenesis of thrombotic microaniopathies, Prog. Mol. Biol. Transl. Sci. 99, 105–144, 2011. 449. Plaimauer, B. and Scheilinger, F., Expression and characterization of recombinant human ADAMTS-13, Semin. Hematol. 41, 24–33, 2004. 450. Norris, P. and Balduini, C.L., Investigational drugs in thrombotic thrombocytopenic purpura, Expert Opin. Investig. Drugs 20, 1087–1098, 2011. 451. Plaimauer, B., Kremer Hovinga, J.A, Juno, C., et al., Recombinant ADAMTS13 normalized von Willebrand factor cleaving activity in plasma of acquired TTP patients by overriding inhibitory antibodies, J. Thromb. Haemost. 9, 936–944, 2011. 452. Terrell, D.R., Williams, L.A., Vesely, S.K., et al., The incidence of thrombotic thrombocytopenic purpura-hemolytic uremic syndrome: All patients, idiopathic patients and patient with severe ADAMTS-13 deiciency, J. Thromb. Haemost. 3, 1432–1436, 2005. 453. Jorpes, J.E., Erik von Willebrand and von Willebrand’s disease, Thromb. Diath. Haemorrh. 9 (Suppl. 11), 95–101, 1962. 454. Marques, M.B. and Fritsma, G.A., von Willebrand disease laboratory diagnosis, Am. J. Clin. Pathol. 135, 818–820, 2011. 455. Chandler, A.B., Stormorken, H., Solum, N.O., and Gaarder, A., An attempt to assay the the ‘anti-Willebrand factor,’ Scand. J. Clin. Lab. Invest. 17(Suppl. 84), 129–132, 1965. 456. Budde, U., Drewke, E., Mainusch, K., and Schneppenheim, R., Laboratory diagnosis of congenital von Willebrand disease, Semin. Thromb. Hemost. 28, 173–190, 2002. 457. Federici, A.B., Diagnosis of inherited von Willebrand disease: A clinical perspective, Semin. Thromb. Hemost. 32, 555–565, 2006. 458. Chandler, W.L., Peerschke, E.I., Castellone, D.D., and Meijer, P., Von Willebrand factor assay proiciency testing. The North American Specialized Coagulatlion Laboratory Association experience, Am. J. Clin. Pathol. 135, 862–869, 2011. 459. Sadler, J.E., Low von Willebrand factor: Sometimes a risk factor and sometimes a disease, Hematology Am. Soc. Hematol. Educ. Program 2009, 106–112, 2009. 460. Böhm, M., Täschner, S., Kretzschmar, E., et al., Cold storage of citrated whole blood induces drastic time-dependent losses in factor VIII and von Willebrand factor: Potential for misdiagnosis of haemophilia and von Willebrand disease, Blood Coagul. Fibrinolysis 17, 39–45, 2006. 461. Totonchi, A., Eshraghi, Y., Beck, D., et al., Von Willebrand Disease: Screening, Diagnosis, and Management, Aesthetic Surg. J. 28, 189–194, 2008. 462. Budde, U., Metzner, H.J., and Müller, H.G., Comparative analysis and classiication of von Willebrand factor/factor VIII concentrates: Impact on treatment of patients with von Willebrand disease, Semin. Thromb. Hemost. 32, 626–635, 2006. 463. Favaloro, E.J., Kershaw, G., McLachlan, A.J., and Lloyd, J., Time to think outside the box? Proposals for a new approach to future pharmacokinetic studies of von Willebrand factor concentrates in people with von Willebrand disease, Semin. Thromb. Hemost. 33, 745–758, 2007. 464. Auerswald, G. and Kreuz, W., Haemate P/Humate-P for the treatment of von Willebrand disease: Considerations for use and clinical experience, Haemophilia 14 (Suppl. 5), 39–46, 2008. 465. Batlle, J., López-Fernández, M.F., Fraga, E.L., et al., Von Willebrand factor/factor VIII concentrates in the treatment of von Willebrand disease, Blood Coagul. Fibrinolysis 20, 89–100, 2009. 466. Windyga, J. and von Depka-Prondzinski, M., Eficacy and safety of a new generation von Willebrand factor/factor VIII concentrate (Wilate®) in the management of perioperative haemostasis in von Willebrand disease patients undergoing surgery, Thromb. Haemost. 105, 1072–1079, 2011.

280

Biotechnology of Plasma Proteins

467. Fisher, B.E., Recombinant von Willebrand factor: Potential therapeutic use, J. Thromb. Thrombolysis 8, 197–205, 1999. 468. Plaimauer, B., Schlokat, U., Turecek, P.L., et al., Recombinant von Willebrand factor: Preclinical development, Semin. Thromb. Hemost. 27, 395–403, 2001. 469. De Meyer, S.F., Deckmyn, H., and Vanhoorelbeke, K., von Willebrand factor to the rescue, Blood 113, 5049–5057, 2009. 470. Michiels, J.J., van Vliet, H.H., Berneman, Z., et al., Intravenous DDAVP and factor VIII-von Willebrand factor concentrate for the treatment and prophylaxis of bleeding patients with von Willebrand disease type 1, 2, and 3, Clin. Appl. Thromb. Hemost. 13, 14–34, 2007. 471. Federici, A.B., The use of desmopressin in von Willebrand disease: The experience of the irst 30 years (1977–2007), Haemophilia 14(Suppl. 1), 5–14, 2008. 472. Rodeghiero, F., Castaman, G., and Tosetto, A., How I treat von Willebrand disease, Blood 114, 1158–1165, 2009. 473. Castaman, G., Montgomery, R.R., Meschengieser, S.S., et al., von Willebrand’s disease diagnosis and laboratory issues, Haemophilia 16(Suppl. 5), 67–73, 2010. 474. Nauenberg, E. and Sullivan, S.D., Firm behavior in the U.S. market for factor VIII: A need for policy?, Soc. Sci. Med. 39, 1591–1603, 1994. 475. Iserson, K.V., The limits of health care resources, Am. J. Emerg. Med. 10, 588–592, 1992. 476. De Vresse, L., Evidence-based medicine and progress in the medical sciences, J. Eval. Clin. Pract. 17, 852–856, 2011. 477. Gilbert, A. and Tonkovic, B., Case report of specialty pharmacy management of hemophilia, J. Mang. Care Pharm. 17, 175–176, 2011. 478. Siddiqi, A.E., Ebrahim, S.H., Soucie, J.M., et al., Burden of disease resulting from hemophilia in the U.S., Am. J. Prev. Med. 38, S482–S488, 2010. 479. Levine, P.H., Deiciency in current hemophilia therapy: Need for factor XIV, J. Am. Med. Assoc. 219, 213–215, 1972. 480. Green, D., Therapeutic materials, in Hemophilia, ed. D. Green, Chapter 2, pp. 18–30, Charles C. Thomas, Springield, IL, 1972. 481. Lazerson, J., Hemophilia home transfusion program: Analysis of cost data, J. Pediatr. 83, 623–625, 1973. 482. Oldenburg, J., Dolan, G., and Lemm, G., Haemophilia care then, now and in the future, Haemophilia 15(Suppl. 1), 2–7, 2009. 483. Tuddenham, E.G.D. and Laffan, M., Puriied factor VIII, Brit. Med. J. 311(7003), 465, 1995. 484. Anon., Factor VIII Baxter: rAHF-PFM, recombinant factor VIII-protein free, Drugs 4, 366–368, 2003. 485. Shord, S.S. and Lindlay, C.M., Coagulation products and their uses, Am. J. Health Syst. Pharm. 57, 1403–1417, 2000. 486. Dwyre, D.M., Fernando, L.P., and Holland, P.V., Hepatitis B, hepatitis C and HIV transfusion-transmitted infections in the 21st century, Vox Sang. 100, 92–98, 2011. 487. Hay, J.W., Ernst, R.L., and Kessler, C.M., Cost effectiveness analysis alternative factor VIII products in treatment of haemophilia A, Haemophilia 5, 191–202, 1999. 488. Brown, S.A., and Aledort, L.M., Economic challenges in haemophilia, Haemophilia 11, 64–72, 2005. 489. Ragni, M.V., Rationale for a randomized controlled trial comparing two prophylaxis regimens in adults with severe hemophilia A: The Hemophilia Adult Prophylaxis Trial, Expert Rev. Hematol. 4, 495–507, 2011. 490. Columbo, G.L., Di Matteo, S., Mascusio, M.E., and Santagastina, E., Cost-utility of prophylaxis versus treatment on demand in severe hemophilia, Clinicoecon. Outcomes Res. 3, 55–61, 2011.

Factor VIII and von Willebrand Factor

281

491. Gringeri, A., Factor VIII safety: Plasma-derived versus recombinant products, Blood Transfusion 9, 366–370, 2011. 492. Koren, C., Zuckerman, L.A., and Mire-Sluis, A.R., Immune responses to therapeutic proteins in human—Clinical signiicance, assessment, and prediction, Curr. Pharm. Biotechnol. 3, 349–360, 2002. 493. McKeage, K. and Wagstaff, A.J., Subcutaneous interferon-β-1a: New formulation, CNS Drugs 21, 871–876, 2007. 494. Jaber, A., Driebergen, R., Giovannoni, G., et al., The Rebif new formulation story: It’s not trials and error, Drugs R&D 8, 335–348, 2007. 495. Sauerborn, M. and Schellekens, H., B-1 cells and naturally occuring antibodies: Inluencing the immunogenicity of recombinant human therapeutic proteins?, Curr. Opin. Biotechnol. 20, 715–721, 2009. 496. Singh, S.K., Impact of product-related factors on immunogenicity of biotherapeutics, J. Pharm. Sci. 100, 354–387, 2011. 497. Mohammed, A.E., Epstein, J.D., and Li-McLeod, J.M., Patient and parent preferences for haemophilia A treatment, Haemophilia 17, 209–214, 2011. 498. Mantovani, L.G., Monzini, M.S., Manucci, P.M., et al., Differences between patients’, physcicians’ and pharmacists’ preferences for treatment products in haemophilia: A discrete choice experiment, Haemophilia 11, 589–597, 2005. 499. Flesland, O., Seghatchian, J., and Solheim, B.G., The Norwegian plasma fractionation proejct—A 12 year clinical and economic success story, Transfus. Apher. Med. 29, 93–100, 2003. 500. Bocciardo, L., Martinengo, M., Ardenghi, D., et al., Plasma derivatives and strategies for reaching self-suficiency in Liguria: The role of the Transfusion Medicine Service of the Gaslini Institute, Blood Transfus. 5, 85–92, 2007. 501. Cheraghali, A.M. and Aboofazeli, R., Economical impact of plasma fractionation project in Iran on affordability of plasma-derived medicines, Transfus. Med. 19, 363–368, 2009. 502. Park, Q., Kim, M.J., Lee, J., et al., Plasma fractionation in Korea: Working towards selfsuficiency, Korean J. Hematol. 45, 3–5, 2010. 503. Ghosh, K., Ghosh, K., and Shetty, S., Hemostasis research in India: Past, present, and future, Clin. Appl. Thromb. Hemost. 18, 128–133, 2012. 504. Rautonen, J., Self-suficiency, free trade and safety, Biologicals 38, 97–99, 2010. 505. High, K.A. and Skinner, M.W., Cell phones and landlines: The impact of gene therapy on the cost and availability of treatment for hemophilia, Mol. Ther. 19, 1749–1750, 2011. 506. Wang, X.F., Zhao, Y.Q., Yang, R.C., et al., The prevalence of factor VIII inhibitors and genetic aspects of inhibitor development in Chinese patients with haemophlia A, Haemophilia 16, 632–639, 2010. 507. Bafunno, V., Santacroce, R., Chetta, M., et al., Polymorphisms in genes involved in autoimmune disease and the risk of FVIII inhibitor development in Italian patients with haemophilia A, Haemophilia 16, 469–473, 2010. 508. De Barros, M.F., Herrero, J.C., Sell, A.M., et al., Inluence of class I and II HLA alleles on inhibitor development in severe haemophilia A patients from the South of Brazil, Haemophilia (in press). 509. Di Paolo, J., Aledort, L., Britton, H., et al., Application of current knowledge to the management of bleeding events during immune tolerance induction, Haemophilia 12, 591–597, 2006. 510. Zakarija, A., Harris, S., Rademaker, A.W., et al., Alloantibodies to factor VIII in haemophilia, Haemophilia 17, 636–640, 2011. 511. Pool, J.G. and Shannon, A.E., Production of high-potency concentrates of antihemophilic globulin in a closed-bag system, N. Engl. J. Med. 273, 1443–1447, 1965.

282

Biotechnology of Plasma Proteins

512. Gjerset, G.F., Pike, M.C., Mosley, J.W., et al., Effect of low- and intermediate-purity clotting factor therapy on progress of human immunodeiciency virus infection in congenital clotting disorders, Blood 84, 1666–1671, 1994. 513. Dobrkovska, A., Krzensk, U., and Chediak, J.R., Pharmacokinetics, eficacy and safety of Humate-P® in von Willebrand disease, Haemophilia 4(Suppl. 3), 33–39, 1998. 514. Kessler, C.M., Friedman, K., Schwartz, B.A., et al., The pharmacokinetic diversity of two von Willebrand factor(VWF)/factor VIII (FVIII) concentrates in subjects with congenital von Willebrand disease. Results from a prospective, randomized crossover study, Thromb. Haemost. 106, 279–288, 2011. 515. Hay, C.R., Ludlam, C.A, Lowe, G.D., et al., The effect of monoclonal or ion-exchange puriied factor VIII concentrate on HIV disease progression: A prospective cohort comparison, Brit. J. Haematol. 101, 632–637, 1998. 516. Rivera, J., Escolar, G., Casmiquela, R., et al., von Willebrand factor contained in a high purity FVIII concentrate (Fanhdi) binds to platelet glycoproteins and supports platelet adhesion to subendothelium under low conditions, Haematologica 84, 5–11, 199. 517. Auerswald, G., Eberspächer, B., Engl, W., et al., Successful treatment of patients with von Wilebrand disease using a high-purity double-virus inactivated factor VIII/von Willebrand factor concentrate (Immunate®), Semin. Thromb. Hemost. 28, 203–214, 2002. 518. Weinstein, R.E., Immunoafinity puriication of factor VIII, Ann. Clin. Lab. Sci. 19, 84–91, 1989. 519. Kasper, C.K., Kim, H.C., Gomperts, E.D., et al., In vivo recovery and survival of monoclonal-antibody-puriied factor VIII concentrates, Thromb. Haemost. 66, 730–733, 1991. 520. Grifith, M., Kingdon, H., Liu, S.L., and Burkart, W., In-process controls and characterization of recombinate antihemophilic factor (recombinant), Ann. Hematol. 63, 166– 171, 1991. 521. Boedeker, B.G., The manufacturing of the recombinant factor VIII, Kogenate, Transfus. Med. Rev. 6, 256–260, 1992. 522. Kelley, B., Jankowski, M., and Booth, J., An improved manufacturing process of Xyntha/ Refacto AF, Haemophilia 16, 717–725, 2010. 523. Allen, S.H., Kellermeyer, R.W., Stjernholm, R.L., and Wood, H.G., Puriication and properties of enzymes involved in the propionic acid fermentation, J. Bacteriol. 87, 171–187, 1964. 524. Löf, A.-L., Gustafson, G., Novak, V., et al., Determination of total protein in highly puriied factor IX concentrates, Vox Sang. 63, 172–177, 1992. 525. Scharrer, I. and Becker, T., Products used to hemophilia: Evolution of treatment for hemophilia A and B, in Textbook of Hemophilia, eds. C.A. Lee, E.E. Berntorp, and W.K. Hoots, Chapter 23, pp. 131–135, Blackwell, Malden, MA, 2005. 526. Barrowcliffe, T.W., Curtis, A.D., and Thomas, D.P., Standardization of factor VIII–IV. Establishment of the 3rd International Standard for Factor VIII: C concentrate, Thromb. Haemost. 50, 697–702, 1983. 527. Newman, J., Johnson, A.J., Karpatkin, M.H., and Puszkin, S., Methods for the production of clinically effective intermediate- and high-purity factor-VIII concentrates, Brit. J. Haematol. 21, 1–20, 1972. 528. Margolis, J. and Rhoades, P., Preparation of stable intermediate-purity factor VIII concentrate with a note on high-purity factor VIII, Vox Sang. 36, 369–374, 1979. 529. Roberts, P.L., Virus inactivation by solvent/detergent treatment using Triton X-100 in a high purity factor VIII, Biologicals 36, 330–335, 2008. 530. Dmoszynska, A., Hellman, A., Baglin, T., et al., Pharmacokinetics of Optivate®, a highpurity concentrate of factor VIII with von Willebrand factor, in patients with severe haemophilia A, Haemophilia 17, 185–190, 2011.

Factor VIII and von Willebrand Factor

283

531. Nauenberg, E. and Sullivan, S.D., Firm behavior in the U.S. market for factor VIII: A need for policy?, Soc. Sci. Med. 39, 1591–1603, 1994. 532. Suiter, T.M., First and next generation native rFVIII in the treatment of hemophilia A. What has been achieved? Can patients be switched safely?, Semin. Thromb. Hemost. 28, 277–284, 2002. 533. Voorberg, J. and van den Brink, E.N., Phage display technology: A tool to explore the diversity of inhibitors to blood coagulation factor VIII, Semin. Thromb. Hemost. 26, 143–150, 2000. 534. Mersich, C., Billes, W., Paginger, I., and Jungbauer, A., Peptides derived from a secretory yeast library restore factor VIII activity in the presence of an inhibitory antibody, Biotechnol. Bioeng. 98, 12–21, 2007. 535. Hermans, C., Brackmann, H.H., Schinco, P., and Auerswald, G., The case for wider use of recombinant factor VIII concentrates, Crit. Rev. Oncol. Hematol. (in press). 536. Farrugia, A., Product delivery in the developing world: Options, opportunities and threats, Haemophilia 10(Suppl. 4), 77–82, 2004. 537. Nemerson, Y., Tissue factor and hemostasis, Blood 71, 1–8, 1988. 538. Pedersen, A.H., Lund-Hansen, T., Bisgaard-Franzen, H., et al., Autoactivation of human recombinant coagulation factor VII, Biochemistry 28, 9331–9336, 1989. 539. Soejima, K., Mizuguchi, J., Yuguchi, M., et al., Factor VIIa modiied in the 170 loop shows enhanced catalytic activity but does not change the zymogen-like property, J. Biol. Chem. 276, 17229–17235, 2001. 540. Persson, E. and Olsen, O.H., Current status on tissue factor activation of factor VIIa, Thromb. Res. 125(Suppl. 1), S11–S12, 2010. 541. Gertler, A., Walsh, K.A., and Neurath, H., Catalysis by chymotrypsinogen. Demonstration of an acyl-zymogen intermediate, Biochemistry 13, 1302–1310, 1974. 542. Aronson, D.L. and Mustafa, A.J., The activation of human factor X in sodium citrate: The role of factor VII, Thromb. Haemost. 36, 104–114, 1976. 543. White II, G.C., Roberts, H.R., Kingdon, H.S., and Lundblad, R.L., Prothombin complex concentrates: Potentially thromogenic materials and clues to the mechanism of thrombosis in vivo, Blood 49, 159–170, 1977. 544. Jeong, E.K., Sung, H.M., and Kim, I.S., Inactivation and removal of inluenza A virus H1N1 during the manufacture of plasma derivatives, Biologicals 38, 652–657, 2010. 545. Groenevald, A.B., Navickis, R.J., and Wilkes, M.M., Update on the comparative safety of colloids: A systematic review of clinical studies, Ann. Surg. 253, 470–483, 2011. 546. Alter, H.J., Stramer, S.L., and Dodd, R.Y., Emerging infectious diseases that threaten the blood supply, Semin. Hematol. 44, 32–42, 2007. 547. Allain, J.P., Stramer, S.L., Carneiro-Proietti, A.B., et al., Transfusion-transmitted infectious disease, Biologicals 37, 71–77, 2009. 548. Vamvakas, E.C., Relative risk of reducing the lifetime blood donation deferral for men who have had sex with men versus currently tolerated transfusion risks, Transfus. Med. Rev. 25, 47–60, 2011. 549. Lindholm, P.F., Annen, K., and Ramsey, G., Approaches to minimize infection risk in blood banking and transfusion practice, Infect. Disord. Drug Targets 11, 45–56, 2011.

7

Plasma Proteinase Inhibitors

It is estimated that plasma proteinase inhibitors constitute some 10% of the total plasma proteins.1 Most of the proteinase inhibitors inhibit serine proteinase such as thrombin and are known by the collective term serpins (serine proteinase inhibitors).2,3 Serpins are considered to have a common reaction mechanism that can be considered as a stabilized acyl-enzyme intermediate.4 While the formation of an enzyme–inhibitor complex is reversible to the point of the formation of this intermediate, progression through the complete reaction results in the transition of the inhibitor from a virgin or native state to a modiied form which is inactive.5 A partial list of human plasma proteinase inhibitors is presented in Table 7.1. This list may be incomplete as new inhibitors continue to be discovered and the physiological relevance of others has not been irmly established.6–8 It is of some interest that Laskowski and Kato commented some 30 years ago4 that the precise physiological function of inhibitors is not known. Two plasma proteinase inhibitors—antithrombin (SERPINC1; earlier known as antithrombin III) and α-antiproteinase inhibitor (SERPINA1; earlier known as α-1-antitrypsin)—are of signiicant importance, as biopharmaceutical products and their complexes with proteases such as thrombin–antithrombin9 are valuable biomarkers. Other plasma proteinase inhibitors, such as tissue factor pathway inhibitor (TFPI)10,11 and C1 esterase inhibitor,12,13 are also of interest as commercial products, but do not have the project maturity of antithrombin and α-1-antitrypsin. One of the major advantages of inhibitors such as antithrombin is that they are biological buffers that react with enzymes such as thrombin in an equilibrium reaction similar to that of a substrate and enzymes; thus, it is most unlikely that hemorrhagic events are of importance as adverse events. Other inhibitors are heparin cofactor II, α2-macroglobulin, and α2-antiplasmin (SERPINF2 will also be discussed). Finally, both antithrombin and α1-antitrypsin have potentially important functions tangential to their function as inhibitors, while the antiprotease function of α2-macroglobulin may be truly secondary to other activities.

ANTITHROMBIN Antithrombin was not suficiently characterized as a protein to be included in Frank Putnam’s list of puriied plasma proteins in 1960.14 Antithrombin did merit inclusion in the second list15 published in 1975, in which it was deined as an α2-globulin of molecular weight 65,000 and a plasma concentration of 17–30 mg/100 mL. Commercial preparations have been derived from human plasma and recombinant DNA technology. The half-life of infused antithrombin is 2–3 days in normal subjects with either a recombinant or a plasma-derived product.16–18 There are only 285

Thrombin, factor Xa, factor IXa, factor XIIa, factor VIIa, matriptase

Leukocyte elastase; trypsin, plasmin, caspase-3

Many

65 kDa; 170–300 mg/L (2–5 μM); t1/2 = 2–3 days

50 kDa; 100–200 mg/L (4–8 μM); t1/2 = 3–5 days

720 kDa (tetramer; a dimer of disulide-linked dimers; monomer is 120 kDa); 2000 mg/L; 3 μM; t1/2 = 8–11 days (n = 6)

Inhibitor

Antithrombin (SERPINC1)

α1-Antitrypsin (α1-proteinase inhibitor; SERPINA1)

α2-Macroglobulin

Target Protease(s)d

Molecular Weight;a Plasma Concentration;b Half-Lifec

TABLE 7.1 Some Protease Inhibitors Found in Human Plasma Comments Originally described as antithrombin III and considered to be the primary inhibitor of thrombin and factor Xa. Reaction with target proteases is greatly accelerated by heparin. Plasma-derived and recombinant products are available for therapeutic use; approved for use in hereditary deiciency and in the study regarding its use in sepsis. Activity unrelated to blood coagulation is still being explored. α1-Antitrypsin deiciency is associated with chronic obstructive pulmonary disease. Severe deiciency results in abnormal protein retained in the hepatocyte, causing ibrosis. Plasma-derived and recombinant therapeutic products are available and gene therapy is being developed. It is clear that α1-antitrypsin has biological activity beyond protease inhibition. α2-Macroglobulin is not a serpin and “inactivates” enzymes by a trapping mechanism that does not modify the enzyme-active site of the target protease. The trapped target protein may retain full activity toward small substrates but greatly reduced activity toward high molecular substrates. α2-Macroglobulin has diverse biological activities in addition to inhibition of proteinase, including the binding of biologically active peptides such as cytokines.

References

[794–798]

[12,787–793]

[4,81,785,786]

286 Biotechnology of Plasma Proteins

65–70 kDa; 50–60 mg/L; ~1 μMe

65 kDa; 90 mg/L; approximately 1 μM; t1/2 = 2–3 days

71 kd; 70 mg/L; ~1 μM, t1/2 = 32.7 h

45–50 kDa, 20 μg/L; ~0.6 nM; t1/2 = 5–30 minf

α2-Antiplasmin (fast-acting plasmin inhibitor; SERPINF2)

Heparin cofactor II (SERPIND1)

C1-inhibitor (C1-esterase inhibitor; SERPING1)

Plasminogen activator inhibitor-1 (SERPINE1) Urokinase-type plasminogen activator; tissue plasminogen activator, thrombing

C1s, factor XIIa, factor XIa, plasmin, MASP-2, hepatitis virus NS3

Plasmin, trypsin α2-Antiplasmin is the primary protein inhibitor of plasmin in blood. α2-Antiplasmin can be covalently bound to ibrinogen in the circulation. There is interest in modifying α2-anitiplasmin activity to improve ibrinolysis. α2-Antiplasmin contains two disulide bridges, which can be reduced without losing activity and one cysteine that may or may not be in a mixed disulide. While heparin cofactor II can inhibit thrombin, it is not considered an important factor in the control of intravascular hemostasis. The majority of heparin cofactor II is in the extravascular space. Deinition of function for heparin cofactor II has not been accomplished. C1-inhibitor is the primary control of the classic pathway of complement formation by blocking the action of C1s on C2C4. C1-inhibitor also inhibits other proteases. The amino-terminal segment is rich in carbohydrate and may be important for C1-inhibitor function in inlammation. Plasminogen activator inhibitor-1 (PAI-1) is present in plasma in very low concentration and has a relatively short half-life for a native serpin. PAI-1 is a metastable protein that has stabilized interaction with vitronectin, both in the circulation and in the extravascular space. PAI-1 inhibits tissue plasminogen activator in the vascular space and urokinase-type plasminogen activator in the extravascular space. PAI-1 is synthesized and secreted from a variety of cell/tissue sources, including the endothelium and vesicular adipose tissue. PAI-1 has a broad range of activities, which may or may not be associated with protease inhibition. (continued)

[811–815]

[581,806–810]

[273,803–805]

[799–802]

Plasma Proteinase Inhibitors 287

57 kDa; 5.3 mg/L; 80–100 nM; t1/2 = 23 h

47 kDa(intracellular); 60–70 kDa (glycosylated and secreted)

Plasminogen activator inhibitor-2 (SERPINB2)

Molecular Weight;a Plasma Concentration;b Half-Lifec

Protein C inhibitor (SERPINA5); plasminogen activator inhibitor-3

Inhibitor

Urokinase-type plasminogen activator; tPA

Activated protein C, thrombin, factor Xa, urokinase, plasma kallikrein

Target Protease(s)d

TABLE 7.1 (Continued) Some Protease Inhibitors Found in Human Plasma

Protein C inhibitor (PCI) is the most signiicant of the various plasma protein proteinase inhibitors of activated protein C. PCI has a broad speciicity and a broad tissue distribution, suggesting that a critical function has yet to be described. The high concentration in semen has suggested a role in reproductive physiology. Plasminogen activator inhibitor-2 is present in the plasma of pregnant women being derived from the placenta. Plasminogen activator inhibitor-2 is also derived from other sources, including leukocytes and macrophages. Plasminogen activator inhibitor-2 is also suggested as a biomarker for certain tumors. As with other serpins in blood, a speciic role or roles remains to be elucidated.

Comments

[681,779,820–827]

[765,766,816–819]

References

288 Biotechnology of Plasma Proteins

g

f

e

d

c

b

a

53 kDa; ~4 mg/L; 73 nM

N/A

Thrombin-activatable ibrinolysis inhibitor (TAFI) was puriied from plasma as carboxypeptidase U, which exists in a precursor form bound to plasminogen. Once activated by thrombin, TAFI leaves carboxyl terminal lysine from ibrin decreasing ibrin afinity for plasminogen and decreasing the rate of ibrinolysis.

[828–833]

Approximate; accuracy depends on method(s) used. mg/L; molar concentration. Half-life given is for terminal phase in a two-compartment model. Many of the serpins are approximately equally distributed between intravascular and extravascular space. Signiicant targets listed; may not include all potential targets. Possibly considering how much α2-antiplasmin is covalently linked to ibrinogen.533 α and β phases for elimination. In the presence of heparin or vitronectin.

Thrombin-activatable ibrinolysis inhibitor (carboxypeptidase U; TAFI)

Plasma Proteinase Inhibitors 289

290

Biotechnology of Plasma Proteins

limited studies on the pharmacokinetics of the transgenic product,19,20 showing a shorter half-life for the recombinant product. The transgenic protein does differ in glycosylation patterns with respect to the plasma-derived protein,21,22 and it is possible that such differences inluence the half-life. The half-life of antithrombin is decreased in acquired antithrombin deiciency states.23–25 The decreased half-life in acquired antithrombin deiciency is likely a result of the increased rate of clearance of antithrombin–enzyme complexes. When thrombin is injected into the circulation, it appears to be immediately complexed with antithrombin.26–28 Coagulant enzymes such as factor VIIa and factor IXa, which are not rapidly inactivated by antithrombin, persist in the circulation for a longer period of time.29–31 Studies on the pharmacokinetics of antithrombin have, however, demonstrated a complex behavior that is seen with many other protein therapeutics. Two-compartment32 and three-compartment33 models have been proposed for the pharmacokinetics of antithrombin. A common observation in antithrombin pharmacokinetics studies is the rapid equilibration with the extravascular space.34 Collen and coworkers32 estimated that 45% of the antithrombin remained in the intravascular pool, while Carlson and coworkers33 estimated that 40% of the antithrombin was in the intravascular pool. Carlson and coworkers proposed a three-compartment model where 10% of the antithrombin was bound to the vascular wall in a noncirculating fraction. Both of these groups used radiolabeled proteins and demonstrated that the rapid initial loss of infused material was not due to denatured protein. Carlson and coworkers had performed earlier studies in a rabbit model system,35 where the issue of protein denaturation secondary to the iodination process was addressed by a “irst pass” of radiolabeled material through a rabbit, and plasma containing radiolabeled antithrombin was taken to a second rabbit. Antithrombin is active as a protease inhibitor in the extravascular space with matriptase.36 Variant forms of antithrombin are present in individuals with congenital antithrombin deiciency.37,38 Luxembourg and coworkers described 87 different mutations in a cohort of 272 patients with antithrombin deiciency. Thus, antithrombin deiciency is a heterogeneous disorder that is inherited as an autosomal defect, with most of the subjects being heterozygous.38 Mutations range from benign to clinically signiicant. Antithrombin deiciency is usually characterized by 50%–60% of normal activity. Antithrombin is glycosylated, and there is a naturally occurring form, β-antithrombin, which lacks glycosylation at asparagine-135.39 The presence of these two isoforms of antithrombin in normal plasma was established by Carlson and Atencio in 1982.40 However, these researchers noted that there were earlier studies that suggested the presence of multiple molecular forms. Peterson and Blackburn41 showed that the isoforms differed in glycosylation and afinity for heparin. The variant form with less carbohydrate has increased afinity for heparin. Later studies42,43 demonstrated that glycosylation at Asn135 inhibited the conformational change of antithrombin in the presence of heparin. There are variant forms of antithrombin in normal plasma and therapeutic concentrates, some with novel properties, relecting altered conformation. The thrombin–antithrombin complex can undergo cleavage44–46 resulting in the formation of a two-chain, disulide-linked derivative, which has lost inhibitory activity but

Plasma Proteinase Inhibitors

291

acquired antiangiogenic activity.47 Latent antithrombin (L-antithrombin) is a conformationally altered form of antithrombin that can be formed by heating (60°C) in the presence of citrate to avoid polymerization.48,49 Latent antithrombin has lost anticoagulant activity, including the ability to bind heparin tightly, but has acquired activity as demonstrated by O’Reilly and coworkers47 and Larsson and coworkers.50 Larsson and coworkers51 subsequently showed that a second conformationally altered form of antithrombin was formed in conditions of heat and citrate as shown by Wardell and coworkers.49 This form was designated prelatent antithrombin and was characterized by the retention of anticoagulant activity, heparin binding, and antiangiogenic activity. Karlsson and Winge52 used hydrophobic afinity chromatography to separate latent and native antithrombin. Subsequent work by these researchers53 demonstrated that heparin afinity was more effective and could separate prelatent, latent, and native antithrombin. These researchers and others54 have demonstrated the presence of prelatent and latent antithrombin in commercial concentrates most likely arising from the heating step used for viral inactivation. Latent antithrombin can also be found in recombinant antithrombin expressed in Chinese hamster ovary cells.55 Latent antithrombin and cleaved antithrombin are also found in normal blood plasma.56,57 It was suggested that elevated levels of latent antithrombin might be a risk factor for thrombosis on the basis of the association with the native antithrombin.58 However, Corral and coworkers59 have demonstrated that latent antithrombin does not pose a risk for thrombosis. These researchers also showed that latent human antithrombin and native human antithrombin were cleared at the same rate in a rat model system. A variety of mechanisms exist to control the blood coagulation process, including antithrombin. Other regulatory proteins such as TFPI and α2-macroglobulin are discussed elsewhere in this chapter, and other proteins such as protein C and protein S are discussed in Chapter 8. This chapter focuses on antithrombin, and it is useful to clarify some terminology before moving forward. A variety of phenomena have been described as antithrombin. Thus, thrombin is adsorbed to ibrin and released on ibrinolysis; this “antithrombin” activity was known as antithrombin I.60–62 Antithrombin II60 was a term used to distinguish progressive antithrombin activity from heparin cofactor activity.63,64 There was occasional use65 of ATII/III to describe these activities. Briginshaw and Shanberge63 were able to physically separate the two activities. Pratt and coworkers64 provide an excellent review of the development of heparin cofactor II, which is the term used now to describe antithrombin II. Antithrombin IV66 was described to have the ability to immediately inhibit thrombin upon formation. Antithrombin activity has been described in blood platelets,67 most likely relecting the various binding modes of thrombin to platelets.68 Verstrate69 provided some clariication in showing that there is one antithrombin which was designated as antithrombin III. Current nomenclature has dropped the Roman numeral designation, resulting in the use of the term antithrombin. However, there are other serpins in blood which can inhibit thrombin but which have other designations. Antithrombin as a physiological function has been known for more than 100 years but it was poorly understood.70,71 Steady progress in the understanding of antithrombin occurred over the years.72–75 The current world of antithrombin can be traced to the work of Bob Rosenberg and his colleagues at Harvard.76 I remember when

292

Biotechnology of Plasma Proteins

Bob presented the work at a Gordon Conference in the early 1970s. It was very well received and brought clariication into a cloudy ield. Subsequent work by Mike Grifith at the University of North Carolina at Chapel Hill77 and others clariied the fundamental relationship between antithrombin and heparin. While the reaction is greatly accelerated by the presence of heparin, antithrombin is a kinetically signiicant inhibitor (3.36 × 105 M/min) of thrombin.78 Antithrombin also inhibits other coagulation proteases, including factor Xa, in a reaction that is also accelerated by heparin. The use of low-molecular-weight heparin enhances the speciicity of the inactivation of factor Xa and is thought to be easier to manage79 with respect to adverse reactions, but there is the question regarding clear clinical advantage.80 The effect of heparin on the reaction of antithrombin with factor Xa is thought to relect the effect of a speciic heparin sequence on the conformation of antithrombin, while the reaction of antithrombin with thrombin in the presence of heparin is thought to involve a template process.81 There are two problems with the clinical use of antithrombin. The irst, which is discussed in more detail later, is the necessity of the early use of antithrombin in the clinical course of acquired antithrombin deiciencies such as sepsis. The second is the need for supranormal levels of antithrombin in the treatment of acquired antithrombin deiciencies.82 Inherited deiciency of antithrombin poses a similar therapeutic issue. A recent paper from a group of German researchers35 described the molecular basis of antithrombin deiciency. These researchers noted that inherited antithrombin deiciency is mostly heterozygous, leading to observed levels of 50%–60% of normal values (reference range); a decreased concentration of antithrombin is associated with an increased risk of thrombosis. As with acquired antithrombin deiciency, successful treatment of inherited antithrombin deiciency may require the initial establishment of higher than normal levels (120%) followed by maintenance at greater than 80%.83 While antithrombin is approved for use in congenital deiciency, the anticipated market is in acquired antithrombin deiciency, where even higher levels are used with marginal success.84,85 It is noted that in the pediatric study described earlier, the four subjects with disseminated intravascular coagulation/organ failure were treated with high-dose antithrombin in the absence of heparin with success. In addition to antithrombin, there are other SERPINS (antithrombin is SERPINC1) in the blood such that it is remarkable that blood does clot.86 The molar excess of inhibitors to enzymes is substantial,87 suggesting that enzymes should be inactivated immediately on formation. It is likely that the separation of clot formation from bulk solution is critical for protecting the activated enzymes from premature inactivation,88,89 as demonstrated by the effect of phospholipid, calcium ions, and factor V on the inactivation of factor Xa by antithrombin.90 Walker and Esmon90 do make the observation (which I missed some 30 years ago) that as antithrombin levels are extrapolated to ininity, the protective effect was eliminated and the rate of inhibition was accelerated. Reeve91 modeled the various reactions in coagulation to show that there was a reasonable explanation for increased susceptibility to thrombosis associated with modest reductions in antithrombin concentration. As cited earlier, there are studies84,85 in which there is a suggestion that antithrombin may be of value in acquired antithrombin deiciency such as that seen in sepsis.92,93 In

Plasma Proteinase Inhibitors

293

addition to these studies, there are other studies that support therapeutic effectiveness in sepsis94,95 when given at high levels and early in the clinical course.96 Inthorn and coworkers94 observed that antithrombin concentrate eliminated disseminated intravascular coagulation in severe sepsis. The multiorgan dysfunction (MOD) syndrome is a complex problem. Haire97 has an excellent discussion of the relationship of systemic inlammatory response syndrome (SIRS), MOD, and sepsis; sepsis can be deined as SIRS resulting from infection. He observes that the risk of progression from singleorgan dysfunction to MOD is substantial when antithrombin level was less than 84% of normal level. In previous studies, Haire and coworkers95 as well as Eid and coworkers96 emphasize the importance of the early use of antithrombin concentrate in obtaining a favorable therapeutic outcome. The following conclusions can be derived from the aforementioned information; it is understood that the data are not as solid as one would wish: • Congenital antithrombin deiciency is a heterogeneous disorder. • Acquired antithrombin deiciency is observed in a variety of clinical situations such as MOD in hematopoietic stem cell transplantation and sepsis. This is likely a relection of disseminated intravascular coagulation. Disseminated intravascular coagulation is relieved by the administration of antithrombin concentrate. • High levels of antithrombin concentrate may have a therapeutic effect in sepsis and related clinical problems. The effect is variable in a challenging the clinical population. A successful clinical outcome is dependent on early diagnosis and early use of the therapeutic product. • The conformational variants of the antithrombin, latent antithrombin and prelatent antithrombin, which have antiangiogenic activity, may be important in the therapeutic magic and variability of antithrombin in acquired antithrombin deiciency. It should be noted that this concept has been addressed by Heemskerk98 in a short comment.

α1-ANTITRYPSIN (α1-ANTIPROTEASE INHIBITOR, SERPINA1) α1-Antiprotease inhibitor (originally known as α1-antitrypsin) is a signiicant component of blood present at a concentration of 100–200 μg/mL.99–101 α 2-Macroglobulin has a similar concentration and is an eficient inhibitor of proteases97 but is secondary to α1-antitrypsin as a general physiologic proteinase inhibitor as α1-antitrypsin is small (and therefore has a higher molar concentration).102 α1-Antitrypsin is considered to be an acute phase protein.103–109 Serum levels of α1-antitrypsin increase during pregnancy110–112 and with the use of oral contraceptives.113 A cDNA for human α1-antitrypsin coding for a mature polypeptide of 394 amino acid residues has been isolated and sequenced114,115 and expressed in an Escherichia coli system yielding an unglycosylated protein with a mass of 45 kDa with antiproteinase activity.116,117 Recombinant α1-antitrypsin has been developed for clinical use as a parenteral drug118,119 and gene therapy is being developed.120

294

Biotechnology of Plasma Proteins

The mature α1-antitrypsin protein contains three carbohydrate chains (12% carbohydrate) and has a molecular mass of approximately 50 kDa.121–125 It is an α1-globulin (see Chapter 3), originally referred to as the 3.5 α1-glycoprotein,3 and contains a free cysteine residue.121,126 The presence of cysteine in α1-antitrypsin could be inferred from Laurell’s studies127 in 1970, showing the in vivo formation of a complex between immunoglobulin κ chains and α1-antitrypsin. Tomasi and Hauptman128 demonstrated α1-antitrypsin binding to IgA by the formation of disulide bonds. Musiani and coworkers129 showed that α1-antitrypsin maintains protease inhibitor activity when bound to IgA. Vaerman and coworkers130 showed that both albumin and α1-antitrypsin are bound via a disulide bridge to the same cysteine residue in the α-chain of IgA. IgA with bound α1-antitrypsin is found in fecal material, suggesting a serum origin of this material.131 Serum albumin also has a free sulfhydryl group (Chapter 4), which has been under considerable study. It is, thus, a bit surprising that the cysteine residues in α1-antitrypsin have been of little apparent interest. Grifiths and workers126 comment on this lack of interest in their paper on the reactivity of this cysteine residue in recombinant human albumin. In a work cited earlier, Crawford121 established the presence of one mole of cysteine/protein by performic acid oxidation as well as S-carboxymethylcysteine after reaction with iodoacetate. Musiani and Tomasi123 found one mole of cysteine/mole protein after reaction with iodoacetamide. Laurell and coworkers132,133 used disulide exchange to purify α1-antitrypsin from human plasma. Pierce and coworkers134 showed that conversion of α1-antitrypsin to a mixed disulide with cysteine improved resolution of α1-antitrypsin phenotypes. The pKa of the cysteine group in α1-antitrypsin has been determined to be 6.85 based on the pH dependence of reaction with hydrogen peroxide.126 This is approximately 1–1.5 pH units below the pKa of cysteine residues measured in glutathione (pH 9)135 and in a model pentapeptide (pH 8.6),136 but not unlike the average value (pH 6.8) for 25 proteins.137 The range for pKa values in proteins is 2.5–11.1, similar to that observed in albumin (Chapter 4). Naor and Jensen137 used computational methods to provide the basis for the low pKa of Cys232 in α1-antitrypsin, suggesting that the formation of a hydrogen bond with an amide is factor-responsible. The sulfhydryl group can be converted to the S-nitrosyl derivative by reaction with nitric oxide and the S-nitrosylated derivative has bacteriostatic activity.138,139 Subsequent work by this group showed that S-nitrosylated α1-antitrypsin has a protective effect on ischemia-reperfusion injury.140 The nitrosylation did not inluence the inhibition of trypsin or elastase. α1-Antitrypsin is a product of hepatocytes,141 although other sites of synthesis have been suggested.142 Putnam and coworkers141 showed that liver transplantation could cure the most severe deiciency of α1-antitrypsin (PiZZ) and that the phenotype (see later) of α1-antitrypsin matched that of the transplanted liver. Liver transplantation is used for the treatment of PiZZ α1-antitrypsin deiciency,143 where the abnormal protein is retained in the hepatocyte,144 which causes liver ibrosis.145 α1-Antitrypsin is considered to have a circulatory half-life of 3–5 days. Makino and Reed99 showed that there was a rapid distribution of α1-antitrypsin into the extravascular space after administration of partially puriied α1-antitrypsin to three patients who were homozygous for α1-antrypsin deiciency. Jones and coworkers146 studied the clearance of native and desialylated (disialylated) α1-antitrypsin using radiolabeled protein in

Plasma Proteinase Inhibitors

295

healthy human subjects (type MM). Approximately 50% of the native radiolabeled protein was taken rapidly to the extravascular space, while the desialylated protein was cleared from the circulation in a process associated with the uptake of radiolabel into the liver. The ratio of the amount of native α1-antitrypsin in the extravascular space to that in the intravascular space was approximately unity. α1-Antitrypsin deiciency is a complex disorder arising either from a congenital problem associated with the synthesis of defective protein or from an in vivo posttranslational modiication such as oxidation.142 The genetics of α1-antitrypsin deiciency is complicated and beyond the scope of this work. Sufice it to say that there are a range of variants from normal MM to the ZZ and Znull deiciencies.142,147 The reader is directed to several excellent discussions of this problem.148–151 α1-Antitrypsin deiciency is considered to be the major issue in the etiology of chronic obstructive pulmonary disease (COPD).152 The existence of α1-antitrypsin phenotypes was irst detected by Laurell and Eriksson in 1963 on the altered electrophoretic (paper/agar gel) pattern of α-globulin serum obtained from subjects with α1-antitrypsin deiciency.153 Subsequent work by Laurell154 using electrophoresis and immunoelectrophoresis to study serum samples from normal and α1-antitrypsin–deicient subjects obtained results, which showed that α1-antitrypsin heterogeneity could be explained either by multiple forms of α1-antitrypsin in serum or by the binding of α1-antitrypsin to another material. This study and a later study155 provided data, suggesting a relationship between α1-antitrypsin deiciency and obstructive lung disease. Fagerhol and Laurell156 used electrophoresis to identify homozygous variants of α1-antitrypsin and heterozygotes and developed Pi as an abbreviation to describe the polymorphic forms. Most of the variants are not associated with a major decrease in activity.157 Fagerhol157 showed that the MZ variant had the lowest activity (15% with 100% value from MM homozygous population) of seven phenotypes. The homozygous ZZ variant has only 15% activity. α1-Antitrypsin obtained its name on the basis of its ability to inhibit trypsin. As our understanding of the function of this protein increased, it was clear that while α1-antitrypsin did inhibit trypsin, the primary in vivo target was neutrophil elastase in preventing pulmonary damage.158–160 The broad spectrum of protease inhibition exhibited by α1-antitrypsin led Pannell and coworkers161 to use α1-antiproteinase inhibitor to refer to α1-antitrypsin. A committee on serpin nomenclature was convened in 1999 at the request of the HUGO Gene Nomenclature Committee, which suggested that SERPIN be used as the gene symbol for serpins.162 SERPINA1 was suggested as the term for α1-antiproteinase inhibitor/α1-antitrypsin. It is not clear whether α1-proteinase inhibitor or SERPINA1 has been greeted with enthusiasm by the scientiic community; a PUBMED search exclusive for α1-proteinase inhibitor garnered 1093 citations in June 2011, while SERPINA1 garnered 9. Thus, the author is comfortable with the use of α1-antitrypsin in the current work. The emphasis to this point has been the description of the protein and the congenital deiciency states. It is also important to note that the issue with pulmonary disease and α1-antitrypsin is the ability of neutrophil elastase to degrade pulmonary tissue (elastin), and α1-antitrypsin serves to inhibit this process. Likewise, it is important to note that α1-antitrypsin is an acute phase reactant and blood levels will increase with inlammation. COPD is a local disease resulting from the destruction of the

296

Biotechnology of Plasma Proteins

pulmonary tissue by neutrophil elastase, which is enhanced by a decrease in systemic levels of α1-antitrypsin. Effective prophylaxis requires inhibition of neutrophil elastase in the pulmonary bed, not in the circulation; α1-antitrypsin can inhibit pulmonary elastase, but delivery of therapeutic α1-antitrypsin must occur in the pulmonary bed. The molecular basis of α1-antitrypsin deiciency has been reviewed by Brantly and coworkers;144 the clinically signiicant ZZ has an allelic frequency of 0.01–0.02 in the caucasian population and is essentially absent in the black or Asian population. The presence of PiZZ also results in liver disease (see above). While COPD can result from the PiZZ deiciency state (≤ 1% of COPD can be ascribed to PiZZ individuals), the dominant cause of COPD is tobacco use, with occupation exposure and air pollution also being factors.163 Smoking (and other airborne reactive substances) results in the oxidation of a methionine residue at the “active site” of α1-antitrypsin.164–167 Oxidants such as myeloperoxidase derived from neutrophils can also contribute to in situ oxidation of α1-antitrypsin. To the naive person, such as the current author, it was a bit of a surprise then to ind that systemic levels of α1-antitrypsin are largely unchanged and sometimes increased in individuals who smoke cigarettes.168–171 As pointed out by Ogushi and coworkers,172 while there is compelling evidence for the protease–antiprotease concept173,174 for the development of emphysema in α1-antitrypsin deiciency, it was somewhat more dificult to rationalize the development of emphysema in cigarette smokers with normal or supranormal blood levels of α1-antitrypsin. There are several studies that provide some insight into this issue. First, the development of emphysema in cigarette smokers depends on individual phenotype. Mittman and coworkers175 showed that α1-trypsin deiciency predisposed individuals who smoked cigarettes to the development of emphysema; individuals with intermediate deiciency (heterozygotes with approximately 50% trypsin inhibitory capacity176) who smoked were far more likely to develop emphysema than a control of normal population who smoked. Lieberman176 had previously noted that homozygous-deicient individuals (ZZ) developed emphysema at 34 years of age, while heterozygotes developed emphysema at 44 years of age. The development of pulmonary pathology with smoking is a local, rather than a systemic, event.177 Hill and coworkers177 showed a reduction in interleukin-8 levels in ex-smokers with decreased neutrophil inlux. Individuals with α1-antitrypsin had greater airway inlammation. Ogushi and coworkers172 showed that the α1-antitrypsin isolated from the lavage luid of smokers was similar to that of α1-antitrypsin isolated from the lavage luid obtained from nonsmokers; proteins isolated from the lower respiratory tract showed decreased rate of reaction of α1-antitrypsin obtained from smokers compared to nonsmokers; the rate of inactivation of neutrophil elastase by α1-antitrypsin obtained from the lower respiratory tract of normal individuals was twice that of α1-antitrypsin from the lower respiratory tract of smokers. In addition to environmental oxidants, inactivation of α1-antitrypsin can be effected by the action of myeloperoxidase derived from neutrophils.178 This information is intended to support the use of α1-antitrypsin as a prophylactic agent for the treatment of pulmonary disease, secondary to α1-antitrypsin deiciency. The problem is that a therapeutic (α1-antitrypsin) is needed for local, not systemic, action in the lower lung. Aboussouan and Stoller179 suggest that only 2%–3% of the product is delivered to the pulmonary site with parenteral administration of an α1-antitrypsin therapeutic product.

Plasma Proteinase Inhibitors

297

The development of replacement/augmentation therapy with α1-antitrypsin for the treatment of COPD was suggested by the demonstration of a relationship between the α1-antitrypsin concentration and disease.155 Makino and Reed99 observed that it is impractical to give whole blood plasma to patients for hereditary α1-antitrypsin deiciency (PiZZ, 15%–25% activity180,181). Gadek and coworkers182 used a fraction derived from human plasma by poly(ethylene)glycol precipitation to restore protease–antiprotease173,174 balance in the lung; this was determined by the measurement of elastase inhibitory activity in the lower respiratory tract. The development of a plasma therapeutic α1-antitrypsin dates to the work of Michael Coan and coworkers183–187 at Cutter Laboratories in Berkeley, California, resulting in Prolastin-C,188 which is/was a product of Talecris Biologicals (Clayton, NC); Talecris is a successor to part of Bayer, which purchased Cutter Laboratories; Talecris has just been purchased by Grifols, a plasma product company based in Spain, which also purchased Alpha Therapeutics (Los Angeles, CA) from the Green Cross. Grifols also has a therapeutic product (Trypsone), Baxter Healthcare produces Aralast,189,190 and Zemaira is a product of CSL Behring. To the best of the author’s knowledge, these products are obtained from the Cohn Fraction IV(IV-1) (see Chapter 2). This fraction also serves as a source for the manufacture of antithrombin and the vitamin K–dependent factors (see Chapter 8). Development of α1-antitrypsin was initiated to maximize the use of this material, particularly after recombinant factor IX created marketing issues for the plasma-derived product. So, to some extent, the Cohn Fraction IV-1 was a waste fraction that was available for use.185 The same rationale was used for the puriication of ibrinogen for ibrin sealant from cold precipitate (see Chapters 2 and 9). It is quite likely that a superior product could be obtained by alternative puriication procedures.191,192 It is noted that a new process for puriication of α1-antitrypsin from the Cohn Fraction IV-1 had been developed, resulting in an improved product.193 It is not clear as to whether this work has resulted in a commercial product; Baxter has a new α1-antitrypsin product, Aralast-NP, but it was not possible to ind literature on this product. It is noted that the Baxter process193 provides a 15% yield from starting plasma, while the Cutter/Bayer/Talecris process185 provides a 50% yield from the Cohn Fraction IV (which has an approximate 40% yield from plasma) so that both processes have approximately the same yield. The Cutter/Bayer/Talecris group194 improved the yield from the Cohn Fraction IV-1 by 60%–70%. It is likely that the yield could be improved by using one of the alternative puriication processes.191,192 The development of alternative processes is discussed in Chapter 2. α1-Antitrypsin, as with antithrombin, can adopt a latent conformation195 associated with the loss of inhibitory activity. Lomas and coworkers195 prepared latent α1-antitrypsin by heating at 67°C in 0.7 M sodium citrate. The loss of activity could be partially reversed by refolding from 6.0 M guanidine hydrochloride. Latent α1-antitrypsin could be separated from the native protein by chromatography on a strong anion-exchange matrix (Q-Sepharose); resolution was also possible on native gel electrophoresis. Heating in the absence of citrate under these conditions of time and temperature resulted in polymerization of the protein and loss of activity. Lomas and coworkers195 did observe some polymerization with 0.7 M sodium citrate. Subsequent work from this group196 demonstrated the presence of latent α1-antitrypsin in a commercial preparation of α1-antitrypsin. It was suggested

298

Biotechnology of Plasma Proteins

that the heat step used for viral inactivation185 could be responsible for the presence of latent α1-antitrypsin in the inal drug product. However, this step involves heating at 60°C for 10 h in the presence of 0.38 M citrate, and Bottomley and Tew197 observed that 0.5 M sodium citrate stabilized α1-antitrypsin. Coan and Mitra198 had previously observed that 0.5 M sodium citrate (in the presence of sucrose) stabilized α1-antitrypsin to pasteurization (60°C/10 h). These results suggest that heat treatment is not responsible for the presence of latent α1-antitrypsin in the commercial product. The proteins in the Cohn Fraction IV and albumin fraction have been exposed to substantial concentrations of ethanol for a considerable period of time and, depending on the manufacturing process, the Cohn Fraction IV, the parent material for the α1-antitrypsin, may be frozen prior to subsequent processing. The concentration of ethanol is approximately 0.91 mole fraction for the preparation of the Cohn Fraction II + III, increasing to 0.163 for the preparation of the Cohn Fraction IV-1.199 There are also changes in ionic strength and pH and, while these processes occur in the cold, it takes days to complete the entire fractionation process.200 An organic solvent such as ethanol must be added to a concentrated protein solution such as plasma or cryo-poor plasma slowly to avoid local denaturation. Ethanol has been demonstrated to destabilize protein stucture,201–203 and St-Amour and coworkers204 demonstrated the unmasking of a cryptic IgG during ethanol plasma fractionation. These researchers describe this “unmasking” as a consequence of the “slightly denaturing” conditions of the Cohn fractionation. Denaturation originally referred to a process resulting in the loss of solubility but has evolved into a process deined as change in conformation without change in the covalent structure,205,206 which can be reversible or irreversible.207 Ethanol has been demonstrated to have an effect on the conformation of α1-antitrypsin. Jirgensons208 showed that the conformation of α1-antitrypsin was altered in the presence of 25% ethanol as shown by the changes in circular dichroism spectra. It was possible to reverse the changes by solvent exchange, but the extent of reversal depended on the time and temperature of ethanol exposure. These results provide the basis for the loss of α1-antitrypsin activity from starting plasma to the Cohn Fraction IV-1 precipitate.209 Zimmerman210 reported modiication of the Cohn Fraction IV-1 extraction step, which improved the yield of α1-antitrypsin. The assurance of reproducible alkaline pH of the Tris buffer and the addition of leuptin increased the yield of active material from the Cohn Fraction IV-1 paste. The leuptin likely blocks degradation by alcohol-stimulated proteases.211,212 Proteolytic inactivation of α1-antitrypsin by neutrophil metalloproteinases is thought to occur in vivo.213–215 The studies described have resulted in the development of several therapeutic concentrates derived from plasma and investigation in recombinant products and gene therapy. The use of α1-antitrypsin concentrates is referred to as an augmentation therapy rather than replacement therapy, as the therapy increases existing concentration. The use of therapeutic concentrates of α1-antitrypsin has given mixed results.216 Positive results have been obtained using augmentation therapy with individuals with severe α1-antitrypsin deiiciency,217 and such therapy appears to halt, but not reverse, further decline in COPD patients.218 Other researchers219,220 have suggested that intravenous augmentation therapy is not justiied on the basis of eficacy and cost. However, as noted by both Kueppers218 and Tonelli and Brantly,220 the therapeutic concentrates of α1-antitrypsin are the only therapeutic option, other than liver

Plasma Proteinase Inhibitors

299

transplantation, for the treatment of severe α1-antitrypsin deiciency. There are, however, other options for the treatment of acquired COPD.221–224 Several groups have advanced a personalized medicine approach to the parenteral use of α1-antitrypsin concentrates, which are therapeutically eficacious and cost-effective.225,226 The major issue with the use of α1-antitrypsin therapeutic concentrate is that the use is local, but parenteral administration is a systemic therapy.179 Since the need for therapeutic products is in the lower lung, it made sense to consider direct delivery to the pulmonary system. The author has had some experience in this area and is well aware that the pulmonary administration of a protein product is not a trivial exercise.227–229 The advantage is that α1-antitrypsin does not need to be adsorbed into the systemic circulation but is active in the lung. The major problem is formulating the protein product into a form that can be delivered by aerosol or related devices.227 It is possible that this is one problem that may be amenable to nanotechnology approaches.227 Hubbard and coworkers230 established a proof of principle for pulmonary delivery of α1-antitrypsin in an animal model. Sometime later, Kropp and coworkes231 showed that radiolabeled α1-antitrypsin could be delivered via inhalation to provide eficacy; 14.2% of administered α1-antitrypsin could be delivered to the pulmonary site, while 2% was delivered by parenteral means. Continued development of aerosol technology has led to a steady improvement in eficiency.232,233 The varied information presented earlier suggests that the parenteral use of therapeutic concentrates of α1-antitrypsin is useful with appropriate patient selection and pharmacokinetic analysis. Development of a pulmonary drug delivery system would be preferable to parenteral administration. The use of gene therapy for α1-trypsin deiciency234 is a potentially useful approach with the caveat that this is a technology still attempting to achieve anything close to the original expectations and/or concerns. Gene therapy for α1-antitrypsin deiciency is complicated in that severe (PiZZ) deiciency is associated with both liver and lung pathologies.235 The liver concern stems from the deposition of abnormal protein in the hepatocyte; as described by Greene and coworkers,235 this is a toxic gain-of-function mutation. COPD is the pulmonary disorder stemming from the loss of function as a result of the gain-of-function mutation. A gene therapy approach to the treatment of severe α1-antitrypsin deiciency must have two approaches: one is downregulation of the production of abnormal protein in the liver,234 while the other approach must solve augmentation of function in the lung. Considerable progress has been made in the development of expression systems for α1-antitrypsin236,237 and methods to deliver gene expression to the lung.238–241 Other biological activities of α1-antitrypsin may well be more commercially signiicant than the use in augmentation therapy for α1-antitrypsin deiciency. Work with latent antithrombin discussed earlier showed that while inhibitor activity was lost, antiangeogenic activity was generated. A latent form of α1-antitrypsin, which has lost inhibitor activity, but there was no comment about new activities, has been described.195 Breit and coworkers242 demonstrated that α1-antitrypsin inhibited lymphocyte response to phytohemagglutinin, suggesting that α1-antitrypsin could modulate the activation of T cells by monocytes. This study was based on immunological abnormality such as rheumatoid arthritis in individuals with α1-trypsin deiciency. A relationship is suggested between immune disorders and α1-antitrypsin.243,244

300

Biotechnology of Plasma Proteins

Lindström and coworkers244 do advise careful interpretations of changes in α1-antitrysin in patients with vasculitis, as α1-antitrypsin is an acute phase reactant, and deiciency could be masked by changes secondary to the presence of inlammation. Banda and coworkers245 showed that human α1-antitrypsin was inactivated by murine macrophage elastase; inactivated human α1-antitrypsin was a potent chemotactic factor for human neutrophils, suggesting a role in promoting inlammation. Stockley and coworkers246 showed that intact α1-antitrypsin inhibited neutrophil migration in response to FMLP, while oxidized α1-antitrypsin or a peptide cleaved from α1-antitrypsin increased cell migration. These researchers suggest that the observed inhibition of chemotaxis results directly from the protease-inhibitory activity; α1-antichymotrypsin is more potent as an inhibitor of chemotaxis, while antileukoprotease is less effective. The oxidized and cleaved forms of α1-antitrypsin are present in diseased lung tissue and might function to regulate the early phase of inlammation. The results do suggest that α1-antitrypsin has divergent effects on cell migration measured in vitro. There are a number of other studies247–252 that address the role of α1-antitrypsin in the inlammatory response, but considerable additional work is required to provide clariication. Bergin and coworkers253 showed that α1-antitrypsin could bind to IL-8, preventing the interaction of IL-8 with CXCR1 (chemokine receptor 1); α1-antitrypsin also binds to neutrophil membranes, thereby decreasing FcγIIIb release. Both these processes serve to regulate chemotaxis and may be important in the role of α1-antitrypsin in augmentation therapy. Subramaniyam and coworkers254 presented data (mouse model) showing that early administration of α1-antitrypsin enhances lipopolysaccharide (LPS)-induced cytokine/chemokine production in the pulmonary tissue. The general concept is that α1-antitrypsin has anti-inlammatory properties, while proteolysis/oxidation generates proinlammatory activity. There are also several studies on the role of α1-antitrypsin in angiogenesis.255–257 There is one report258 suggesting that elevated α1-antitrypsin is a risk factor for arterial ischemic stroke in childhood based on the inhibition of activated protein C; an earlier report259 suggested that a relationship between elevated levels of α1-antitrypsin and stroke is a function of elevation of inlammation-sensitive proteins (acute-phase proteins). There is another suggestion that α1-antitrypsin is a factor in the development of atherosclerosis on the basis of the inhibition of neutrophil elastase.260 Zsila261 has suggested a chaperone-like function for α1-trypsin in the clearance of defective proteins. Congote and colleagues have published a series of papers describing the activity of a C-terminal peptide derived from α1-antitrypsin in stimulating the proliferations of breast and liver cancer cells,262 blocking human immunodeiciency virus (HIV-1) infection,263 and promoting wound healing.264 There is certainly a collection of eclectic information concerning α1-antitrypsin, and it is possible that there are potential products to be obtained from a careful study of this area. It would be useful to know the effect of the Cohn fractionation process on the various observed activities. Four other plasma proteinase inhibitors require mention before moving to the complement issue. Several have been considered for development as therapeutic products but have not developed suficient traction for commercial development. Clarity of function is a major obstacle as product development has been a challenge with both antithrombin and α1-antitrypsin where it can be argued that function is understood.

Plasma Proteinase Inhibitors

301

Heparin cofactor II has been mentioned earlier in comparison to antithrombin and later with respect to the generation of chemotactic products resulting from proteolysis. α2-Macroglobulin is a large protein with a usual mechanism and an increasingly diverse number of putative functions. TFPI acts by inhibiting a complex involved in the “extrinsic coagulation pathway” and α2-antiplasmin inhibitor inhibits the ibrinolytic system.

HEPARIN COFACTOR II (SERPIND1) Heparin cofactor II is similar to antithrombin in that it is an inhibitor of thrombin in a reaction that can be stimulated by heparin. Heparin cofactor II may have been described earlier as antithrombin II60,63,64 or as one of the progressive antithrombin activities in plasma. While substantial portions of antithrombin and α1-antitrypsin are in the extravascular space, an even greater proportion of heparin cofactor II is found in the extravascular space.265,266 Hatton and coworkers265 proposed a threecompartment model for heparin cofactor II with 20% distribution in the intravascular space and 60%–70% in the extravascular space. It is then not surprising that heparin cofactor II is not thought to be an important factor in the regulation of solution blood coagulation but likely functions at the vascular wall or in the extravascular space.267 The tissue distribution data support this concept as well as the stimulation of the heparin cofactor II reactions by dermatan sulfate268 and smooth muscle cells.269 Heparin cofactor II was puriied and characterized by Tollefsen and coworkers270 at Washington University in Saint Lous, Missouri. Washington University has been responsible for major contributions to our understanding of hemostasis. Heparin cofactor II was shown to have a molecular weight of 65 kDa and is present in plasma at a concentration of approximately 1 μM.260 Heparin cofactor II inhibits thrombin in the absence of heparin (5 × 105 M/min) and is accelerated by the presence of heparin (4 × 108 M/min), resulting in a stabilized acyl-enzyme form which is inactive. Heparin cofactor II is not effective in the inhibition of factor Xa.223 Heparin cofactor II deiciency has been reviewed by Tollefsen271 and is not considered to be a signiicant issue in thrombosis risk. Heparin cofactor II will inhibit thrombin bound to ibrin in the presence of dermatan sulfate but not in the presence of heparin.272 Liaw272 and coworkers suggest that dermatan sulfate forms a ternary complex with thrombin and ibrin, which is not formed by heparin. Current thinking273 suggests that heparin cofactor II is important in the response to arterial injury. There is continued interest274 in the role of heparin cofactor II and dermatan sulfate in arterial lesions, but it is not clear whether such a role represents a therapeutic opportunity. A recent study275 showed that recombinant lamprey (Lampetra luviatrilis) angiotensinogen could inhibit human α-thrombin; primary sequence analysis shows considerable homology between the lamprey angiotensinogen and heparin factor II. Chemotactic products can be derived from heparin cofactor II.276–280

α2-MACROGLOBULIN α2-Macroglobulin is present in plasma at a concentration of approximately 2 mg/mL and is an α2-globulin on the basis of electrophoretic migration, which is found in the

302

Biotechnology of Plasma Proteins

Cohn Fraction II-III from alcohol fractionation of plasma.281–283 α2-Macroglobulin is unusual for a plasma protein in that it is a tetramer of 180 kDa subunits284 with a molecular weight of 720 kDa for the mature protein. α2-Macroglobulin has been reported to have a frictional ratio285 of 1.67, but some lower values have been reported.286 The value for frictional ratio argues for an asymmetric protein, but Kyte287 suggests caution in the interpretation of this assumption in the absence of intrinsic viscosity data. I was unable to ind intrinsic viscosity measurements for α2-macroglobulin; extracorporeal elimination of α2-macroglobulin has been shown to reduce blood viscosity improving luidity.288,289 Asymmetry of α-macroglobulin is also supported by electron microscopy.290 α2-Macroglobulin has the ability to bind reversibly or irreversibly to a variety of proteins and peptides.291 A variety of proteases bind to α2-macroglobulin via a reaction involving proteolysis of a “bait” region, which exposes an internal thioester resulting in the formation of a lysyl-γ-glutamyl peptide bond similar to those formed in transpeptidation reactions.292 The thioester bond is also seen in complement components C3 and C4.293–295 As with α2-macroglobulin, activation of complement component C4 exposes the thioester bond for reaction with various nucleophiles. The thioester in both complement C3 and α2-macroglobulin can also be exposed by reaction with primary amines.295 Mitchell and coworkers295 observed that activation of complement component C3 with methylamine enabled the formation of C3–protein complexes that are similar to those formed by physiological activation. A broad spectrum of proteases reacts with α2-macroglobulin,284 with broadspectrum proteases (digestive proteases) reacting more rapidly than more speciic proteases (regulatory proteases). Proteolysis of the “bait” region is associated with a conformational change in α2-macroglobulin.296 Native electrophoresis shows a transition from a “slow” form to a “fast” form. Electrophoresis under denaturing conditions shows 100 kDa and 85 kDa components as well as a 100 kDa component bound to the protease. This conformational change is associated with exposure of a hydrophobic region that is important in binding to the α2-macroglobulin receptor/ low-density lipoprotein receptor-related protein (CD91).297 The transpeptidation reaction of activated α2-macroglobulin as a consequence of activation results in the formation of a covalent bond with the protease. Differing from the reaction of proteases with antithrombin and α1-antitrypsin, which involves the enzyme active site, the reaction between α2-macroglobulin and protease allows for retention of activity.298 α2-Macroglobulin is one of several protease inhibitors in blood which inactivates enzymes in blood coagulation. Together with α1-antitrypsin, α2-macroglobulin can also inhibit enzymes unrelated to blood coagulation such as elastase, trypsin, and chymotrypsin. The ability to cleave a residue or residues in the “bait” region seems to be the major determinant of speciicity. Enzymes such as elastase or trypsin, which are digestive enzymes, are inhibited quite rapidly by α2-macroglobulin (6.4 × 105 M/s for pancreatic elastase299), while thrombin is inhibited more slowly (4.2 × 102 M/s).78,299 Downing and coworkers78 estimated that α2-macroglobulin is more than tenfold less effective than antithrombin in the inhibition of thrombin. Sottrup-Jensen284 argues that α2-macroglobulin is a broad-spectrum inhibitor that could trap proteases released from other inhibitors, proteases involved in ibrinolysis

Plasma Proteinase Inhibitors

303

and tissue remodeling, as well as proteases associated with invading pathogens.300 Chu and coworkers301 suggest that α2-macroglobulin is a protease-activated sensor for signal transmission. Borth302 and Chu and Pizzo303 summarized the broad spectrum of activities exhibited by activated α2-macroglobulin. α2-Macroglobulin was shown to activate B cells and T cells more than 30 years ago.304,305 James306,307 has reviewed the early work on the interaction between cytokines and α2-macroglobulin. James observes that much of the earlier observations on the effect of α2-macroglobulin on immune cells could be explained by bound bioactive substances.306 Binding to α2-macroglobulin results in activity modulation and clearance.308 Binding to α2-macroglobulin can also inluence the determination of cytokines in immunoassays.309 Binding of cytokines involves the “fast” or activated form of α2-macroglobulin, which would imply the rapid clearance of these complexes. On the other hand, complexes of peptides/proteins with α2-macroglobulin seem to persist long enough to permit consideration of isolation of cytokine complexes with α2-macroglobulin.310,311 In the latter study, Burgess and coworkers311 suggested that α2-macroglobulin complexes could be used as biomarkers for prostate cancer. French and workers312 showed that native α2-macroglobulin could form complexes with partially unfolded proteins; complexes were not formed with activated α2-macroglobulin. However, activation of the soluble complex by a protease could facilitate removal. Webb and coworkers313 suggested that α2-macroglobulin can regulate growth by binding the transforming growth factor beta (TGFβ). TGFβ can bind to native α2-macroglobulin, but binding is enhanced in a plasma–α2-macroglobulin complex; a similar effect was seen with thrombin. Native α2-macroglobulin has also been shown to form complexes with apolipoprotein E314 and with an osteogenic growth peptide.315 Complexing of antigen with α2-macroglobulin enhanced immunogenicity and has been proposed for use in vaccine manufacture.316–318 Another group has proposed the use of peptide–α2-macroglobulin complexes for the development of cancer vaccines.319–321 α2-Macroglobulin and its derivative forms have the potential to be used as biomarkers. Their use as a biomarker in prostate cancer has been described earlier.311 Prostate-speciic antigen, a member of the tissue kallikrein family, has been shown to form a complex with α2-macroglobulin.322,323 Misra and coworkers324 have reported that α2-macroglobulin forms a complex with prostate-speciic antigen, which binds to GRP78 on the surface of prostate cancer cells, activating the cells potentially and promoting more aggressive behavior. The levels of the TGFβ–α2-macroglobulin have been reported to be elevated in prostate cancer325 and may be of prognostic beneit. α2-Macroglobulin is used on a member of a multivariate test (FibroTest) for liver ibrosis.326–328 It has also been used in combination with vitamin D–binding protein and apolipoprotein A1 to evaluate liver ibrosis in hepatitis C.329 A highmolecular-weight protein, which could induce cardiac hypertrophy in rats, was found by Rajmanickam and coworkers330 and later identiied as a cardiac isoform of α2-macroglobulin that has differences in the region binding to the α2-macroglobulin receptor, but with identity in the sequence responsible for the interaction with growth factors.331 These researchers suggested that the differences in the receptor-binding region explain the change in targeting from liver to cardiac tissue. Injection of the puriied protein into normal rats resulted in the development of cardiac hypertrophy.332

304

Biotechnology of Plasma Proteins

Subsequent work showed the presence of this protein in humans associated with cardiac hypertrophy333 and suggested that the isoform may be of particular importance for cardiac changes in HIV and diabetic patients.334 α2-Macroglobulin has been suggested as a biomarker for glaucoma335 and is suggested to be neurotoxic on the basis of interaction with nerve growth factor blocking a neuroprotective effect via the TrkA receptor. Oh and coworkers336 suggested that α2-macroglobulin is a biomarker for the radiation of pneumonitis in lung cancer patients. There has been considerable interest in the involvement of α2-macroglobulin in Alzheimer’s disease.337–341 It is an acute-phase protein, so speciicity as a biomarker is complicated by the consideration of Alzheimer’s disease as an inlammatory disorder,342–344 and a recent review on biomarkers in Alzheimer’s disease345 did not include α2-macroglobulin. There is some interest in the potential function of it as a chaperone,346,347 and it has been shown that α2-macroglobulin inhibits β2-microglobulin amyloid formation.348 It is also suggested that complex formation of amyloid β-peptide prevents clearance across the blood–brain barrier.349 Both α1-antitrypsin and α2-macroglobulin are acute-phase proteins in that there can be a greater than 25% change (positive) in the concentration following an inlammatory stimulus.350 While acute-phase response implies a positive change in concentration, there are proteins such as albumin, apolipoproteins, and transferrin that are negative acute-phase reactants.351 Acute-phase response/inlammation is a confounding factor in the identiication of speciic biomarkers in pathologies such as Alzheimer’s disease and cancer, which have substantial inlammatory components.352

TISSUE FACTOR PATHWAY INHIBITOR When I started work on the proteins participating in blood coagulation in 1962, the blood coagulation process was viewed as a combination of two pathways: the intrinsic pathway and the extrinsic pathway. The conceptual separation between the two pathways is not very rigid today; however, the extrinsic concept continues in the literature.353 A defect in either pathway results in a bleeding problem. Current work in hemostasis supports the concept of the initiation of coagulation by tissue factor (extrinsic) followed by ampliication through the interactions of factor IX and factor VIII (intrinsic).354 This overlap between the extrinsic system and the intrinsic system has given rise to the use of the term tissue factor pathway to refer to the extrinsic pathway. Tissue factor is also known as tissue thromboplastin, which is considered to be a complete thromboplastin as opposed to the phospholipid derived from platelets (platelet factor 3) which is a partial or incomplete thromboplastin. The partial thromboplastin time derives its name from this distinction.355,356 The partial thromboplastin time (PTT) assay is used for the monitoring of heparin therapy and for the evaluation of hemophilia A and hemophilia B (see Chapters 6 and 8, respectively) initiated by glass or similar material.357–359 The prothrombin time (PT)360 is the diagnostic application of tissue factor and is used to monitor vitamin K antagonist therapy. The assay was developed using crude thromboplastin preparations derived from lung. Comparison of data between laboratories was a major

Plasma Proteinase Inhibitors

305

consideration as this assay was developed,360,361 and the controversy resulted in the International Normalized Ratio (INR).362 The sequence of reactions is initiated by the interaction of tissue factor with factor VII, forming factor VIIa in an autocatalytic reaction. The factor VIIa/tissue factor complex can then activate factor IX to factor IXa, providing a link between the extrinsic system and the intrinsic system. The factor VIIa/tissue factor complex also converts factor X to factor Xa; factor X is also converted to factor Xa by factor IXa. Factor Xa converts prothrombin to thrombin followed by platelet aggregation and ibrin formation. TFPI binds to factor Xa and subsequently to factor VIIa/tissue factor, forming a quaternary complex resulting in inhibition of the activity of factor VIIa. As such, TFPI functions to regulate the extrinsic pathway by inhibiting the activity of factor VIIa.363 However, a recent study of the clearance of recombinant factor VIIa364 shows persistence in plasma after parenteral administration for hours, not minutes as with other activated coagulation factors such as thrombin or factor Xa (see earlier), with the formation of complexes with antithrombin and α2-macroglobulin. Seligsohn and coworkers365 had previously reported the persistence of factor VIIa in the circulation after administration of factor IX concentrates. A congenital deiciency of tissue factor or TFPI has not been reported, suggesting that this could be a lethal mutation.366 A decrease in the plasma concentration of TFPI may be associated with an increased risk of thrombosis,366–368 and TFPI is reduced in disseminated intravascular coagulation but not in severe liver disease.369 The primary site of TFPI synthesis is the endothelial cell with contribution from platelets;370 so hepatic disease should not affect plasma levels unless related to a secondary disease process.371 The expression of TFPI is reduced in sepsis372,373 and may contribute to the associated coagulopathy.374 TFPI has been known in the past as placental protein 5,375 extrinsic pathway inhibitor,376 and lipoprotein-associated inhibitor.377 Placental protein 5 has subsequently shown to be identical with TFPI-2.337–380 Placental protein 5 was shown to interact with heparin381–385 and was suggested to be a product of tumors.386,387 Nisbet and coworkers387 found placental protein 5 to be present in a high percentage of gestational trophoblast tumors; one case had high levels of “free” placental protein 5 and disseminated intravascular coagulation. Jin and coworkers379 observed that expression of PP5/TFPI-2 decreased the invasive potential of human choriocarcinoma cells. The inhibition of tissue factor/factor VII by a plasma factor was described by Carson388 in 1981 who assigned the activity to plasma high-density lipoproteins. Inhibition of tissue factor/VIIa in plasma requiring factor Xa was subsequently observed by Morrison and Jesty.389 Sanders and coworkers390 observed the inhibition of tissue factor/factor VIIa by a factor in plasma in a reaction that requires factor Xa. Subsequent work from the Rapaport laboratory presented data supporting a two-step process for the reaction of extrinsic pathway inhibitors.391,392 The irst step is the reaction of the plasma factor, designated as extrinsic pathway inhibitor,391 with factor Xa, followed by a second reaction where the factor Xa–extrinsic pathway inhibitor is reacted with tissue factor/factor VIIa, forming a quaternary complex.392 Huang and coworkers393 observed that the rate of inhibition of factor Xa by TFPI was reduced in the presence of calcium ions, but subsequently the rate of inactivation increased by

306

Biotechnology of Plasma Proteins

phospholipid and factor Va, suggesting that factor Xa was more rapidly inactivated when present in the prothrombinase complex. This would appear to be contrary to the observations by Walker and Esmon90 who observed a decrease in the rate of factor Xa inactivation by anthrombin in the presence of other components of the prothrombinase complex. Later, Baugh and coworkers394 suggested that the inactivation of factor Xa by TFPI occurred mostly rapidly when factor Xa was physically close to tissue factor/factor VII. Broze and coworkers isolated lipoprotein-associated coagulation inhibitor produced in HepG23 (human hepatoma) cells395 in 1987 in a process that used afinity chromatography with immobilized bovine factor Xa using elution with benzamidine, a competitive inhibitor of factor Xa, to obtain the puriied inhibitor. Early work used factor Xa inactivated with diisopropylluorophosphate and was unsuccessful; the use of active factor Xa was also successful, yielding a homogeneous product (sodium dodecyl sulfate-polyacrylamide gel electrophoresis, SDS-PAGE) with a mass of approximately 39 kDa. The isolated material bound radiolabeled factor Xa (Western blot) and inhibited tissue factor-initiated clotting. The puriied material did not bind to factor Xa modiied at the enzyme active site with either diisopropyl phosphoroluoridate or the peptide chloromethylketone. The Washington University group subsequently isolated the lipoprotein-associated coagulation inhibitor from human plasma using a combination of hydrophobic afinity chromatography, ionexchange chromatography, and factor Xa afinity chromatography.396 The various studies cited resulted in a mechanism of action where TFPI is a multivalent protease inhibitor.397 There are other examples of multivalent protease inhibitors,4 including inter-α-trypsin inhibitor (inter-α-inhibitor).398–401 There is no evidence of a relationship between inter-α-inhibitor and TFPI.402 Factor Xa is not inhibited by inter-α-inhibitor but is weakly inhibited by bikunin (SPINT2)401 derived from interα-inhibitor.402 Earlier work with recombinant placental bikunin403 showed potent inhibition of human plasmin, plasma and tissue kallikrein, and factor XIa with lesser activity against factor Xa, factor XIa, and tissue factor/factor VIIa. Placental bikunin is composed of two linked Kunitz domains; individual domains were also expressed and showed reduced potency. Structural analysis of TFPI has been accomplished by molecular biology techniques.404,405 Wun and coworkers404 presented data supporting the basic structure of TFPI as three linked Kunitz domains with a negatively charged amino-terminal region and a positively charged carboxyl region. The sequence derived from the cDNA predicts several glycosylation sites which have been subsequently identiied by Nakahara and coworkers.405 Subsequent analysis of an endothelial cell library by Northern blotting demonstrated the presence of two separate messages—one a 1.4 kb domain and the other a 4.0 kb region,406 which contained the 1.4 kb message—for the expression of lipoprotein-associated coagulation inhibitor. One Kunitz domain binds factor Xa followed by the binding of factor VIIa/tissue factor to a second Kunitz domain. A precise function for the third domain remains to be established, but it has been suggested that the third domain interacts with plasma lipoproteins407 and cell surfaces387,408–410 and is important in the protein S stimulation of factor Xa inhibition by TFPI.411 The designation of a protease inhibitor domain as a Kunitz domain is based on a nomenclature developed some 30 years ago.4 The term

Plasma Proteinase Inhibitors

307

Kunitz is derived from Moses Kunitz who was responsible for much of the early work on protease inhibitors.412 The Kunitz domain is based on the structure of a pancreatic trypsin inhibitor, which was the irst to be crystallized, and from soybean trypsin inhibitor.413,414 The author met Professor Kunitz when they were both at the Rockefeller University in New York City. While Kunitz was at the end of his career, he remained a vital member of the community. I can recall the day when he came down the hall with a handful of papers to ind Stanford Moore; Professor Kunitz had just been elected to membership in the National Academy of Sciences and needed help with the associated paperwork. As a further note, the Worthington Biochemical Corporation owes its start to Kunitz, as Charles Worthington was a laboratory assistant with Kunitz at the Princeton laboratories of the Rockefeller Institute. While I do not know this, I suspect that Professor Kunitz would be somewhat embarrassed by the term “Kunitz domain.” At a time when computers trump laboratory work, it is useful to designate regions on proteins as domains, and homology is used to identify related domains. These domains, which usually have structural or functional implications, are named after an index protein resulting in, for example, sh3 domains, kinase domains, pdz domains, egf domains, and so on. As noted previously, the Kunitz domain is based on the structure of a pancreatic trypsin inhibitor; as far as I know, this is the only example of a protein domain named after a scientist. A deinition of a Kunitz domain was suggested by Markland and Ladner in the 1998 U.S. patent,415 which consisted of 50–60 amino acid sequence homologous to the bovine pancreatic trypsin inhibitor; there is particular reference to the position of the disulide bonds.4 The irst use of a Kunitz domain in the literature appears in the work of Hochstasser and Wachter416 in 1979. These researchers prepared a “Kunitz-type proteinase inhibitor” from inter-α-trypsin inhibitor by digestion or by isolation from urine. This material had an approximate molecular weight of 8 kDa, and the primary structure analysis showed homology with bovine pancreatic trypsin inhibitor. Kunitz domains are found in proteins such as amyloid precursor proteins, which are not intuitively identiied as protease inhibitors; the soluble form of amyloid precursor proteins is a potent protease inhibitor, also known as protease nexin 2.417 Kunitz domains may have function(s) other than protease inhibition suggested by possible roles in inlammation and tissue remodeling.418 Finally, Kunitz domains are not a prerequisite for inhibitor function, as α1-antitrypsin, for example, does not contain a Kunitz domain.419 Studies on the distribution of TFPI in the body suggest that the presence of TFPI in the vascular space is a secondary phenomena, with the focal point of activity in the extravascular space. Some 80%–90% of TFPI is associated with the endothelial cell surface420 and the TFPI in plasma is associated with lipoprotein.421 A small amount of TFPI is produced by platelets.422 Tissue factor pathway is also subject to proteolytic degradation, resulting in degradation and loss of activity.418,423,424 Kothari and coworkers reported that while thrombin increased tissue factor activity in endothelial cell and pleural mesoendothelial cell culture, plasmin increased tissue factor activity by inactivating cell-associated TFPI.425 In addition to the heterogeneity associated with the binding to lipoprotein and limited proteolysis, Piro and Broze reported two forms of TFPI—TFPIα and TFPIβ—which are suggested

308

Biotechnology of Plasma Proteins

to differ in the extent of glycosylation and the chemistry of attachment to the endothelial surface.426,427 In addition, there is TFPI-2.380 TFPI-2 expression in monocytes is stimulated by thrombin.428 Schuepbach and coworkers429 showed that endothelial cell tissue factor activity is enhanced by activated protein C in a mechanism involving cleavage of TFPI with the release of the irst Kunitz domain, which is thought to interact with tissue factor/factor VIIa. TFPI is released from the endothelial cell surface with heparin.430–436 Harenberg and coworkers437 reported that the heparininduced mobilization of TFPI could be reversed by protamine; these researchers also reported that dermatan sulfate was much less effective than heparin in the mobilization of TFPI, implying speciicity in the structure of sulfated glycosaminoglycans in the reaction. Valentin and coworkers438 reported that the presence of heparin and TFPI may inluence coagulation assays. Heparanase is an enzyme that hydrolyzes the heparan sulfate proteoglycans on the endothelial surface; it is thought that these highly negatively charged proteoglycans are responsible for binding the positively charged carboxyl terminal region of TFPI.437 Heparanase has been shown to release TFPI, creating a procoagulant effect.439,440 TFPI has been suggested to have activities other than those in coagulation;441 however, the caveat must be made that, in principle, TFPI controls the expression of other biologically active substances such as factor Xa, thrombin, and plateletderived growth factors. Animal model studies have shown that TFPI exhibits antiangiogenic and antimetastatic activity,442 and it is possible that some of the nonanticoagulant activity of low-molecular-weight heparin might be related to the release of TFPI.434,435 Tissue factor is suggested to promote angiogenesis;443–446 such inhibition of tissue factor and/or tissue factor expression can be antiangiogenic.447–449 A second form of TFPI—TFPI-2—is upregulated in tumor cells and appears to induce apoptosis and inhibit angiogenesis.450 In addition to TFPI-2, there are alternatively spliced forms of TFPI—TFPIα and TFPIβ—that are produced by endothelial cells;451 both TFPIα and TFPIβ induce apoptosis in breast cancer cells.452 It is reported that the expression of TFPIα and TFPIβ is developmentally regulated; both TFPIα and TFPIβ are expressed in murine embryonic tissue, while only TFPIβ is expressed in murine adult tissue.453 Van Den Boogaard and coworkers454 reported that recombinant TFPI reduced bacterial growth (Streptococcus pneumonia) in a murine model of community-acquired pneumonia with concurrent antibiotic therapy. There was a decrease in neutrophil iniltration into pulmonary tissue and reduction in cytokines and chemokines with the use of recombinant TFPI; however, these effects were only observed in the absence of antibiotics. Other researchers455 have demonstrated an antibacterial effect of TFPI fragments in vitro against resistant E. coli. Regnault and coworkers456 show a positive relationship between increased free TFPI and arterial stiffness and suggest that free TFPI increases as vascular wall function deteriorates, with a possible relationship between free TFPI and cardiovascular risk. The aforementioned studies provide a limited basis for the use of TFPI as a diagnostic tool and as a therapeutic agent. The studies by Regnault and coworkers456 are suggestive, but as the authors note, a substantial longitudinal study is required to establish its value as a predictive biomarker. A clinical trial on the use of recombinant TFPI in community-acquired pneumonia showed a reduction in some

Plasma Proteinase Inhibitors

309

coagulation-based biomarkers (prothrombin fragment 1 + 2, thrombin–antithrombin) and there was no effect on mortality.457 An earlier clinical trial458 with TFPI in sepsis suggested a reduction in the mortality, but there was no signiicant difference between the treated and the control groups. A subsequent larger trial459 in severe sepsis showed no difference in mortality (all-cause 28-day mortality) between treated and placebo; there was an increase in bleeding in the treatment group. This is not unlike the trials with antithrombin, which suggest to the writer who is not a clinician, that success will require more careful patient selection and also a better understanding of the relationship between free TFPI and TFPI bound to endothelial cells. In work cited earlier, Regnault and coworkers456 suggest that increased plasma TFPI is predictive of cardiovascular death. Earlier studies by Morange and coworkers460 reported that free plasma TFPI was associated with the severity of cardiovascular damage on admission but not independently related to outcome; free soluble tissue factor was predictive of cardiovascular mortality in acute coronary syndrome. This information suggests that TFPI functions bound to a membrane and that plasma levels may be an indication of vascular pathology.461–468 Kamikura and colleagues461 observed elevated plasma TFPI in patients with acute myocardial infarction and suggested that it was released from ischemic tissues. This same research group 462 observed elevated TFPI in disseminated intravascular coagulation and suggested that such elevation was a result of injury to the vascular endothelium. Mitchell and coworkers463 presented data suggesting that TFPI is elevated in atherosclerosis as a result of endothelial dysfunction. Yasuda and coworkers464 showed elevated TFPI in acute pancreatitis. Decreases in free TFPI were observed in COPD patients,465 resulting in increased thrombin generation. Golino and coworkers466 observed consumption of TFPI in acute coronary syndrome and Ignjatovic and coworkers467 observed an increase in TFPI with heparin administration in coronary bypass surgery. Golino and coworkers466 also reported an increase in TFPI with heparin administration. Van Dreden and coworkers468 reported a decrease in TFPI in factor V Leiden. The use of TFPI as a parenteral therapeutic or biomarker is complicated, and the extremely rapid clearance of TFPI from the circulation is characterized by a rapid dissociation/reassociation from the vascular endothelium. Donahue and coworkers469 studied the disposition of TFPI during cardiopulmonary bypass; as would be expected, the administration of heparin resulted in the release of TFPI and the administration of protamine resulted in a decrease of full-length TFPI while free TFPI antigen and total plasma TFPI remained elevated. Electrophoresis/Western blot analysis showed that TFPI remaining in the solution after protamine was a 38 kDa form missing the C-terminal domain. Kemme and coworkers470 observed an increase in the rate of TFPI release at low-dose heparin, which did not increase with additional heparin and decreased upon stopping heparin administration. The most striking aspect of studies on the pharmacokinetics of TFPI is the very rapid irst phase (α-phase) of clearance, which is on the order of seconds to minutes, followed by a second phase (β-phase), which is in the range of 1–3 h.471 The α-phase likely represents the rapid association of TFPI with sulfated proteoglycans on the vascular endothelium and, as would be expected, the administration of heparin decreased the rate of clearance of full-length TFPI (recovery at 2 min for

310

Biotechnology of Plasma Proteins

full-length TFPI of 3.2 min vs. 45.9 min in the presence of low-molecular-weight heparin).472 These researchers showed that the α-phase for a TFPI derivative lacking the C-terminal positively charged domain (two-domain TFPI) was longer (recovery at 2 min of 48.8%) and was not inluenced by heparin to the same extent as the fulllength material (70.5 min). This group subsequently reported473 that a nonglycosylated derivative lacking the Kunitz domain 3 (two-domain TFPI) had similar α-phase clearance to the glycosylated two-domain TFPI but a longer β-phase. Other studies also support a very rapid clearance of TFPI.474 There has been considerable interest in the use of factor VIIa to treat factor VIII inhibitors (see Chapter 6). Van’t Veer and coworkers475 have suggested that the rapid inactivation of tissue factor/factor VIIa by TFPI, combined with the absence of factor VIII in hemophilia A or factor IX in hemophilia B, is responsible for the hemorrhagic diathesis in these disorders. Erhardtsen and coworkers476 demonstrated a shortening of the bleeding time in “hemophilic rabbits” with an antibody against TFPI, suggesting that this might be an approach to the treatment of hemophilia. Liu and coworkers477 reported inhibition of TFPI with nonanticoagulant sulfated polysaccharides and suggested that this as an approach for the treatment of the hemophiloid disorders. More recently, an aptamer, which inhibits TFPI, has been developed as an approach to treat hemophilia.478,479 There does not appear to be a rationale for the use of TFPI as a parenteral drug, as the clearance rate is quite rapid. It could be argued that the rapid clearance increases concentration at the site of action. On the other hand, a deiciency state with consequences remains to be demonstrated. The rapid clearance is likely due to rapid binding to the vascular wall; however, the β-phase is quite rapid as well and likely represents renal clearance. It is possible that modiication with poly(ethylene)glycol would decrease renal clearance and might decrease binding to the vascular wall. However, it is not clear that TFPI functions in the luid phase and probably requires membrane binding to function. Inhibition of TFPI is a mechanism by which the activity of factor VIIa would be increased in those providing a therapeutic approach to the treatment of hemophilia. This seems to the author to be a somewhat convoluted approach to solving a clinical problem. A bit of history on the development of factor VIIa might be useful here (see also Chapter 8). It is also useful to consider Walt Kisiel’s relections480 on the development of factor VIIa which resulted in the development of the Novo Nordisk product (Novo7 RT) for the treatment of hemophilia A patients with inhibitors. Prothrombin complex concentrates were plasma therapeutic concentrates derived from the Cohn Fraction IV and were used to treat patients with factor IX deiciency (Christmas disease) and liver disease.481 Physicians also found some of these concentrates to be useful in the treatment of hemophilia A patients with inhibitors.482 Thrombogenic activities were found in these early prothrombin complex concentrates,483 raising concern about their continued clinical use.484 The companies responsible for the manufacture of the prothrombin complex concentrates made changes in the manufacturing process resulting in therapeutic products that were not thrombogenic but were no longer as useful in the treatment of factor VIII inhibitors. As a result, prothrombin complex concentrates, which were deliberately activated, were developed for the treatment of factor VIII inhibitor patients starting with Autoplex.485–487 Analysis of the composition of Autoplex showed the presence

Plasma Proteinase Inhibitors

311

of large amounts of factor VIIa and factor IXa with smaller amounts of factor Xa.29 It was not unreasonable then to use puriied factor VIIa for the treatment of inhibitors in hemophilia A patients, and an approach, such as the use of a speciic aptamer, which would decrease the activity of TFPI, would be expected to increase factor VIIa activity.

α2-ANTIPLASMIN α2-Antiplasmin (SERPINF2, fast-reacting inhibitor of plasmin) is the primary physiological inhibitor of plasmin, the enzyme responsible for the dissolution of ibrin clots (ibrinolysis). Plasmin is derived from plasminogen in the vascular space by tissue plasminogen activator (tPA) and by urokinase in the extravascular/interstitial space. The reader is recommended to a group of excellent reviews on the ibrinolytic system.488–493 α2-Antiplasmin was originally described as the fast-reacting plasmin inhibitor of plasma494 as opposed to a slower-acting inhibitor, subsequently shown to be α2-macroglobulin. α2-Antiplasmin was shown to be distinct from α1-antitrypsin;495 The former has been shown to inhibit both chymotrypsin and trypsin.496 The rate of inactivation of chymotrypsin by α2-antiplasmin was similar to those observed for plasmin (~7 × 105 M/s), while trypsin is inhibited more rapidly (1.3 × 107 M/s) than plasmin. Nobar and coworkers497 observed the rapid inhibition of trypsin by antiplasmin and by antithrombin; the inhibition of trypsin by α2-antiplasmin was more rapid than by α1-antitrypsin. Levi and coworkers498 developed speciic immunoassays for plasmin–protease inhibitors in plasma. As would be expected, the majority of plasmin was associated with α2-antiplasmin with a far smaller amount complexed with α2-macroglobulin; smaller amounts of plasmin were associated with antithrombin, α1-antitrypsin, or C1 inhibitor. The distribution did depend on the subject; patients with DIC or on thrombolytic therapy had higher levels of plasmin–α1-antitrypsin and plasmin–antithrombin than did healthy controls. A later study by Banbula and coworkers499 showed that whole blood had increased plasmin inhibitor activity. C1-inhibitor is susceptible to digestion by plasmin;500,501 Brown and coworkers500 suggest that susceptibility of Ci-IHN, to digestion by plasmin relects the presence of a partially denatured, polymerized form of Ci-INH, which is formed on storage or on heating. Latent forms of antithrombin and α1-antitrypsin with reduced potency as inhibitors have been described earlier. Both α2-antiplasmin and C1-inhibitor are inactivated with proteolytic digestion by elastase derived from neutrophils.502 Another consideration is the overall mechanism, with the exception of α2-macroglobulin, for protease inhibition by protein inhibitors which involves the formation of a stabilized acyl-enzyme or tetrahedral intermediate.4,503 Sazonova and coworkers504 converted α2-antiplasmin from an inhibitor to a substrate for plasmin by using a monoclonal antibody and suggested that binding of the monoclonal body promoted the dissociation of the acyl-enzyme/tetrahedral intermediate to cleaved peptide chains and free enzyme. Congenital deiciency of α2-antiplasmin is a rare disorder,505 with some 40 cases reported until 2008. Acquired deiciency of α2-antiplasmin is observed in a variety of clinical conditions, including liver cirrhosis.506,507 Decreased α2-antiplasmin is also seen in disseminated intravascular coagulation.507 Whether congenital or acquired, α-antiplasmin deiciency is characterized by increased bleeding.508,509

312

Biotechnology of Plasma Proteins

α2-Antiplasmin deiciency can also occur with ibrinolytic therapy.510 Simpson and coworkers511 have recently shown that plasmin generation in plasma was enhanced by α2-antiplasmin deiciency or excess tPA; plasmin generation was decreased with ibrinogen deiciency. Treatment of α 2-antiplasmin deiciency (excess ibrinolysis) is based on the use of plasmin inhibitors. The major inhibitor is ɛ-aminocaproic acid (EACA; 6-aminohexanoic acid; Amicar).512 ɛ-Aminocaproic acid is used to treat α2-antiplasmin deiciency as well as to supplement endogenous α2-antiplasmin513,514 in various surgical procedures with more use since the removal of aprotinin (bovine pancreatic trypsin inhibitor; Trasylol) from the market.515 Aprotinin is a potent inhibitor of plasmin516 and was included in early ibrin sealant products to improve derivative product stability.517 Aprotinin was removed from the market as a result of adverse reactions but is in the process of a clinical trial to provide more information.518,519 As far as I know, there is neither a plasma-derived α2-antiplasmin therapeutic nor one obtained by recombinant DNA technology. I do recall being in meetings, both industrial and academic, where such a product was discussed. Such a product never came to fruition most likely because of the very rare nature of congenital deiciency, the complex nature of the acquired deiciency being secondary to a primary pathology, and the success of EACA and aprotinin. The clinical concerns regarding α2-plasmin activity have focused on increasing focal ibrinolysis (reducing the effectiveness of α2-antiplasmin). The use of a monoclonal antibody to convert α2-antiplasmin from an inhibitor to a substrate504 was mentioned earlier. Lee and coworkers520 identiied an enzyme (antiplasmin-cleaving enzyme) that converts native α2-antiplasmin, a single polypeptide chain of 464 residues with an N-terminal methionine, to a 452 residue with an N-terminal asparagine. The 452 residue inhibitor is cross-linked to ibrin521 more rapidly than the native protein, providing more resistance to clot lysis. Lee and coworkers520 observed that clot lysis times were reduced as the amount of the 452 residue material decreases. These researchers suggest that blocking the activity of the antiplasmin-cleaving enzyme would promote ibrinolysis. α2-Antiplasmin is a single-chain protein of molecular weight 65–70 kDa containing four carbohydrate side chains in the human protein together with a sulfated tyrosine residue.493,522 The sequence of what appeared to be the mature protein was determined by cDNA technology in 1987 by two groups.523,524 These researchers presented data supporting a 452 residue mature protein with an N-terminal asparagine associated with a 30+ residue signal peptide. Lijnen and coworkers525 reported the sequence of a 452 residue human α2-antiplasmin obtained by classical protein chemistry with two differences from the cDNA sequence. Glucosamine-based carbohydrate is present in four asparagine residues. Högstorp and Carlin526 showed that glycosylation was not necessary for the secretion of α2-antiplasmin from rat hepatocytes in culture or for subsequent binding to plasminogen kringles. These researchers obtained evidence for two disulide bonds. Koyama and coworkers527 subsequently isolated two forms of α2-plasmin inhibitor from plasma and Hep G2 cell culture by the use of immunoafinity chromatography. The major component isolated from plasma was the 452 residue protein, while a 464 residue protein with N-terminal methionine was isolated from the serum-free conditioned media from Hep G2 cells. In a work cited

Plasma Proteinase Inhibitors

313

earlier, Lee and coworkers420 described an enzyme, antiplasmin-cleaving enzyme, which converted the high-molecular-weight form (464 residues) to the lower-molecularweight form (452 residues). The x-ray crystal structure of a truncated form of the murine protein has been reported.528 The sum of the various structural studies supports a model containing a serpin domain529,530 with amino- and carboxyl-terminal extensions. The carboxyl terminal contains multiple lysine residues, which interact with plasminogen kringles,529 and the amino-terminal region can be cross-linked to ibrin531,532 and ibrinogen533 in transpeptidation reactions catalyzed by factor XIIIa. Mosesson and coworkers533 showed that α2-antiplasmin was covalently linked to ibrinogen via a transpeptidation reaction catalyzed by factor XIII providing resistance to ibrinolysis. This work provided considerable insight into an earlier observation by Mosesson and Finlayson534 who demonstrated heterogeneity of puriied human plasma ibrinogen on DEAE-cellulose chromatography. There was one fraction with proibrinolytic activity and another fraction with ibrinolysis inhibitory activity; inhibition was demonstrated against both trypsin and plasmin. It is possible to identify this inhibitory activity as α2-antiplasmin covalently bound to ibrinogen. Mosesson and coworkers533 showed at least stoichiometric amounts of α2-antiplasmin associated with ibrinogen, which implies a higher concentration of α2-antiplasmin in plasma (10–15 μM) than the ~1 μM value, that is generally accepted.493 This observation would imply resistance to ibrinolysis intrinsic within plasma ibrinogen. There are several subsequent studies which suggest that this is the situation. Jámbor and coworkers535 showed that the addition of a polyclonal antibody to the whole increased observed ibrinolytic activity as measured by thromboelastography. When I was just starting in graduate school years ago, the use of the thromboelastogram along with the two-stage assay for factor VIII seemed to have vanished from the landscape. The two-stage assay has been reinvented as the chromogenic assay, while the use of the thromboelastogram is gaining in popularity.536,537 There are, of course, several reasons for the increased ibrinolysis observed by Jámbor and coworkers, such as the lack of cross-linked ibrin, but it is not unreasonable to consider the lack of α2-antiplasmin incorporation into ibrinogen as a possibility. Mutch and workers538 showed that a model thrombus in a Chander loop539,540 prepared from factor XIII–deicient blood supplemented with a factor XIII therapeutic concentrate was more resistant to ibrinolysis than with a thrombi prepared from unsupplemented blood. In a related study, Tsurupa and coworkers541 showed noncovalent binding of α2-antiplasmin to ibrin and ibrinogen bound to a surface; binding was not shown to occur with ibrinogen in solution. Zamarron and coworkers542 used monoclonal antibodies to demonstrate conformational changes in ibrinogen bound to a surface similar to those seen in the conversion of ibrinogen to ibrin. There are observations of differences in the relationship of α2-antiplasmin with the resistance to ibrinolysis. Morris and coworkers543 found that ibrin (crude ibrinogen and bovine thrombin) prepared from blood obtained from patients with chronic thromboembolic pulmonary hypertension is resistant to lysis, but there was no relationship between resistance to lysis and content of α2-antiplasmin. The reactive center loop (RCL) is located in the serpin domain of α2-antiplasmin.544,545 Initiation of the cleavage of a speciic peptide bond in the RCL by the target protease results in a conformational change in the serpin with stabilization of the

314

Biotechnology of Plasma Proteins

tetrahedral intermediate/acyl enzyme.546 The RCL in α2-antiplasmin contains the “active site,” Arg364-Met,365 for interaction with plasmin as shown by Shieh and Travis.547 Identiication of this site was accomplished by using trypsin instead of plasmin to form a complex with α2-antiplasmin and taking the mixture directly to the Sequenator. α2-Antiplasmin, depending on species, contains three or four cysteine residues. The human protein has four cysteine residues, two of which are participants in a single disulide bond, with the other two either in a mixed disulide or a free cysteine.548 The bovine protein549 and rabbit protein550 have three cysteine residues with one disulide and present either as a free cysteine residue or mixed disulide. The study on the human protein548 reports differences in the interpretation obtained from earlier primary structure studies.525 It is not possible to resolve this without further study, but it is not consequential as the sulfhydryl/disulide is not important for the function (or structure). Reduction of the disulide bond does not markedly inluence inhibitor activity (9.5 × 106 M/s for native protein vs. 5.3 × 106 M/s for the reduced protein).548 Both the native and reduced proteins unfolded at 3.0 M urea (transverse urea gradient—polyacrylamide gel electrophoresis551–553). The complex of either protein, native or reduced α2-antiplasmin, with trypsin was stable to at least 8.0 M urea using the same analytical techniques. The implication here is that α2-antiplasmin is more stable in the complex than in free solution as is trypsin as measured by physical techniques. Trypsin, as judged by analytical ultracentrifugation, undergoes unfolding between 3.0 M and 5.0 M urea.554 Delaage and Lazdunski555 observed that the binding of trypsin to a protease inhibitor stabilized the bound trypsin to denaturation in the presence of urea as measured by exposure of tyrosine and tryptophan residues when measured by difference spectroscopy. These researchers suggest that stability of trypsin increases with increasing points of contact with the inhibitor. Stevens and Doskoch556 showed that while the eight disulide bonds of lima bean trypsin inhibitor in free solution were reduced with dithiothreitol in the loss of inhibitory activity against either trypsin or chymotrypsin, only one disulide bond was susceptible to reduction when the inhibitor was complexed to trypsin. It would appear that α2-antiplasmin increases in stability in a complex with plasmin. The x-ray structure of α2-antiplasmin was reported by Law and workers.528 These researchers noted that the C-terminal extension could serve as a binding site to move the RCL of α2-antiplasmin into position to act as a “substrate” for plasmin. These researchers also note that α2-antiplasmin is an F-clade serpin,557 suggesting potential antiangiogenic activity as discussed earlier for antithrombin. There are few studies on the stability and conformation of α2-antiplamin, as there was little interest in the development of a therapeutic product, derived either from plasma or via recombinant DNA technology. Implicit in the discussion of the formation of the α2-antiplasmin complex is the suggestion that α2-antiplasmin undergoes a conformation change on the binding to and formation of a complex with plasmin. This concept is supported in studies by Plow and coworkers558 who demonstrated differences in antigenic determinants in α2-antiplasmin on the formation of a complex with plasmin and later by circular dichroism studies.559 Nilsson and coworkers559 also showed that the reduction of α2-antiplasmin, which does not result in the loss of activity, does result in major changes in structure (circular dichroism). Solvent–detergent is a method developed at the New York Blood Center to inactivate lipid-enveloped

Plasma Proteinase Inhibitors

315

viruses such as HIV.560,561 Solvent–detergent treatment of plasma results in decreased α1-antitrypsin activity and total inactivation of α2-antiplasmin; antithrombin activity is unchanged.562 Subsequent work from another laboratory563 demonstrated that Triton X-100 detergent was responsible for the inactivation of α2-antiplasmin. While the removal of aprotinin presented problems to the management of excess ibrinolysis,564 EACA appears to be useful but still presents problems.565 The use of EACA to prevent unwanted ibrinolysis in hemophilia566–568 stems from the observation that a trypsin inhibitor in peanuts was useful in hemophilia.567 Presently, there appears to be more interest in plasminogen activator inhibitor-1 (PAI-1) as a therapeutic target.569–571 Treatment of congenital deiciency of α2-antiplasmin with whole plasma appears to be satisfactory572 with the caveat that α2-antiplasmin activity may be variable; EACA can be useful.527,568 The lack of the need for an α2-antiplasmin therapeutic product does not necessarily suggest the lack of therapeutic opportunities based on modulation of α2-antiplasmin. One study suggests that a decrease in α2-antiplasmin is necessary for plasmin activity to remove the ibrin matrix in a wound prior to the development of granulation tissue.573 Another more recent study574 suggests that a decrease in α2-antiplasmin upregulates the release of vascular endothelial growth factor (VEGF), inducing angiogenesis. Hatziapostolou and coworkers575 observed that EACA inhibited angiogenesis as well as endothelial cell proliferation, while α2-antiplasmin enhanced these effects. These researchers also showed that EACA and α2-antiplasmin had different effects on angiostatin production. Van Leer and coworkers576 showed that there is a ibrinolytic optimum for in vitro alveolar wound healing. The importance of α2-antiplasmin in the regulation of plasmin is also supported by the decrease of α2-antiplasmin in hepatocellular cancer development in hepatitis B-infected patients; a decrease in α2-antiplasmin resulted in increased degradation of the extracellular matrix.577 In support of this observation, Chan and coworkers578 observed downregulation of the SERPINF2 gene expression in hepatocellular carcinoma. These observations (and many others) emphasize the importance of the ibrinolytic system in wound healing in that plasmin, as with thrombin, is a regulatory multifunctional protease. As an example, Lei and coworkers579 suggest that plasmin is the major protease for processing platelet-derived growth factor in the development of proliferative vitreoretinopathy. α2-Antiplasmin, at high concentration, inhibits human kallikrein H5, which may be involved in tumorogeneis.580 It would appear that most of the actions of α2-antiplasmin are directed at the inhibition of plasmin. Compared to the other serpins, there is not much information on the structure of human α2-antiplasmin, nor has it been the subject of direct study. Most of the work has focused on its role as a component in ibrinolysis. Antiangiogenic and other cell-based activities have been ascribed to α2-antiplasmin, but it is dificult to differentiate a speciic effect of α2-antiplasmin from the inhibition of plasmin.

C1-INHIBITOR (C1-ESTERASE INHIBITOR) Complement is another blood system which, as with coagulation and ibrinolysis, involves a sequential series of protein interactions resulting in a inal physiological event. The binding and activation of the C1 complex, composed of C1q and a dimer

316

Biotechnology of Plasma Proteins

of two dimers, C1r and C1s, to antigen–antibody complexes via C1q result irst in the activation of C1r and subsequently of C1s.581 The binding and activation of the C1 component of complement depends on antibody–antigen density with IgG antibodies582–584 but not with IgM antibodies.585 C1r and C1s are both serine proteases, and C1s, after activation by C1r, forms the C4bC2a complex by the limited proteolysis of C4 and C2. The C4bC2a functions as the C3 convertase, which activates the remaining pathway.586 C1-esterase (C1s587) was identiied by Ratnoff and Lepow in 1957.588 An inhibitor to C1-esterase was partially puriied from serum by Pensky and colleagues589 in 1961. The reader is commended to several other sources for a discussion of complement and the regulation of complement.590,591 There are several earlier books592–596 that the author found quite useful and, inally, after more than 40 years of pretending, inally understood complement ixation. The book by Osler595 and the one by Kabat and Mayer593 were of particular value. C1-inhibitor (C1-esterase inhibitor; C1 INH) inhibits C1r and C1s in classical pathway of complement as well as plasma kallikrein and blood coagulation factor XIIa.597,598 The deiciency of C1-inhibitor results in the clinical problem of hereditary angioedema.598–600 Hereditary angioedema is a suficiently large clinical problem that a therapeutic concentrate has been developed by plasma fractionation.601,602 The incidence of hereditary angioedema is approximately 1 in 50,000,603 while acquired angioedema is a comparatively rare disorder with few reported cases.600,604 The reader is directed to an extremely useful chapter by Davis,598 which discusses the gene structure, expression, and molecular defects in hereditary angioedema. It should be noted that the use of a low-molecular-weight inhibitor of plasma kallikrein has proved useful in the treatment of hereditary angioedema.605,606 Andogen and androgen derivatives have been useful607,608 but are being supplanted by other therapeutic options.608 In addition to inhibiting the C1 activities, C1-inhibitor also inhibits plasma kallikrein and factor XIIa (activated Hageman factor).609,610 Given the spectrum of activities, C1 INH has a role in inlammation and other disorders.611,612 Dorresteijn and coworkers612 observed the anti-inlammatory action in the absence of complement activation. Thorgersen and coworkers613 observed anti-inlammatory activity of C1 INH in the absence of protease inhibitor activity; these studies were performed using E. coli challenge in vitro in porcine and human blood. C1-inhibitor is a bit unusual compared to other serpins in that it contains a large amount of carbohydrate (25%–30% by mass) in an approximate 100 amino acid extension amino-terminal from the serpin domain.598,614 The heavily glycosylated amino-terminal domain is suggested to be important in the role of CI INH in inlammatory disorders.614 Early studies on the puriication and characterization of C1 INH suggested a glycoprotein consisting of 478 amino acids with a single amino-terminal asparagine with a molecular mass of 104 kDa.615–617 Perkins and coworkers618 used neutron scattering to determine the structure of C1 INH. The results of the analysis were consistent with an Mr of 76 kDa and a length of 16–19 nm. The length is far greater than can be accommodated by the serpin domain, and these authors suggested that the length was due to the elongated structure of the N-terminal domain of 113 residues with most of the covalently linked carbohydrates. This suggests an asymmetric protein compared to α1-antitrypsin (length 7.0–7.8 nm). The asymmetry of the

Plasma Proteinase Inhibitors

317

protein explains the high molecular values obtained in earlier studies. Jack Kyte has an excellent discussion of factors inluencing the physical measurement of the protein structure, which is quite useful in understanding this problem when a protein is clearly not a sphere.619 It is not unusual for glycosylated proteins to show anomalous behavior on gel iltration (size exclusion chromatography) or SDS-PAGE.598,620–624 Analytical ultracentrifugation can present similar problems but can be managed.625 Ye626 has developed a useful approach for the determination of molecular mass by a combination of gel iltration and multiangle light scattering of the efluent fractions to measure protein and glycoprotein molecular weights with some accuracy. Mass spectrometry has proved useful for the determination of the molecular weights of glycoproteins627,628 and avoids the problems associated with the earlier techniques of gel iltration and SDS-PAGE. There is a substantial amount of information regarding the function of C1 INH and somewhat less on the chemistry of this protein. The mechanism of action for protease inhibition is a mechanism based on homology with other serpins, most notably α1-antitrypsin618,629–631 where there is inactivation via reaction within the RCL.544 The RCL of C1 INH is short compared to other serpins and elongation did not improve reactivity with plasma kallikrein.632 The reaction of C1 INH with C1s was shown by Harpel and Cooper633 to form an apparently covalent complex that could be dissociated by hydroxylamine,634 suggesting an ester linkage between the enzyme and the inhibitor. Lennick and coworkers635 studied the interaction of C1 INH with C1s and obtained a second-order rate constant of 6.0 × 104 M/s at 30°C in the absence of calcium ions; the reaction rate was modestly enhanced (35-fold) in the presence of heparin. Heparin was suggested to bind to both the protease and the inhibitor. Subsequent work from other researchers has shown interaction of heparin with the protease and with the serpin domain of C1 INH.636,637 Murray-Rust and coworkers636 showed that binding of heparin and dextran sulfate to C1s enhanced activity at low concentration and inhibited activity at high concentrations. Heparin has been demonstrated to have a modest effect on the reaction between human plasma kallikrein and C1 INH.638 C1 inhibitor is the major plasma protein inhibitor of factor XIIa,639,640 but the few studies on the effect of heparin on the reaction suggest little effect of heparin.641,642 It should be emphasized that when the action of C1 INH is examined in complex systems such as contact activation, the kinetic importance of individual reactions is dificult to determine with reasonable accuracy.643–646 Complicating this is the demonstration of the inactivation of factor XIa by C1 INH647 in a reaction that is stimulated by heparin. C1 inhibitor can inhibit a number of plasma proteases, with inhibition of plasma kallikrein being the one for which there is clear clinical support in hereditary angioedema; it is not known whether there are others. Type III hereditary angioedema occurs with normal functional and antigenic levels of C1 INH.648,649 Zuraw and Christiansen648 note that the primary mediator of swelling in hereditary angioedema is bradykinin derived from high-molecular-weight kininogen presumably by the action of plasma kallikrein generated from prekallikrein in contact activation.650–652 A gain-of-function mutation653 in factor XII has been discussed by Zuraw and Christiansen648 to explain the clinical indings with normal levels of C1 INH in type III hereditary angioedema.

318

Biotechnology of Plasma Proteins

C1-inhibitor is similar to other serpins in that C1 INH has important function(s) other than protease inhibition.654,655 It must be acknowledged that this statement is based on observations in complex systems such that inhibition of protease(s) cannot be excluded as part of the observed physiological effect. Cheng and coworkers656 observed that C1 INH prevented LPS-induced vascular permeability. Liu and coworkers657 showed that C1 INH prevented endotoxin shock by binding LPS; binding involved the glycosylated amino-terminal domain C1 INH. Cai and coworkers658 presented studies supporting a role for C1 inhibitor in the regulation of leukocyte adhesion. All of these functions are based on the amino-terminal 113 residue segment which contains most of the carbohydrates of C1 INH. Gesuete and coworkers659 evaluated the use of a recombinant form of C1 INH in a murine model for stroke. Recombinant C1 INH (intravenous) markedly reduced cerebral damage resulting from ischemia and was shown to bind to mannose-binding lectin (MBL); plasma-derived C1 INH had no effect. There are, as would be expected, differences between the plasma-derived protein and the recombinant protein. Mannose-binding lectin is part of the lectin pathway of complement activation and C1 INH is a potent inhibitor of the mannose-binding lectin-associated serine protease-2 (MASP-2), the enzyme responsible for cleaving the C2 and C4 components of complement.660 Osthoff and coworkers661 have reported that deiciency of MBL is associated with a favorable outcome in ischemic stroke patients and suggest that mannose-binding lectin is a target for patients with ischemic stroke where thrombolytic intervention is contraindicated. Earlier studies662 from another laboratory had suggested that deiciency of MBL results in better outcome with stroke in mouse models and human patients. Morrison and coworkers663 suggested that while the deiciency of MBL results in some protection in an ischemic mouse model, it is suggested that “redundant complement pathway activation” may negate the positive effects of MBL deiciency. However, the combination of binding MBL and subsequent blocking of the lectin pathway and the classical pathway might be useful. Tomasi and coworkers664 have used a rat model to demonstrate that C1 INH with a mutant prourokinase is effective in stroke and may be safer than tPA. Banz and Rieben665 have reviewed the role of complement and complement inhibitors in ischemia–reperfusion. C1 inhibitor can modulate the activity of a large number of enzymes and heparin may inluence the reaction with some proteases. The most exciting prospect is ischemic stroke where recombinant C1-INH may prove useful. The usefulness of rC1-INH in ischemic–reperfusion injury appears to depend on the carbohydrate component, and information on the structural differences between the plasmaderived protein and the recombinant protein is critical for explaining the differences observed between the plasma-derived material and the recombinant protein described by Gesuete and coworkers.659 There is some work on the characterization of the carbohydrate sidechains in the recombinant protein expressed in rabbits,666,667 but no direct comparison with the human protein. Wolff and coworkers668 have expressed C1 INH in baculovirus, and Bos and coworkers669 have expressed C1 INH in Pichia. Both proteins were glycosylated but the pattern of glycosylation was different from the plasma protein and differed between the two expression systems. The protein expressed in the baculovirus system was smaller (SDS-PAGE) than the

Plasma Proteinase Inhibitors

319

plasma-derived protein and possessed one-half of the speciic activity.668 The authors suggest that the lower molecular mass relects a decreased extent of glycosylation. The C1 INH expressed in Pichia pastoris had equivalent inhibitor activity as the plasma-derived protein toward C1s, plasma kallikrein, factor XIIa, and factor XIa; covalent complex formation was demonstrated for C1s and factor XIIa. Lamark and coworkers670 expressed an N-terminal truncated form of C1 INH (98–478), which had inhibitor activity against C1s and appeared to form a complex with C1s; integrity of disulide bonding is critical. The bulk of the data suggests that the effectiveness of C1 INH as a protease inhibitor requires the entire protein, including the amino-terminal extension, and the glycosylation is critical for biological function614,657,658,671 in inlammatory disorders and ischemic–reperfusion injury.659 Glycoengineering672,673 of the oligosaccharide chains will likely prove useful, considering the observed differences between the plasma-derived product and the recombinant product.659 The recombinant product is available674 and has been shown to be clinically effective in hereditary angioedema.675 However, there are other promising approaches to the management of hereditary angioedema, such as ecallantide.676 More work is required on the understanding of the role of the carbohydrate moiety in C1 INH biological function before the expanded use of recombinant C1 INH in ischemia–reperfusion injury.

PLASMINOGEN ACTIVATOR INHIBITOR-1 There are several plasminogen activator inhibitors,677 including plasminogen activator-2678 and plasminogen activator inhibitor-3, which is also known as protein c inhibitor.679,680 Plasminogen activator-2 is suggested as a primary inhibitor of the urokinase-type plasminogen activator, but there may be a broader function.681 However, the major factor regulating plasminogen activation is plasminogen activator inhibitor-1. Plasminogen activator inhibitor-1, which inhibits both tissue plasminogen activator and urokinase-type plasminogen activator, is a 45–50 kDa single-chain glycoprotein (15%–20% carbohydrate).682–691 The protein is present in plasma at a concentration of 20 μg/L.692,693 Plasminogen activator inhibitor-1 is an acute-phase protein and, as such, the concentration is quite variable694 and markedly increased in trauma patients.693 Plasminogen activator inhibitor-1 is also affected by circadian variation695–698 and there is speculation that this increase in PAI-1 activity might have a role in susceptibility to acute myocardial infarction. Plasminogen activator inhibitor-1 is associated in vivo with vitronectin, which can serve as a cofactor in the reaction with some proteases such as thrombin.699–703 Binding to vitronectin also maintains PAI-1 in an active conformation as opposed to the latent conformation.704–706 Plasminogen activator inhibitor-1 is a metastable protein707,708 somewhat similar to antithrombin and α1-antitrypsin, which also have latent conformations.709,710 Active PAI-1 binds to vitronectin, while latent PAI-1 does not bind to vitronectin. Plasminogen activator inhibitor-1 is unstable in solution and readily converts to the latent conformation.711 Metal ions and vitronectin have a major effect on the stability of PAI-1. The t1/2 for native PAI-1 in a 100 mM Tris buffer, pH 7.4, at 37°C is 1.14 h (measured by inhibition of tissue plasminogen activator), which is increased to 1.51 h in the presence of vitronectin. Metal ions had a differential effect, type 1 metal ions (i.e., magnesium, calcium) modestly increased the stability of PAI-1 in the presence

320

Biotechnology of Plasma Proteins

or absence of vitronectin, while type II metal ions (i.e., copper, cobalt) decreased stability in the absence of vitronectin, but there was a major increase in the stability in the presence of vitronectin (and somatomedin).712 Deiciency of PAI-1 is very rare and is associated with mild to moderate bleeding.713,714 Plasminogen activator inhibitor-1 is thought to control the activity of tissue plasminogen activator in the vascular space and urokinase-type plasminogen activator on cell surface and in the extravascular space.715 Plasminogen activator inhibitor-1 is synthesized and secreted from a variety of tissues, with the endothelium likely being the most important for the regulation of vascular ibrinolysis.716–719 It is possible to determine the number of tPA–PAI-1 complexes in blood720,721 and these are considered risk factors for myocardial infarction.722 The tPA–PAI-1 complex is elevated in type 2 diabetics as is PAI-1 while tPA activity is decreased.723 There is increasing evidence that an increase in PAI-1 predisposes to thrombosis.724–727 Thus, as with α2-antiplasmin, there is more concern about excessive PAI-1 activity and therapeutic approaches to modulate PAI-1 activity.728–731 However, the broad range of suggested PAI-1-mediated effects presents challenges for speciic function-targeted effects. Examination of the data for tPA and PAI-1 shows that while there is substantial complex formation, free tPA remains even with excess PAI-1. In the study by Sahli and coworkers,723 PAI-1 was markedly elevated in type 2 diabetics as was tPA– PAI-1 complex compared to nondiabetic controls. tPA activity was modestly reduced compared to nondiabetic controls (0.40 ± 0.36 IU/mL vs. 0.82 ± 0.47 IU/mL). It is, however, not clear that the assay measured tPA enzyme activity as such but rather the binding of tPA to immobilized antibody.732 Studies on a irst-generation recombinant ibrinolytic agent did have two approaches to the measurement of tPA which actually measured catalytic activity.733 Seifried and coworkers733 used a coupled assay with plasminogen and a CNBr fragment of ibrinogen, with the hydrolysis of H-D-Val-Leu-Lys-p-nitroanilide generating the signal.734 A second assay used ibrin plates prepared from plasminogen-rich bovine ibrinogen to measure tPA in acid-treated plasma samples. Samples were obtained from normal volunteers after a bolus infusion of tPA. Tissue plasminogen-activator and antigen demonstrated similar pharmacokinetics showing persistence of activity. These studies were discussed later by Tanswell.735 Plasminogen activator inhibitor-1 does appear to be depleted during thrombolytic therapy but does “rebound” following cessation of thrombolytic therapy.736–738 Paganelli and coworkers737 showed that the increase in PAI-1 was more pronounced following streptokinase than with tPA; there was no change in PAI-1 in patients subjected to percutaneous transluminal coronary angioplasty (PTCA). There is another issue regarding the reaction of PAI-1 with plasminogen activators, which appears to be unresolved. Lindahl and coworkers739 showed that the tPA– PAI-1 complex slowly dissociated at physiological pH and ionic strength. Takada and Takada740 suggested that the tPA–PAI-1 complex retained the ability to activate plasminogen and such activity was enhanced by the presence of ibrin. These studies were performed using zymography with SDS-PAGE gels, and it is suggested that SDS dissociated the complex with the release of active enzymes as these researchers subsequently observed for the α2-antiplasmin–plasmin complex.741 There are other studies on the characterization of the tPA–PAI-1 complex,742,743 but none that addresses the

Plasma Proteinase Inhibitors

321

stability of this complex in the circulation. Nilsson and coworkers744 evaluated the anticoagulant condition on the storage stability of the tPA–PAI-1 complex in plasma samples stored for at least 8 years at −70°C and observed lower values in acidiied citrated plasma (Stabilyte) than in citrated plasma, Ethylenediaminetetraacetic acid (EDTA) plasma, or serum. The highest values were obtained in EDTA plasma and serum and were attributed to the formation of complex on sample storage. HernestålBoman and coworkers745 showed that the concentration of tPA–PAI-1 complex decreased to a quantitatively negligible, but statistically signiicant extent on storage at −80°C for more than 11 years. The bulk of the information would suggest that the tPA–PAI-1 complex is stable on the formation and will not dissociate to form active tPA before the clearance of the complex from the circulation. Plasminogen activator inhibitor-1 is important in thrombosis and hemostasis but is likely more important for reasons other than vascular biology. The extravascular effects appear to be related to the inactivation of urokinase-type plasminogen activator as well as mechanisms still poorly understood but presumably unrelated to intravascular function; these effects will be discussed only briely. The expression of PAI-1 gene is highly responsive to changes in cell shape such as disruption of microtubule network and shear stress.746 PAI-1 gene expression is elevated in asthma, suggesting its potential as a biomarker and therapeutic target.747 Recent studies from this group748 have shown that cyclic adenosine monophosphate (AMP) induces PAI-1 expression in mast cells. Expression of PAI-1 in brain has an antiapoptotic effect on neurons and appears to be neuroprotective.749 Increased expression of PAI-1 gene is observed with oxidative challenge and results in tissue ibrosis.750 The proibrotic activity is suggested to be caused by the ability of PAI-1 to recruit macrophages into the interstitial space. The expression of PAI-1 gene is increased by inlammatory stimuli, such as TNF, IL-1, and IL6, while PAI-1 expression in vascular tissue is decreased by statins.751 Statins (simvastatin) reduces dexamethasone-induced expression of PAI-1 in human bone marrow adipocytes.752 Plasminogen activator inhibitor-1 is suggested to have value as a biomarker for a variety of diseases. Schmitt and coworkers found that both urokinase-type plasminogen activator and PAI-1 reached a high level of utility for a prognostic and predictive breastcancer biomarker in tumor tissue extracts.753–755 These researchers also developed techniques for the measurement of urokinase-type plasminogen activator and PAI-1 in formalin-ixed, parafin-embedded tissue sections.672 Malinowsky and coworkers756 reviewed the role of PAI-1 in tumor-associated processes such as extracellular matrix degradation, cell adhesion, and cell migration. PAI-1 functions in association with urokinase-type plasminogen activator and PAI-1 in tumor progression and metastasis and as such is an attractive target for therapeutic intervention.757 An increase in PAI-1 expression is associated with the progression of obesity-related cancers such as colorectal cancer.758 Plasminogen activator inhibitor-1 is synthesized and secreted by visceral adipose tissue and is considered an adipokine.759,760 Plasma levels of PAI-1 increase with the use of glucocorticoid during inlammation.761 Shen762 reported an increase in PAI-1 expression in endothelial cells by oxidized or glycated LDL. Cesari and coworkers have reviewed the various age-related disorders in which PAI-1 might be useful as a biomarker.763 The road to the development and validation of biomarkers is dificult,352 and it is not clear

322

Biotechnology of Plasma Proteins

whether PAI-1 will be useful. However, the ongoing study cited755 earlier might prove most useful in node-negative breast cancer. There might be some value to PAI-1 as a biomarker in cancer, but it is not clear whether it is more useful than currently available diagnostics; it is possible that continued work in the biomarker area will yield better understanding of various pathologies. There does not appear to be any value to the development of PAI-1 as a therapeutic product. As suggested earlier, there is more interest in inhibiting PAI-1 activity. However, given the diverse actions of PAI-1, speciicity will be a challenge.

PROTEIN C INHIBITOR (PLASMINOGEN ACTIVATOR INHIBITOR-3) The presence of an inhibitor of activated protein C (APC) in plasma was established by Canield and Kisiel in 1982.764 Protein C inhibitor (PCI; SERPINA5) is found in blood and other biological luids, including semen.765 Laurell and coworkers765 reported that the concentration of PCI in plasma is 5.3 mg/L, while the concentration in semen is 220 mg/L. The half-life of 125I-labeled PCI in a rabbit model was 23.4 h while the half-life of the APC PCI was 19.6 min.766 Laurell and coworkers766 did observe that the APC–PCI complex is somewhat unstable; similar instability was not observed for the complex of APC with α1-antitrypsin. PCI inhibits a number of proteases, including APC, thrombin, factor Xa, trypsin, chymotrypsin, and plasma kallikrein767,768 with a reaction mechanism767,769 shown by other serpins.544 The reaction of PCI with APC, thrombin, and urokinase is modestly accelerated by heparin.770 The identity of PCI with plasminogen activator inhibitor-3 was established by Stief and coworkers771 in 1987. A consideration of the literature suggests that there is no strong evidence for a critical role of PCI in hemostasis or thrombosis. Also, as noted by Cooper and Church,768 APC might not be the primary target for PCI in the circulation. There is a suggestion of PCI function in tissue regeneration and tumor growth.772–774 Given the high concentration of PCI in semen,765 there was great interest in a possible function for PCI in male reproductive physiology775–777 but little recent activity. Table 7.1 lists two other inhibitors which are not discussed in the text. Plasminogen activator inhibitor-2 is thought to function primarily inside the cell778 or in the extravascular space as an inhibitor of urokinase-type plasminogen activator779 and lacks deinition of function.681 Thrombin-activatable ibrinolysis inhibitor (carboxypeptidase U) does not inhibit proteases but rather inluences the ibrinolytic process by making ibrin less likely to promote plasminogen activation. There is more discussion of TAFI in Chapter 9, but it is noted that there is substantial interest in developing inhibitors of TAFI activity.780–782 It is possible to conclude that, with the exception of antithrombin, the various plasma proteinase inhibitors are intended to function in the extravascular space rather than in the intravascular space. It is my sense that only the development of therapeutic concentrates of antithrombin and α1-antitrypsin has merit. Antithrombin has potential as a parenteral drug for antithrombin deiciency, but the successful use of an antithrombin product in the larger acquired deiciency market will require the

Plasma Proteinase Inhibitors

323

early identiication of opportunity and aggressive use of the therapeutic product. Parenteral use of an α1-antitrypsin concentrate has potential for severe α1-antitrypsin deiciency with the caveat that liver ibrosis may be more critical than pulmonary dysfunction. The larger market for COPD will require the development of local delivery to the lower lung. It is possible that gene therapy such as the one developed by Neurologix for Parkinson’s disease783 would provide insight into device development for this purpose. There are observations that, with the exception of α2-macroglobulin, a substantial amount of every other inhibitor is in the extravascular space; however, both heparin cofactor II and plasminogen activator inhibitor-2 appear to function only in the intravascular space. Redundancy in speciicity of inhibition might imply our lack of understanding of function; alternatively, one could consider an extremely thoughtful article by Joe Gally and Gerry Edelman on degeneracy in biological systems.784 It is likely that the most effective approach to therapeutic intervention via plasma protein proteinase inhibitors other than antithrombin and α1-antitrypsin will involve modulation of gene expression.

REFERENCES 1. Travis, J. and Salvesen, G.S., Human plasma proteinase inhibitors, Annu. Rev. Biochem. 52, 655–709, 1983. 2. Boswell, D.R. and Carroll, R.W., The clinical signiicance of the serpins—A family of plasma enzyme inhibitors, Rec. Adv. Clin. Immunol. 4, 1–17, 1986. 3. Boswell, D.R. and Carroll, R.W., Genetic engineering and the serpins, BioEssays 8, 83–87, 1988. 4. Laskowski, M. and Kato, I., Protein inhibitors of proteinases, Ann. Rev. Biochem. 49, 593–626, 1980. 5. Travis, J. and Salvesen, G.S., Human plasma proteinase inhibitiors, Ann. Rev. Biochem. 52, 655–709, 1983. 6. Huang, X., Rezaie, A.R., Broze, G.J., Jr., and Olson, S.T., Heparin is a major activator of the anticoagulant serpin, protein Z-dependent protease inhibitor, J. Biol. Chem. 286, 8740–8751, 2011. 7. Hackeng, T.M. and Rosing, J., Protein S as cofactor for TFPI, Arterioscler. Thromb. Vasc. Biol. 29, 2015–2020, 2009. 8. Halili, M.A., Ruiz-Gómez, G., Le, G.T., et al., Complement component C2, inhibiting a latent serine protease in the classical pathway of complement activation, Biochemistry 48, 8466–8472, 2009. 9. Parker, B., Augeri, A., Capizzi, J., et al., Effect of air travel on exercise-induced coagulability and ibrinolytic activation in marathon runners, Clin. J. Sport Med. 21, 126–130, 2011. 10. Laterre, P.-F., Opal, S.M., and Abraham, E., A clinical evaluation committee assessment of recombinant human tissue factor pathway inhibitor (tifacogin) in patients with severe community-acquired pneumonia, Crit. Care 13, R36, 2009. 11. Van Den Boogaard, F.E., Brands, X., Schultz, M.J., et al., Recombinant human tissue factor pathway inhibitor exerts anticoagulant, anti-inlammatory and antimicrobial effects in murine pneumococcal pneumonia, J. Thromb. Haemost. 9, 122–132, 2011. 12. Emlen, W., Li, W., and Kirschink, M., Therapeutic complement inhibition: New developments, Semin. Thromb. Hemost. 36, 660–668, 2010. 13. Wasserman, R.L., Levy, R.J., Bewtra, A.K., et al., Prospective study of C1 esterase inhibitor in the treatment of successive acute abdominal and facial hereditary angiodema attacks, Ann. Allergy Asthma Immunol. 106, 62–68, 2011.

324

Biotechnology of Plasma Proteins

14. Phelps, R.A. and Putham, F.W., Chemical composition and molecular parameters of the puriied plasma proteins, in The Plasma Proteins, ed. F.W. Putnam, Vol. 1, Chapter 5, pp. 143–178, Academic Press, New York, 1960. 15. Putnam, F.W., Alpha, beta, gamma, omega—The roster of the plasma proteins, in The Plasma Proteins, 2nd edn., ed. F.W. Putnam, Vol. 1, Chapter 2, pp. 57–131, Academic Press, New York, 1975. 16. Chen, V., Lai, C., and Chan, T.K., Metabolism of antithrombin III in cirrhosis and carcinoma of the liver, Clinical Sci. 60, 681–688, 1981. 17. De Swart, C.A.M., Nijmeyer, B., Andersson, L.O., et al., Elimination of intravenously administered radiolabeled antithrombin III and heparin in humans, Thromb. Haemost. 52, 66–70, 1984. 18. Menache, D., O’Malley, J.P., Schorr, J.B., et al., Evaluation of the safety, recovery, half-life and clinical eficacy of antithrombin III (human) in patients with hereditary antithrombin III deiciency. Cooperative study group, Blood 75, 33–39, 1990. 19. Lu, W., Mant, T.G.K., Levy, J.H., and Bailey, J.M., Pharmacokinetics of a recombinant transgenic antithrombin in volunteers, Anesth. Analg. 90, 531–534, 2000. 20. Pal, N., Kettai, M.D., Lakshiminarasimhacher, A., and Avidan, M.S., Pharacology and clinical applications of human recombinant antithrombin, Expert Opin. Biol. Ther. 10, 1155–1168, 2010. 21. Edmunds, T., van Patten, S.M, Pollack, J., et al., Transgenically produced human antithrombin: Structural and functional comparison to human plasma-derived antithrombin, Blood 91, 4561–4571, 1998. 22. Zhou, Q., Kyasike, J., Echelard, Y., et al., Effect of genetic background on glycosylation heterogeneity in human antithrombin produced in mammary gland of transgenic goats, J. Biotechnol. 117, 57–72, 2005. 23. Schmidt, B., Wais, U., Pringsheim, W., et al., Decreased production or increased turnover of antithrombin III in severe acquired coagulopathy, Klin. Wochenschrift 59, 1349–1351, 1981. 24. Knot, E.A., ten Cate, J.W., Bruin, T., et al., Antithrombin III metabolism in two colitis patients with acquired antithrombin III deiciency, Gastroenterology 89, 421– 425, 1985. 25. Weiner, C.P., Herrig, J.E., Peizer, G.D., and Heilskof, J., Elimination of of antithrombin III concentrate in healthy and preeclamptic women with an acquired antithrombin III deiciency, Thromb. Res. 58, 395–401, 1990. 26. Vogel, C.N., Kingdon, H.S., and Lundblad, R.L., Correlation of in vivo and in vitro inhibition of thrombin by plasma inhibitors, J. Lab. Clin. Med. 93, 661–673, 1979. 27. Shifman, M.A. and Pizzo, S.V., The in vivo metabolism of antithrombin III and antithrombin III complexes, J. Biol. Chem. 257, 3243–3248, 1982. 28. Visich, J.E., Byrnes-Blake, K.A., Lewis, K.B., et al., Bioavailability and relative tissue distribution of [125I]-recombinant human thrombin following intravenous or subcutaneous administration to non-human primates, J. Thromb. Haemost. 4, 1962–1968, 2006. 29. Lundblad, R.L., Bergstrom, J., De Vreker, R., et al., Measurement of active coagulation factors in Autoplex-T with colorimetric active site-speciic assay technology, Thromb. Haemost. 80, 811–815, 1998. 30. Takahashi, I., Kato, K., Sugiura, I., et al., Activated factor IXa-antithrombin complexes in human blood: Quantiication byan enzyme-linked differential antibody immunoassay and determination of the in vivo half-life, J. Lab. Clin. Med. 118, 317–324, 1991. 31. Seested, T., Appa, R.S., Christensen, E.I., et al., In vivo clearance of recombinant activated Factor VII (r FVIIa) and its complexes with plasma protease inhibitors in the liver, Thromb. Res. 127, 356–362, 2011.

Plasma Proteinase Inhibitors

325

32. Collen, D., Schetz, J., De Cock, E., et al., Metabolism of antithrombin III (heparin cofactor) in man: Effects of thrombosis and heparin administration, Europ. J. Clin. Invest. 7, 27–35, 1977. 33. Carlson, T.H., Simon, T.L., and Atencio, A.C., In vivo behavior of human radioiodinated antithrombin III: Distribution among three physiologic pools, Blood 66, 13–19, 1985. 34. Manco-Johnson, M.J., Antithrombin-III, in Anticoagulant: Physiologic, Pathologic, and Pharmacologic, ed. D. Green, Chapter 3, pp. 27–40, CRC Press, Boca Raton, FL, 1994. 35. Carlson, T.H., Atencio, A.C., and Simon, T.C., In vivo behavior of radioiodinated rabbit antithrombin III. Demonstration of a noncirculatory vascular compartment, J. Clin. Invest. 74, 191–199, 1984. 36. Chou, F.P., Xu, H., Lee, M.S., et al., Matriptase is inhibited by extravascular antithrombin in epithelial cells but not in most carcinoma cells, Am. J. Physiol. Cell Physiol. 301, C1093–C1103, 2011. 37. Maclean, P.S. and Tait, C., Heriditary and acquired antithrombin deiciency. Epidemiology, pathogenesis and treatment options, Drugs 67, 1429–1440, 2007. 38. Luxembourg, B., Delev, D., Gelsen, C., et al., Molecular basis of antithrombin deiciency, Thromb. Haemost. 105, 635–646, 2011. 39. Picard, V., Ersdal-Badju, E., and Bock, S.C., Partial glycosylation of antithrombin III asparagine-135 is caused by the serine in the third position of its N-glycosylation consensus sequence and is responsible for production of the β-antithrombin III isoform with enhanced heparin afinity, Biochemistry 34, 8433–8440, 1995. 40. Carlson, T.H. and Atencio, A.C., Isolation and partial characterization of two distinct types of antithrombin III from rabbit, Thromb. Res. 27, 23–34, 1982. 41. Peterson, C.B. and Blackburn, M.N., Isolation and characterization of an antithrombin III variant with reduced carbohydrate content and enhanced heparin binding, J. Biol. Chem. 260, 610–615, 1985. 42. Turk, B., Brieditis, I., Bock, S.C., et al., The oligosaccharide side chain on Asn-135 of α-antithrombin, absent in β-antithrombin, decreases the heparin afinity the inhibitor by affecting the heparin-induced conformational change, Biochemistry 36, 6682–6691, 1997. 43. McCoy, A.J., Pel, X.Y., Skinner, R., et al., Structure of β-antithrombin and the effect of glycosylation on antithrombin’s heparin afinity and activity, J. Mol. Biol. 325, 823–833, 2003. 44. Grifith, M.J. and Lundblad, R.L., Dissociation of antithrombin III–thrombin complex. Formation of active and inactive antithrombin III, Biochemistry 20, 105–110, 1981. 45. Fish, W., Orre, K., and Björk, I., The production of an inactive form of antithrombin through limited proteolysis by thrombin, FEBS Lett. 98, 102–106, 1979. 46. Mourey, L., Shamana, J.P., Delarue, M., et al., Antithrombin III: Structural and functional aspects, Biochimie 72, 599–608, 1990. 47. O’Reilly, M.S., Pirie-Shepherd, S., Lane, W.S., and Folkman, J., Antiangiogenic activity of the cleaved conformation of the serpin antithrombin, Science 285, 1926– 1928, 1999. 48. Busby, T.F., Atha, D.H., and Ingham, K.C., Thermal denaturation of antithrombin III. Stabilization of heparin and lyotropic anions, J. Biol. Chem. 256, 12140–12147, 1981. 49. Wardell, M.P., Chang, W.-S.W., Bruce, D., et al., Preparative induction and characterization of L-antithrombin: A structural homologue of latent plasminogen activator inhibitor-1, Biochemistry 36, 13133–13142, 1997. 50. Larsson, H., Sjöblom, T., Dixelius, J., et al., Antiangiogenic activity effects of latent thrombin through perturbed cell–matrix interactions and apoptosis of endothelial cells, Cancer Res. 60, 6723–6729, 2000.

326

Biotechnology of Plasma Proteins

51. Larsson, H., Akerud, P., Nordling, K., et al., A novel anti-angiogenic form of antithrombin with retained proteinase binding ability and heparin afinity, J. Biol. Chem. 276, 11996–12002, 2001. 52. Karlsson, G. and Winge, S., Separation of native and latent forms of human antithrombin by hydrophobic interaction high-performance liquid chromatography, Protein Express. Purif. 21, 149–155, 2001. 53. Karlsson, G. and Winge, S., Separation of latent, prelatent and native forms of human antithrombin by heparin afinity high-performance liquid chromatography, Protein Express. Purif. 33, 339–345, 2004. 54. Chang, W.-S.W. and Harper, P.C., Commercial antithrombin concentrate contains L-forms of antithrombin, Thromb. Haemost. 77, 323–328, 1997. 55. Mochizuki, S., Miyara, K., and Kondo, M., Puriication and characterization of recombinant human antithrombin containing the prelatent forms in Chinese hamster ovary cells, Protein Express. Purif. 41, 323–341, 2005. 56. Mushunje, A., Evans, G., Brennan, S.O., et al., Latent antithrombin and its detection, formation and turnover in the circulation, J. Thromb. Haemost. 2, 2170–2177, 2004. 57. Kjellberg, M., Ikonomou, T., and Stenlo, J., The cleaved and latent forms of antithrombin are normal consituents of blood plasma: A quantitative method to measure cleaved antithrombin, J. Thromb. Haemost. 4, 168–176, 2005. 58. Carrell, R.W., Huntington, J.A., Mushunje, A., and Zhou, A., The conformational basis of thrombosis, Thromb. Haemost. 86, 14–22, 2001. 59. Corral, J., Rivers, J., Guerrera, J.A., et al., Latent and polymeric antithrombin: Clearance and potential thrombosis risk, Exp. Biol. Med. 232, 219–226, 2007. 60. Verstraete, M. and Vermylen, C., Antithrombin activity: Its interference with the estimation of antithromin II activity in certain thrombin inhibitor determinations, Thromb. Diath. Haemorrh. 3, 640–653, 1959. 61. Becker, D.L., Fredenburgh, J.C., Stafford, A.R., and Weitz, J.I., Molecular basis for the resistance of ibrin-based thrombin to inactivation by heparin/serpin complexes, in Chemistry and Biology of Serpins, eds. F.C. Church, D.D. Cunningham, D. Ginsburg, M. Hoffman, S.R. Stone, and D.M. Tollefson, Plenum Press, New York, 1996. 62. Mosesson, M.W., Update on antithrombin I (ibrin), Thromb. Haemost. 98, 105–108, 2007. 63. Briginshaw, G.F. and Shanberge, J.N., Identiication of distinct heparin cofactors in human plasma. Separation and partial puriication, Arch. Biochem. Biophys. 151, 683– 690, 1974. 64. Pratt, C., Whinna, H.C., Meade, J.B., et al., Physicochemical aspects of heparin cofactor II, Ann. N. Y. Acad. Sci. 556, 104–115, 1989. 65. Thaler, E. and Schmer, G., A simple two-step isolation procedure of human and bovine antithrombin II/III (heparin cofactor): A comparison of two methods, Br. J. Haematol. 31, 233–243, 1975. 66. Seegers, W.H., Antithrombin III: A backward glance o’er travel’d roads, Adv. Exp. Med. Biol. 52, 195–215, 1975. 67. Watanabe, K., Chao, F.C., and Tullis, J.L., Antithrombin activity of intact human platelets, Thromb. Diath. Haemorrh. 34, 115–126, 1975. 68. Lundblad, R.L. and White II, G.C., The interaction of thrombin with blood platelets, Platelets 16, 373–385, 2005. 69. Verstrate, M., Antithrombins, Thromb. Diath. Haemorrh. Suppl., 20, 385–389, 1966. 70. Hess, A.F., A test for antithrombin in the blood, J. Exp. Med. 21, 338–344, 1915. 71. Pickering, J.W. and Hewitt, J.A., Studies of the coagulation of the blood: Part II. Thrombin and antithrombins, Biochem. J. 16, 587–598, 1922. 72. Barratt, J.O., The anticoagulant action of antithrombin, Biochem. J. 23, 422–424, 1929.

Plasma Proteinase Inhibitors

327

73. Brinkhous, K.M., Smith, H.P., Jr., Warner, E.D., and Seegers, W.H., Heparin and blood clotting, Science 90, 539, 1939. 74. Lyttleton, J.W., The antithrombin activity of heparin, Biochem. J. 58, 15–23, 1954. 75. Ratnoff, O.D., Some recent advances in the study of hemostasis, Circ. Res. 35, 1–14, 1974. 76. Rosenberg, R.D. and Damus, P.S., The puriication and mechanism of action of human antithrombin-heparin cofactor, J. Biol. Chem. 248, 6490–6505, 1973. 77. Grifith, M.J., Heparin-catalyzed inhibitor/protease reactions: Kinetic evidence for a common mechanism of action of heparin, Proc. Natl. Acad. Sci. USA 80, 5460–5464, 1983. 78. Downing, M.R., Bloom, J.W., and Mann, K.G., Comparison of the inhibition of thrombin by three plasma protease inhibitors, Biochemistry 17, 2649–2653, 1978. 79. Coutinno, J.M., Ferro, J.M., Canhão, P., et al., Unfractionated or low-molecular weight heparin for the treatment of cerebral venous thrombosis, Stroke 41, 2575– 2780, 2010. 80. Cook, D., Meade, M., Guyatt, G., et al., Dalteparin versus unfractionated heparin in critical ill patients, New Engl. J. Med. 364, 1305–1314, 2011. 81. Olson, S.T., Richard, B., Izaguirre, G., et al., Molecular mechanism of antithrombinheparin regulation of blood clotting proteinases. A paradigm for understanding proteinase regulation by serpin family protein proteinase inhibitors, Biochimie 92, 1587–1596, 2010. 82. Fuse, S., Tomita, H., Yoshida, M., et al., High dose of intravenous antithrombin III without heparin in the treatment of disseminated intravascular coagulation and organ failure in four children, Am. J. Hematol. 53, 18–21, 1996. 83. Rodgers, G.M., Role of antithrombin concentrate in treatment of hereditary antithrombin deiciency. An update, Thromb. Haemost. 101, 806–812, 2009. 84. White, B. and Perry, D., Acquired antithrombin deiciency in sepsis, Brit. J. Haematol. 112, 26–31, 2001. 85. Wiederman, C.J., Hoffman, J.W., Juers, M., et al., High-dose antithrombin III in the treatment of severe sepsis in patients with a high risk of death: Eficacy and safety, Crit. Care Med. 34, 285–292, 2006. 86. Rau, J.C., Beaulieu, I., Huntington, J.A., and Church, F.C., Serpins in thrombosis, hemostasis and ibrinolysis, J. Thromb. Hemostas. 5(Suppl. 1), 102–115, 2007. 87. Butenas, S. and Mann, K.G., Blood coagulation, Biochemistry (Moscow) 67, 5–15, 2002. 88. Mann, K.G., Nesheim, M.E., Church, W.R., et al., Surface-dependent reactions of the vitamin K-dependent enzyme complexes, Blood 76, 1–16, 1990. 89. Haynes, L.M., Dubief, Y.C., Orfeo, T., and Mann, K.G., Dilutional control of prothrombin activation as physiologically relevant shear rates, Biophys. J. 100, 765–773, 2011. 90. Walker, F.J. and Esmon, C.T., The effects of phospholipid and factor Va on the inhibition of factor Xa by antithrombin III, Biochem. Biophys. Res. Commun. 90, 641–647, 1979. 91. Reeve, E.B., Steady state reactions between factors X, Xa, II, IIa, antithrombin III and alpha-2-macroglobulin in thrombosis, Thromb. Res. 18, 19–31, 1980. 92. Pettilä, V., Peritti, J., Pettilä, M., et al., Predictive value of antithrombin III and serum C-reactive protein concentration in critically ill patients with suspected sepsis, Crit. Care Med. 30, 271–275, 2002. 93. Wiedermann, C.J., Erfolge und Misserfolge gerinnungsaktiver Substenzen als SepsisTherapeutika, Arzneimittal-Forschung-Drug Res. 56, 792–794, 2006. 94. Inthorn, D., Hoffmann, J.N., Hartl, W.H., et al., Antithrombin III supplementation in severe sepsis: Beneicial effects on organ dysfunction, Shock 8, 329–334, 1997.

328

Biotechnology of Plasma Proteins

95. Haire, W.D., Ruby, E.I., Stephens, L.C., et al., A prospective randomized double-blind trial of antithrombin III concentrate in the treatment of multiple-organ dysfunction syndrome during hematopoietic stem cell transplantation, Biol. Blood Marrow Transplant. 4, 142–158, 1998. 96. Eid, A., Widermann, C.J., and Kinasewitz, G.T., Early administration of high-dose antithrombin in severe sepsis. Single center results from the KyberSept trials, Crit. Care Trauma 107, 1633–1638, 2008. 97. Haire, W.D., Multiple organ dysfunction syndrome in hematopoietic stem cell transplantation, Crit. Care Med. 30(Suppl.), S257–S262, 2002. 98. Heemskerk, J.W.M., Antithrombin extends its job, Thromb. Haemost. 92, 1171, 2004. 99. Makino, S. and Reed, C.E., Distribution and elimination of exogenous alpha 1-antitrypsin, J. Lab. Clin. Med. 73, 742–746, 1970. 100. Gadek, J.E., Klein, H.G., Holland, P.V., and Crystal, R.G., Replacement therapy of alpha 1-antitrypsin deiciency. Reversal of protease-antiprotease imbalance within the aveolar structures of PiZ subjects, J. Clin. Invest. 68, 1158–1165, 1981. 101. Laurell, C.-B. and Jeppsson, J.-O., Protease inhibitors in plasma, in The Plasma Proteins. Structure, Function, and Genetic Control, 2nd edn., ed. F.W. Putnam, Chapter 5, pp. 229–264, Academic Press, New York, 1975. 102. Ohlsson, K. and Olsson, I., Neutral proteases of human granulocytes. III. Interaction between human granulocyte elastase and plasma protease inhibitors, Scand. J. Clin. Lab. Invest. 34, 349–355, 1974. 103. Mackiewicz, A., Kushner, I., and Baumann, H.(eds.), Acute Phase Proteins: Molecular Biology, Biochemistry, and Clinical Applications, CRC Press, Boca Raton, FL, 1993. 104. Jahoor, F., Sivakumar, B., Del Rosario, M., and Frazer, E.M., Isolation of acute-phase proteins from plasma for determination of fractional synthesis rates by a stable isotope tracer technique, Anal. Biochem. 236, 95–100, 1996. 105. Turner, G.A. and Goodarzi, M.T., Oligosaccharide proiling of acute-phaes proteins: A possible strategy toward better markers in disease, Adv. Exp. Med. Biol. 435, 185–194, 1998. 106. Fredriksson, M.I., Figueredo, C.M., Gustafsson, A., et al., Effect of periodontitis and smoking on blood leukocytes and acute-phase proteins, J. Periodontol. 70, 1355–1360, 1999. 107. Buttenschoen, K., Buttenschoen, D.C., Berger, D., et al., Endotoxemia and acute-phase proteins in major abdominal surgery, Am. J. Surg. 181, 36–43, 2001. 108. Brunetti, N.D., Correale, M., Pellegrino, P.L., et al., Acute phase proteins in patients with acute coronary syndrome: Correlations with diagnosis, clinical features, and angiographic indings, Eur. J. Intern. Med. 18, 109–117, 2007. 109. Correale, M., Brunetti, N.D., De Gennaro, L., and Di Biase, M., Acute phase proteins in atherosclerosis (acute coronary syndrome), Cardiovasc. Hematol. Agents Med. Chem. 6, 272–277, 2008. 110. Ganrot, P.O., Variation of the concentrations of some plasma proteins in normal adults, in pregnant women and in newborns, Scand. J. Clin. Lab. Invest. 29(s124), 83–88, 1972. 111. Legge, M., Duff, G.B., Potter, H.C., and Hoetjes, M.M., Material serum alpha 1-antitrypsin concentrations in normotensive and hypertensive pregnancies, J. Clin. Pathol. 37, 867–869, 1984. 112. Lisowska-Myjak, B., Sygitowicz, G., Wolf, B., and Pachecka, J., Serum alpha-1antitrypsin concentration during normal and diabetic pregnancy, Eur. J. Obstet. Gynecol. Reprod. Biol. 99, 53–56, 2001. 113. Nielsen, C.H., Poulsen, H.K., Teisner, B., et al., Changes in blood levels of proteinase inhibitors, pregnancy zone protein, steroid carriers and complement factors induced by oral contraceptives, Eur. J. Obstet. Gynecol. Reprod. Biol. 51, 63–71, 1993.

Plasma Proteinase Inhibitors

329

114. Kurachi, K., Chandra, T., Degen, S.J., et al., Cloning and sequence of cDNA coding for α1-antitrypsin, Proc. Natl. Acad. Sci. USA 78, 6826–6830, 1981. 115. Long, G.L., Chandra, T., Woo, S.L., et al., Complete sequence of the cDNA for human α1-antitrypsin and the gene for the S variant, Biochemistry 23, 4828–4837, 1984. 116. Bollen, A., Loriau, R., Herzog, A., and Hérion, P., Expression of human α1-antitrypsin in Escherichia coli, FEBS Lett. 166, 67–70, 1984. 117. Courtney, M., Buchwalder, A., Tessier, L.H., et al., High-level production of biologically active human α1-antitrypsin in Escherichia coli, Proc. Natl. Acad. Sci. USA 81, 669–673, 1984. 118. Karnaukhova, E., Ophir, Y., and Golding, B., Recombinant human α1-proteinase inhibitor: Toward therapeutic use, Amino Acids 30, 317–332, 2006. 119. Tonelli, A.R. and Brantly, M.L., Augmentation therapy in α1-antitrypsin deiciency: Advances and controversies, Ther. Adv. Respir. Dis. 4, 289–312, 2010. 120. Cruz, P.E., Mueller, C., and Flotte, T.R., The promise of gene therapy for the treatment of α1-antitrypsin deiciency, Pharmacogenomics 8, 1191–1198, 2007. 121. Crawford, I.P., Puriication and properties of normal human α1-antitrypsin, Arch. Biochem. Biophys. 156, 215–222, 1973. 122. Talamo, R.C., Basic and clinical aspects of the alpha1-antitrypsin, Pediatrics 56, 91–99, 1975. 123. Musiani, P. and Tomasi, T.B., Jr., Isolation, chemical, and physical properties of α-1antitrypsin, Biochemistry 15, 798–804, 1976. 124. Travis, J. and Johnson, D., Human α1-proteinase inhibitor, Meth. Enzymol. 80, 754–765, 1981. 125. Boswell, D.R. and Bathurst, I.C., Molecular physiology and pathology of α1-antitrypsin, Biochem. Educ. 13, 98–104, 1985. 126. Grifiths, S.W., King, J., and Cooney, C.L., The reactivity and oxidation pathway of cysteine 232 in recombinant human α1-antitrypsin, J. Biol. Chem. 277, 25486–25492, 2002. 127. Laurell, C.-B., Complexes formed in vivo between immunoglobulin light chain κ, prealbumin, and/or α1-antitrypsin in myeloma sera, Immunochemistry 7, 461–465, 1970. 128. Tomasi, T.B. and Hauptman, S.P., The binding of α-1-antitrypsin in human IgA, J. Immunol. 112, 2274–2277, 1974. 129. Musiani, P., Lauriola, L., and Piantelli, M., Inhibitory activity of α-1-antitrypsin bound to human IgA, Clin. Chim. Acta 85, 61–66, 1978. 130. Vaerman, J.P., Hagiwara, K., Kobayashi, K., and Rits, M., Complexes of albumin and α1-antitrypsin with Fc-fragment of IgA monomer are disulide-bonded to penultimate C-terminal cysteine in the C α 3-domain, Immunol. Lett. 15, 67–72, 1987. 131. Kapel, N., Meillet, D., Iscaki, S., et al., Characterization of the main molecular forms of human fecal immunoglobulin A, Clin. Chim. Acta 195, 67–75, 1990. 132. Laurell, C.-B., Pierce, J., Persson, U., and Thulin, E., Puriication of α1-antitrypsin from plasma through thiol-disulide exchange, Eur. J. Biochem. 57, 107–113, 1975. 133. Laurell, C.-B., Dalhlqvist, I., and Persson, U., The use of thiol-disulide exchange chromatography for the automated isolation of α1-antitrypsin and other plasma proteins with reactive thiol groups, J. Chromatogr. 278, 53–61, 1983. 134. Pierce, J.A., Jeppson, J.-O., and Laurell, C.-B., α-1 Antitrypsin phenotypes determined by isoelectric focusing of the cysteine–antitrypsin mixed disulide in serum, Anal. Biochem. 74, 227–241, 1976. 135. Di Simplcio, P., Franconi, F., Frosali, S., and Di Giuseppe, D., Thiolation and nitrosation of cysteines in biological luids and cells, Amino Acids 25, 323–339, 2003. 136. Grimsley, G.R., Scholtz, J.M., and Pace, C.N., A summary of the measured pK values of the ionizable groups in folded proteins, Protein Sci. 18, 247–251, 2009. 137. Naor, M.M. and Jensen, J.H., Determinants of cysteine pKa values in creatine kinase and α1-antitrypsin, Proteins. Struct. Funct. Bioinform. 57, 799–803, 2004.

330

Biotechnology of Plasma Proteins

138. Miyamoto, Y., Akaike, T., Alam, M.S., et al., Novel functions of human α1-protease inhibitor after S-nitrosylation: Inhibition of cysteine protease and antibacterial activity, Biochem. Biophys. Res. Commun. 267, 918–923, 2000. 139. Miyamoto, Y., Akaike, T., and Maeda, H., S-Nitrosylated human α1-protease inhibitor, Biochim. Biophys. Acta 1477, 90–97, 2000. 140. Ikebe, N., Akaike, T., Miyamoto, Y., et al., Protective effect of S-nitrosylated α1-protease inhibitor on ischemia-reperfusion injury, J. Pharmacol. Exp. Ther. 295, 904–911, 2000. 141. Putnam, C., Porter, K.A., Peters, R.L., et al., Liver replacement for α1-antitrypsin deiciency, Surgery 81, 258–261, 1977. 142. Strange, C. and Jancliauskiene, S., Alpha-1-antitrypsin deiciency, in Molecular Basis of Pulmonary Disease. Insights from Rare Lung Disorders, eds. F.X. McCormack, R.J. Panos, and B.C. Trapnell, Chapter 9, pp. 209–224, Humana/Springer, New York, 2010. 143. Prachalias, A.A., Kalife, M., Francavilla, R., et al., Liver transplantation for alpha-1antitrypsin deiciency in children, Transplant. Int. 13, 207–210, 2000. 144. Brantly, M., Nukiwa, T., and Crystal, R.G., Molecular basis of alpha-1-antitrypsin deiciency, Am. J. Med. 84(Suppl. 6A), 13–33, 1988. 145. Hidvegi, T., Mukherjee, A., Ewing, M., et al., The role of autophagy in alpha-1antitrypsin deiciency, Meth. Enzymol. 499, 33–54, 2011. 146. Jones, E.A., Vergalla, J., Steer, C.J., et al., Metabolism of intact and disialylated α1-antitrypsin, Clin. Sci. Mol. Med. 55, 139–148, 1978. 147. Carrell, R.W., Jeppsson, J.-O., Laurell, C.-B., et al., Structure and variation of human α1-antitrypsin, Nature 298, 329–334, 1982. 148. Luisetti, M. and Seersholm, N., α1-Antitrypsin deiciency. I: Epidemiology of α1-antitrypsin deiciency, Thorax 59, 164–169, 2004. 149. Kelly, E., Geene, C.M., Carroll, T.P., et al., α1-antitrypsin deiciency, Respir. Med. 104, 763–772, 2010. 150. Salahuddin, P., Genetic variants of α1-antitrypsin, Curr. Protein Pept. Sci. 11, 101–117, 2010. 151. Fagerhol, M.K. and Cox, D.W., The Pi polymorphism genetic, biochemical, and clinical aspects of human α1-antitrypsin, Adv. Hum. Genet. 11, 1–62, 1981. 152. Sandhaus, R.A., α1-Antitrypsin deiciency: Whom to test, whom to treat?, Semin. Respir. Crit. Care Med. 31, 343–347, 2010. 153. Laurell, C.-B. and Eriksson, S., The electrophoretic α1-globulin pattern of serum in α1-antitrypsin deiciency, Scand. J. Clin. Lab. Invest. 15, 132–140, 1963. 154. Laurell, C.-B., Electrophoretic microheterogeneity of serum α1-antitrypsin, Scand. J. Lab. Clin. Invest. 17, 271–274, 1965. 155. Ganrot, P.O., Laurell, C.-B., and Eriksson, S., Obstructive lung disease and trypsin inhibitors in α1-antitrypsin deiciency, Scand. J. Clin. Lab. Invest. 19, 205–208, 1967. 156. Fargerhol, M.K. and Laurell, C.B., The polymorphism of “prealbumins” and α1-antitrypsin in human sera, Clin. Chim. Acta 16, 199–203, 1967. 157. Fagerhol, M.K., Quantitative studies on the inherited variants of serum α1-antitrypsin, Scand. J. Clin. Lab. Invest. 23, 97–103, 1969. 158. Virca, G.D. and Schnebli, H.P., The elastase/α1-1-proteinase inhibitor balance in the lung. A review, Schweiz. Med. Wochenschr. 114, 895–989, 1984. 159. Janoff, A., Elastases and emphysema. Current assessment of the protease-antiprotease hypothesis, Am. Rev. Respir. Dis. 132, 417–433, 1985. 160. Coakley, R.J., Taggart, C., O’Neill, S., and McElvaney, N.G., α1-Antitrypsin deiciency: Biological answers to clinical questions, Am. J. Med. Sci. 321, 33–41, 2001. 161. Pannell, R., Johnson, D., and Travis, J., Isolation and properties of human α-1-proteinase inhibitor, Biochemistry 13, 5439–5445, 1974.

Plasma Proteinase Inhibitors

331

162. Silverman, G.A., Bird, P.I., Carrell, R.W., et al., The serpins are an expanding superfamily of structurally similar but functionally diverse proteins, J. Biol. Chem. 276, 33293–33296, 2001. 163. Viegi, G., Pistelli, F., Sherrill, D.L., et al., Deinition, epidemiology and natural history of COPD, Eur. Respir. J. 30, 993–1013, 2007. 164. Johnson, D. and Travis, J., The oxidative inactivation of human α1-proteinase inhibitor. Further evidence for methionine at the reactive center, J. Biol. Chem. 254, 4022–4026, 1979. 165. Janoff, A., Carp, H., and Lee, D.K., Cigarette smoke inhalation decreases α1-antitrypsin activity in rat lung, Science 206, 1313–1314, 1979. 166. Carp, H. and Janoff, A., Potential mediator of inlammation. Phagocyte-derived oxidants suppress the elastase-inhibitory capacity of alpha1-proteinase inhibitor in vitro, J. Clin. Invest. 66, 987–995, 1980. 167. Carp, H., Miller, F., Hoidal, J.R., and Janoff, A., Potential mechanism of emphysema: α1-Proteinase inhibitor recovered from the lungs of cigarette smokers contains oxidized methionine and has decreased elastase inhibitory capacity, Proc. Natl. Acad. Sci. USA 79, 2041–2045, 1982. 168. Ashley, M.J., Corey, P., and Chen-Yeung, P., Smoking, dust exposure and serum alpha1-antitrypsin, Am. Rev. Respir. Dis. 121, 783–788, 1980. 169. Bridges, R.B., Kimmel, D.A., Wyatt, R.J., and Rehm, S.R., Serum antiproteases in smokers and nonsmokers. Relationships to smoking status and pulmonary function, Am. Rev. Respir. Dis. 132, 1162–1169, 1985. 170. Matsuki, H., Kasuga, H., Misawa, K., et al., Impact of smoking on the concentration and activity of alpha-1-antitrypsin in serum in relation to the urinary excretion of hydroxyproline, Tokai J. Exp. Clin. Med. 12, 19–26, 1987. 171. Pongpaew, P., Tungtrongchitr, R., Phonral, B., et al., Tobacco smoking in relation to the phenotype of alpha-1-antitrypsin and serum vitamin C concentration, J. Nutr. Environ. Med. 11, 167–173, 2001. 172. Ogushi, F., Hubbard, R.C., Vogelmeier, C., et al., Risk factors for emphysema. Cigarette smoking is associated with a reduction in the association rate constant of lung α1-antitrypsin for neutrophil elastase, J. Clin. Invest. 87, 1060–1065, 1991. 173. Gadek, J.E., Fells, G.A., Zimmerman, R.L., et al., Antielastases of human aveolar structures. Implications for the protease–antiprotease theory of emphysema, J. Clin. Invest. 68, 889–898, 1981. 174. Stockley, R.A., Neutrophils and protease/antiprotease imbalance, Am. J. Respir. Crit. Care Med. 160, 549–552, 1999. 175. Mittman, C., Lieberman, J., Marasso, F., and Miranda, A., Smoking and chronic obstructive lung disease in alpha1-antitrypsin deiciency, Chest 60, 214–221, 1971. 176. Lieberman, J., Heterozygous and homozygous alpha1-antittrypsin deiciency in patients with pulmonary emphysema, New Engl. J. Med. 281, 279–284, 1969. 177. Hill, A.T., Bayley, D.L., Campbell, E.J., et al., Airways inlammation in chronic bronchitis: The effects of smoking and α1-antitrypsin deiciency, Eur. Respir. J. 15, 886–890, 2000. 178. Summers, F.A., Morgan, P.E., Davies, M.J., and Hawkins, C.L., Identiication of plasma proteins that are susceptible to thiol oxidation by hypochlorous acid and N-chloramines, Chem. Res. Toxicol. 21, 1832–1840, 2008. 179. Aboussouan, L.S. and Stoller, J.K., α1-antitrypsin deiciency, in Chronic Obstructive Pulmonary Disease, eds. R.Schockley, S.Rennard, K.Rebe and B.Celli, Chapter 38, pp. 447–459, Blackwell, Malden, MA, 2007. 180. Ogushi, F., Fells, G.A., Hubbard, R.C., et al., Z-type alpha-1-antitrypsin is less competent than M1-type alpha 1-antitrypsin as an inhibitor of neutrophil elastase, J. Clin. Invest. 80, 1366–1374, 1987.

332

Biotechnology of Plasma Proteins

181. Ellett, M.L., Alpha 1-antitrypsin deiciency, Gastroenterol. Nurs. 14, 138–141, 1991. 182. Gadek, J.E., Klein, H.G., Holland, P.V., and Crystal, R.G., Replacement therapy of alpha 1-antitrypsin deiciency. Reversal of protease–antiprotease imbalance within the alveolar structure of PiZ subjects, J. Clin. Invest. 68, 1158–1165, 1981. 183. Coan, M.N. and Mitra, G., Stabilization of alpha-1-proteinase inhibitor by citrate, Vox Sang. 46, 142–148, 1984. 184. Coan, M.H., Brockway, W.J., Eguizabal, H., et al., Preparation and properties of alpha-1-proteinase inhibitor concentrate from human plasma, Vox. Sang. 48, 333–342, 1985. 185. Coan, M.H., Puriication of alpha-1-proteinase inhibitor. Preparation and properties of a therapeutic concentrate, Am. J. Med. 84(6A), 32–36, 1988. 186. Hein, M.H., Puriication of alpha-1-proteinase inhibitor (human), Eur. Respir. J. Suppl., 9, 16s–20s, 1990. 187. Coan, M.H., Dobkin, M.B., Brockway, W.J., and Mitra, G., Characterisation and virus safety of alpha-1-proteinase inhibitor, Eur. Respir. J. Suppl., 9, 35s–38s, 1990. 188. Stocks, J.M., Brantly, M.L., Wang-Smith, L., et al., Pharmacokinetic comparability of Prolastin®-C to Prolastin® in alpha1-antitrypsin deiciency: A randomized study, BMC Clin. Pharmacol. 10, 13, 2010. 189. Mordinkin, N.M. and Louie, S.G., Aralast®; an alpha-1-protease inhibitor for treatment of alpha-antitrypsin deiciency, Expert Opin. Pharmacother. 8, 2609–2614, 2007. 190. Louie, S.G., Sclar, D.A., and Gill, M.A., Aralast: A new alpha-1-protease inhibitor for treatment of alpha-antitrypsin deiciency, Ann. Pharmacother. 39, 1861–1869, 2005. 191. Kumpalume, P., Podmore, A., LePage, C., and Dalton, J., New process for the manufacture of alpha-1-antitrypsin, J. Chromatogr. A 1148, 31–37, 2007. 192. Lihme, A., Hansen, M.B., Anderson, I.V., and Burnouf, T., A novel core fractionation process of human plasma by expanded bed adsorption chromatography, Anal. Biochem. 399, 102–109, 2010. 193. Mattes, E., Matthiessen, P., Turecek, P.L., and Schwarz, H.P., Preparation and properties of an alpha-1-protease inhibitor concentrate with high speciic activity, Vox Sang. 81, 29–36, 2001. 194. Chen, S.X., Hammond, D.J., Lang, J.M., and Lebing, W.R., Puriication of α1 proteinase inhibitor from human plasma fraction IV-1 by ion exchange chromatography, Vox Sang. 74, 232–241, 1998. 195. Lomas, D.A., Elliott, P.R., Chang, W.-S.W., et al., Preparation and characterization of latent α1-antitrypsin, J. Biol. Chem. 270, 5282–5288, 1995. 196. Lomas, D.A., Elliott, P.R., and Carrell, R.W., Commercial plasma α1-antitrypsin (Prolastin®) contains a conformationally inactive, latent component, Eur. Respir. J. 10, 672–675, 1997. 197. Bottomley, J.P. and Tew, D.J., The citrate ion increases the conformational stability of α1-antitrypsin, Biochim. Biophys. Acta 534, 123–131, 1978. 198. Coan, M.H. and Mitra, G., Stabilization of alpha-1-proteinase inhibitor by citrate, Vox Sang. 46, 142–148, 1984. 199. Cohn, E.J., Strong, L.E., Hughes, W.L., et al., Preparation and properties of serum and plasma proteins. IV. A system for the separation into fractions of proteins and lipoproteins components of biological tissues and luids, J. Am. Chem. Soc. 68, 459– 470, 1946. 200. Ness, P.M. and Pennington, R.M., Plasma fractionation in the United States. A review for clinicians, JAMA 230, 247–250, 1974. 201. Hirota-Nakaoka, N. and Goto, Y., Alcohol-induced denaturation of β-lactoglobulin: A close correlation to the alcohol-induced α-helix formation of melittin, Bioorg. Med. Chem. 7, 67–73, 1999.

Plasma Proteinase Inhibitors

333

202. Tanaka, S., Oda, Y., Ataka, M., et al., Denaturation and aggregation of hen egg lysozyme in aqueous ethanol solution studied by dynamic light scattering, Biopolymers 59, 370– 379, 2001. 203. Miyawaki, O. and Tatsuno, M., Thermodynamic analysis of alcohol effect on thermal stability studied by dynamic light scattering, J. Biosci. Bioeng. 111, 198–203, 2011. 204. St-Amour, I., Laroche, A., Bazin, R., and Lemieux, R., Activation of cryptic IgG reactive with BAFF, amyloid beta peptide and GM-CSF during the industrial fractionation of human plasma into therapeutic immunoglobulins, Clin. Immunol. 133, 52–60, 2009. 205. Putnam, F.W., Protein denaturation, in The Proteins, eds. H. Neurath and K. Bailey, Vol. 1, Pt., B, Chapter 9, pp. 807–892, Academic Press, New York, 1953. 206. Lumry, R. and Eyring, H., Conformation changes of proteins, J. Phys. Chem. 58, 110– 120, 1954. 207. Hatley, R.H.M. and Franks, F., The effect of aqueous methanol cryosolvents on the heat- and cold-induced denaturation of lactate dehydrogenase, Eur. J. Biochem. 184, 237–240, 1989. 208. Jirgensons, B., Circular dichroism studies on the effects of ethanol on the conformation of α1-acid glycoprotein, α1-antitrypsin, deoxyribonuclease, pepsinogen, soybean trypsin inhibitor, and unfolded ribonuclease, Biochim. Biophys. Acta 534, 123–131, 2007. 209. Kumpulume, P., Podmore, A., LePage, C., and Dalton, J., New process for the manufacture of α-1 antitrypsin, J. Chromatogr. A 1148, 31–37, 2007. 210. Zimmerman, T.P., Yield improvement for manufacture of α1-proteinase inhibitor, Vox Sang. 91, 309–315, 2006. 211. Berger, D., Vischer, T.L., and Michel, A., Induction of proteolytic activity in serum by treatment with anionic detergents and organic solvents, Experentia 39, 1109–1111, 1983. 212. Matthiesen, H.P., Willemse, J., Weber, A., et al., Ethanol dependence of α1-antitrypsin C-terminal Lys truncation mediated by basic carboxypeptidase, Transfusion 48, 314– 320, 2008. 213. Desrochers, P.E. and Weiss, S.J., Proteolytic inactivation of alpha-1-proteinase inhibitor by a neutrophil metalloproteinase, J. Clin. Invest. 81, 1646–1650, 1988. 214. Ottonello, L., Dapino, P., and Dallegri, F., Inactivation of alpha-1-proteinase inhibitor by neutrophil metalloproteinase. Critical role of the myeloperoxidase system and effects of the anti-inlammatory drug nimesulide, Respiration 60, 32–37, 1993. 215. Abbink, J.J., Kamp, A.M., Nuijens, J.H., et al., Proteolyic inactivation of alpha-1antitrypsina nd alpha-1-chymotrypsin by neutrophils in arthritic joints, Arthrit. Rheum. 36, 168–180, 1993. 216. Russi, E., Alpha-1-antitrypsin: Now available, but do we need it?, Swiss Med. Weekly 138, 191–196, 2008. 217. Horowitz, I.D. and Schulman, E.S., Alpha-1-Pi for emphysema due to alpha-1-antitrypsin deiciency, Am. Fam. Physician 40, 223–228, 1989. 218. Kueppers, F., The role of augmentation therapy in alpha-1-antitrypsin deiciency, Curr. Med. Res. Opin. 27, 579–588, 2010. 219. Gøtzsche, P.C. and Johansen, H.K., Intravenous alpha-1 antitrypsi augmentation therapy for treating patients with alpha-1-antitrypsin deiciency and lung disease, Cochrane Database Syst. Rev. (7), CD007851, 2010. 220. Tonelli, A.R. and Brantly, M.L., Augmentation therapy in alpha-1-antitrypsin deiciency: Advances and controversies, Ther. Adv. Respir. Dis. 4, 289–312, 2010. 221. Barnes, P.J., Novel approaches and targets for treatment of chronic obstructive pulmonary disease, Am. J. Respir. Crit. Care Med. 160, 572–579, 1999. 222. Viegi, G., Pistilli, F., Sherrill, D.L., et al., Deinition, epidemiology and natural history of COPD, Eur. Respir. J. 30, 993–1013, 2007.

334

Biotechnology of Plasma Proteins

223. Stockley, R., Rennard, S., Rebe, K., and Celli, B. (eds.), Chronic Obstructive Pulmonary Disease, Blackwell, Malden, MA, 2007. 224. Stockley, R.A., Emerging drugs for α1-antitrypsin deiciency, Expert Opin. Emerg. Drugs 15, 685–694, 2010. 225. Piitulainen, E., Bernspång, E., Björkman, S., and Berntorp, E., Tailored pharmacokinetic dosing allows self-administration and reduces the cost of IV augmentation therapy with human α1-antitrypsin, Eur. J. Clin. Pharmacol. 59, 151–156, 2003. 226. Zamora, N.P., Pia, R.V., Del Rio, P.G., et al., Intravenous human plasma-derived augmentation therapy in α1-antitrypsin deiciency: From pharmacokinetic analysis to individualizing therapy, Ann. Pharmacol. 42, 640–646, 2008. 227. Newman, S.P., Aerosols and delivery systems, in Chronic Obstructive Pulmonary Disease, eds. R. Stockey, S. Rennard, K. Rebe, and B. Celli, Chapter 41, pp. 483–489, Blackwell, Malden, MA, 2007. 228. Patton, J.S., Brain, J.D., Davies, L.A., et al., The particle has landed—Characterizing the fate of inhaled pharmaceuticals, J. Aerosol. Med. Pulm. Drug Deliv. 23(Suppl. 2), S71–S87, 2010. 229. Andrade, F., Videira, M., Ferreira, D., and Sarmento, B., Nanocarriers for pulmonary administration of peptides and therapeutic proteins, Nanomedicine (London) 6, 123– 141, 2011. 230. Hubbard, R.C., Brantly, M.L., Sellers, S.E., et al., Anti-neutrophil-elastase defenses of the lower respiratory tract in α1-antitrypsin deiciency directly augmented with an aerosol of α1-antitrypsin, Ann. Intern. Med. 111, 206–212, 1989. 231. Kropp, J., Wencker, M., Hotze, A., et al., Inhalation of [123I] α1-protease inhibitor: Toward a new therapeutic concept of α1-protease inhibitor deiciency?, J. Nucl. Med. 42, 744–751, 2001. 232. Brand, P., Schulte, M., Wencker, M., et al., Lung deposition of inhalbed α1-proteinase inhibitor in cystic ibrosis and α1-antitrypsin deiciency, Eur. Respir. J. 34, 354–360, 2009. 233. Geller, D.E. and Kesser, K.C., The I-neb adaptive aerosol delivery system enhances delivery of α1-antitrypsin with controlled inhalation, J. Aerosol. Med. Pulmonary Drug Deliv. 23(Suppl. 1), S55–S59, 2010. 234. Flotte, T.R. and Mueller, C., Gene therapy for alpha-1-antitrypsin deiciency, Human Mol. Genetics 20, R87–R92, 2011. 235. Greene, C.M., Miller, S.D., Carroll, T., et al., Alpha-1-Antitrypsin deiciency: A conformational disease associated with lung and liver manifestations, J. Inherit. Metab. Dis. 31, 21–34, 2008. 236. Li, H., Lu, Y., Witek, R.P., et al., Ex vivo transduction and transplantation of bone marrow cells for liver gene delivery of α1-antitrypsin, Mol. Ther. 18, 1553–1558, 2010. 237. Chulay, J.D., Ye, G.J., Thomas, D.L., et al., Preclinical evaluation of a recombinant adeno-associated virus vector expressing human alpha-1-antitrypsin made using a recombinant herpes simplex virus production method, Hum. Gene Ther. 22, 155–165, 2011. 238. Rosenfeld, M.A., Siegfried, W., Yoshimura, K., et al., Adenovirus-mediated transfer of a recombinant α1-antitrypsin gene to the lung epithelium in vivo, Science 252, 431–434, 1991. 239. Zhang, D., Wu, M., Nelson, D.E., et al., Alpha-1-antitrypsin expression in the lung is increased by airway delivery of gene-transfected macrophages, Gene Ther. 10, 2148– 2152, 2003. 240. Ligun Wang, R., McLaughlin, T., Cossette, T., et al., Recombinant AAV serotype and capsid mutant comparison for pulmonary gene transfer of α1-antitrypsin using invasive and noninvasive delivery, Mol. Ther. 17, 81–87, 2009.

Plasma Proteinase Inhibitors

335

241. Halbert, C.L., Madtes, D.K., Vaughn, A.E., et al., Expression of human α1-antitrypsin in mice and dogs following AAV6 vector-mediated gene transfer to the lungs, Mol. Ther. 18, 1165–1172, 2010. 242. Breit, S.N., Luckhurst, E., and Penny, R., The effect of α1 antitrypsin on the proliferative response of peripheral blood lymphocytes, J. Immunol. 130, 681–686, 1983. 243. Breit, S.N., Wakeield, D., Robinson, J.P., et al., The role of alpha 1-antitrypsin deiciency in the pathogenesis of immune disorders, Clin. Immunol. Immunopathol. 35, 363–380, 1985. 244. Lindström, F.D., Skogh, T., and Lundström, I.M.C., α1 Antitrypsin deiciency in a patient with systemic vasculitis and primary Sjögren’s syndrome, Ann. Rheum. Dis. 61, 945–946, 2002. 245. Banda, M.J., Rice, A.G., Grifin, G.L., and Senior, R.M., α1-Proteinase inhibitor is a neutrophil chemoattractant after proteolytic inactivation by macrophage elastase, J. Biol. Chem. 263, 4481–4484, 1988. 246. Stockley, R.A., Shaw, J., Afford, S.C., et al., Effect of alpha-1-proteinase inhibitor on neutrophil chemotaxis, Am. J. Respir. Cell Mol. Biol. 2, 163–170, 1990. 247. Joslin, G., Grifin, G.L., August, A.M., et al., The serpin–enzyme complex (SEC) receptor mediates the neutrophil chemotactic effect of α-1antirypsin–elastase complexes and amyloid-β peptide, J. Clin. Invest. 90, 1150–1154, 1992. 248. Kataoka, H, Uchino, H., Iwamura, T., et al., Enhanced tumor growth and invasiveness in vivo by a carboxyl-terminal fragment of α-1-proteinase inhibitor generated by matrix metalloproteinases—A possible modulatory role in natural killer cytotoxicity, Am. J. Pathol. 154, 457–468, 1999. 249. Dabbagh, K., Laurent, G.L., Shock, A., et al., Alpha-1-antitrypsin stimulates ibroblast proliferation and procollagen production and activates classical MAP kinase signalling pathways, J. Cell. Physiol. 186, 73–81, 2001. 250. Churg, A., Dai, J., Zay, K., et al., Alpha-1-antitrypsin and a broad spectrum metalloproteinase inhibitor, RS 113456, have similar acute anti-inlammatory effects, Lab. Invest. 81, 1119–1131, 2001. 251. Janciauskiene, S., Zelvyte, I., Jansson, L., and Stevens, T., Divergent effects of α1-antitrypsin on neutrophil activation, in vitro, Biochem. Biophys. Res. Commun. 315, 288–296, 2004. 252. Aldonyte, R., Jansson, L., and Janciauskiene, S., Concentration-dependent effects of native and polymerized α1-antitrypsin on primary human monocytes, in vitro, BMC Cell Biol. 5, 11, 2004. 253. Bergin, D.A., Reeves, E.P., Melendy, P., et al., α-1-Antitrypsin regulates human neutrophil chemotaxis induced by soluble immune complexes or IL-8, J. Clin. Invest. 120, 4236–4250, 2010. 254. Subramaniyam, D., Steele, C., Köhnlein, I., et al., Effects of alpha-1-antitrypsin on endotoxin-induced lung inlammation in vivo, Inlamm. Res. 59, 571–578, 2010. 255. Shamanian, P., Schwartz, J.D., Pocock, B.J., et al., Activation of progelatinase A (MMP 2) by neutrophil elastase, cathepsin G, and proteinase-3: A role for inflammatory cells in tumor invasion and angiogenesis, J. Cell. Physiol. 189, 197–206, 2001. 256. Huang, H., Campbell, S.C., Nelius, T., et al., α1-Antitrypsin inhibits angiogenesis and tumor growth, Int. J. Cancer 112, 1042–1048, 2004. 257. Roeckel, N., Woerner, S.M., Kloor, M., et al., High frequency of LMAN1 abnormalities in colorectal tumors with microsatellite instability, Cancer Res. 69, 292–299, 2009. 258. Burghaus, B., Langer, C., Thedieck, S., and Nowak-Göttl, U., Elevated α1-antitrypsin is a risk factor for arterial ischemic stroke in childhood, Acta Haematol. 115, 186–191, 2006.

336

Biotechnology of Plasma Proteins

259. Engström, G., Lind, P., Hedblad, B., et al., Long-term effects of inlammation-sensitive plasma proteins and systolic blood pressure on incidence of stroke, Stroke 33, 2744– 2749, 2002. 260. Henriksen, P.A. and Salienave, J.-M., Human neutrophil elastase: Mediator and therapeutic target in atherosclerosis, Int. J. Biochem. Cell Biol. 40, 1095–1100, 2008. 261, Zsila, F., Inhibition of heat- and chemically-induced aggregation of various proteins reveals chaperone-like activity of the acute-phase component and serine protease inhibitor α1-antitrypsin, Biochem. Biophys. Res. Commun. 393, 242–247, 2010. 262. Congote, L.F. and Temmel, N., The C-terminal 26-residue peptide of serpin A1 stimulates proliferation of breast and liver cancer cells: Role of protein kinase C and CD47, Febs Lett. 576, 343–347, 2004. 263. Congote, L.F., Serpin A1 and CD91 as host instruments against HIV-1 infection: Are extracellular antiviral peptides acting as intracellular messengers?, Virus Res. 125, 119– 134, 2007. 264. Congote, L.F., Temmel, N., Sadvakassova, G., and Dobocan, M.C., Comparison of the effects of serpin A1, a recombinant serpin A1-IGF chimera and serpin A1 C-terminal peptide on wound healing, Peptides 29, 39–46, 2008. 265. Hatton, M.W.C., Hoogendoorn, H., Southward, S.M., et al., Comparative metabolism and distribution of rabbit heparin cofactor II and rabbit antithrombin in rabbits, Am. J. Physiol. 272, E824–E831, 1997. 266. Hatton, M.W.C., Ross, B., Southward, S.M.R., and Lucas, A.S., Metabolism and distribution of the virus-encoded serine proteinase inhibitor SERP-1 in healthy rabbits, Metabolism 49, 1449–1452, 2000. 267. Simmons, R.E. and Lane, D.A., Regulation of coagulation, in Thrombosis and Hemorrahage, 2nd edn., eds. J. Loscalzo and A.I. Schafer, Chapter 3, pp. 40–76, Williams & Wilkins, Baltimore, MA, 1998. 268. Tollefsen, D.M., Pestka, C.A., and Monafo, W.J., Activation of heparin cofactor II by dermatan sulfate, J. Biol. Chem. 258, 6713–6716, 1983. 269. McGuire, E.A. and Tollefsen, D.M., Activation of heparin cofactor II by ibroblasts and vascular smooth muscle cells, J. Biol. Chem. 262, 169–175, 1987. 270. Tollefsen, D.M., Majerus, D.W., and Blank, M.K., Heparin cofactor II. Puriication and properties of a heparin-dependent inhibitor of thrombin in human plasma, J. Biol. Chem. 257, 2162–2169, 1982. 271. Tollefsen, D.M., Heparin cofactor II deiciency, Arch. Pathol. Lab. Med. 126, 1394– 1400, 2002. 272. Liaw, P.C.Y., Becker, D.L., Stafford, A.R., et al., Molecular basis for the susceptibility of ibrin-bound thrombin to inactivation by heparin cofactor II in the presence of dermatan sulfate but not heparin, J. Biol. Chem. 276, 20959–20965, 2001. 273. Tollefsen, D.M., Vascular dermatan sulfate and heparin cofactor II, Prog. Mol. Biol. Transl. Sci. 93, 351–372, 2010. 274. Tovar, A.M.F., Teixeira, L.A.C., Marinho, A.C.O., et al., The dermatan sulfatedependent anticoagulant pathway is mostly preserved in aneurism and in severe atherosclerotic lesions while the heparan sulfate pathway is disrupted, Clin. Chim. Acta 412, 909–913, 2011. 275. Wang, Y. and Regg, H. An unexpected link between angiotensinogen and thrombin, FEBS Lett. 585, 2395–2399, 2011. 276. Hoffman, M., Pratt, C.W., Brown, R.L., and Church, F.C., Heparin cofactor II-proteinase reaction products exhibit neutrophil chemoattractant activity, Blood 73, 1682–1685, 1989. 277. Corbin, L.W., Church, F.C., and Hoffman, M., Production of chemotactic peptides by neutrophil degradation of heparin cofactor II, Thromb. Res. 57, 77–85, 1990.

Plasma Proteinase Inhibitors

337

278. Hoffman, M., Pratt, C.W., Corbin, L.W., and Church, F.C., Characteristics of the chemotactic activity of heparin cofactor II proteolysis products, J. Leukoc. Biol. 48, 156–162, 1990. 279. Church, F.C., Pratt, C.W., and Hoffman, M., Leukocyte chemoattractant peptides from the serpin heparin cofactor II, J. Biol. Chem. 266, 704–709, 1991. 280. Hoffman, M., Faulkner, K.A., Iannone, M.A., and Church, F.C., The effects of heparin cofactor II-derived chemotaxis on neutrophil actin conformation and cyclic AMP levels, Biochim. Biophys. Acta 1095, 78–82, 1991. 281. Jones, J.M., Creeth, J.M., and Kekwick, R.A., Thiol reduction of human α2-macroglobulin. The subunit structure, Biochem. J. 127, 187–197, 1972. 282. Hamberg, U., Stelwagen, P., and Ervast, H.S., Human α2-macroglobulin, characterization and trypsin binding. Puriication methodsl, trypsin and plasmin complex formation, Eur. J. Biochem. 40, 439–451, 1973. 283. Heide, K., Haupt, H., and Schwick, H.G., Plasma protein fractionation, in The Plasma Proteins, 2nd edn., ed. F.W. Putnam, Chapter 8, pp. 545–597, Academic Press, New York, 1977. 284. Sottrup-Jensen, L., α-Macroglobulins: Structure, shape, and mechanism of proteinase complex formation, J. Biol. Chem. 264, 11539–11542, 1989. 285. Björk, I. and Fish, W.W., Evidence for similar conformational changes in α2-macroglobulin on reaction with primary amines or proteolytic enzymes, Biochem. J. 207, 347–356, 1982. 286. Barrett, A.J., α2-Macroglobulin, Meth. Enzymol. 80, 737–754, 1981. 287. Kyte, J., Structure in Protein Chemistry, 2nd edn., Chapter 12, Garland Science/Taylor & Francis, New York, 2007. 288. Klingel, R., Fassbender, C., Fassbender, T., et al., Rheopheresis: Rheologic, functional, and structural aspects, Ther. Apher. 4, 345–357, 2000. 289. Kirschkamp, T., Schmid-Schönbein, H., Weinberger, A., and Smeets, R., Effects of ibrinogen and α2-macroglobulin and their apheretic elimination on general blood rheology and rheological characteristics of red blood cell aggregates, Ther. Apher. Dial. 12, 360–367, 2008. 290. Delain, E., Pochon, F., Barray, M., and Van Leuven, F., Ultrastructure of α2-macroglobulins, Electron Microsc. Rev. 5, 231–281, 1992. 291. Borth, W., α2-Macroglobulin, a multifunctional binding protein with targeting characteristics, FASEB J. 6, 3345–3353, 1992. 292. Folk, J.E. and Chung, S.I., Molecular and catalytic properties of transglutaminases, Adv. Enzymol. Relat. Areas Mol. Biol. 38, 109–191, 1973. 293. Dodds, A.W. and Law, S.K., The pylogeny and evolution of the thioester bond-containing proteins C3, C4 and α2-macroglobulin, Immunol. Rev. 166, 15–26, 1998. 294. Reilly, B.D., Structural comparison of human C4A3 and C4B1 after proteolytic activation by C1s, Mol. Immunol. 43, 800–811, 2006. 295. Mitchell, D.A., Ilyas, R., Dodds, A.W., and Sim, R.B., Enzyme-independent, orientationselective conjugation of whole human complement C3 to surfaces, J. Immunol. Meth. 337, 49–54, 2008. 296. Feinman, R.D., The proteinase-bindning reaction of α2 M, Ann. N. Y. Acad. Sci. 737, 245–266, 1994. 297. Williams, S.E., Kounnas, M.Z., Argraves, K.M., et al., The α2-macroglobulin receptor/ low density lipoprotein-related proteins and the receptor-associated protein. An overview, Ann. N. Y. Acad. Sci. 737, 1–13, 1994. 298. Harpel, P.C. and Mosesson, M., Degradation of human ibrinogen by plasma α2-macroglobulin–enzyme complexes, J. Clin. Invest. 52, 2175–2184, 1973. 299. Laurent, P. and Bieth, J.G., Kinetics of the inhibition of free and elastin-bound human pancreatic elastase by α1-proteinase inhibitor and α2-macroglobulin, Biochim. Biophys. Acta 994, 285–288, 1989.

338

Biotechnology of Plasma Proteins

300. Scharfstein, J., Parasite cysteine proteinase interactions with α2-macroglobulin or kininogens: Differential pathways modulating inlammation and inate immunity in infection by pathogenic trypanosomatids, Immunobiology 211, 117–125, 2006. 301. Chu, C.T., Howard, G.C., Misra, U.K., and Pizzo, S.V., α2-Macroglobulin: A sensor for proteolysis, Ann. N. Y. Acad. Sci. 737, 291–307, 1994. 302. Borth, W., α2-Macroglobulin. A multifunctional binding and targeting protein with possible roles in immunity and autoimmunity, Ann. N. Y. Acad. Sci. 737, 267–272, 1994. 303. Chu, C.T. and Pizzo, S.V., α2-Macroglobulin, complement, and biological defense: Antigens, growth factors, microbial proteases, and receptor ligation, Lab. Invest. 71, 792–812, 1994. 304. Chang, J.L., Ganea, D., Dray, S., and Teodorescu, M., Blast transformation of B cells induced by an alpha-macroglobulin-associated lymphokine produced in crowded lymphoid cell cultures, J. Immunol. 130, 267–273, 1983. 305. Chang, J.L., Ganea, D., Dray, S., and Teodorescu, M., An α2-macroglobulin associated factor produced by T lymphocytes which provides polyclonal stimulation of B lymphocytes to maintain the turnover of their surface Ig, Immunology 44, 745–754, 1981. 306. James, K., Alpha2 macroglobulin and its possible importance in immune systems, Trends Biochem. Sci. 5, 43–47, 1980. 307. James, K., Interaction between cytokines and α2-macroglobulin, Immunol. Today 11, 163–166, 1990. 308. Kurdowska, A., Alden, A.M., Noble, J.M., et al., Involvement of α-2-macroglobulin receptor in clearance of interleukin-8-α-2-macroglobulin complexes by human alveolar macrophages, Cytokine 12, 1046–1052, 2000. 309. James, K., Milne, I., Cunningham, A., and Elliott, S.-F., The effect of α2 macroglobulin in commercial cytokine assays, J. Immunol. Meth. 168, 33–37, 1994. 310. Cunningham, A.J., Elliott, S.-F., Black, J.R., and James, K., A simple method for isolating α2-macroglobulin–cytokine complexes, J. Immunol. Meth. 169, 287–292, 1994. 311. Burgess, E.F., Ham, A.J., Tabb, D.L., et al., Prostate cancer serum biomarker discovery through proteomic analysis of α2-macroglobulin protein complexes, Proteomics Clin. Appl. 2, 1223, 2008. 312. French, K., Yerbury, J.J., and Wilson, M.R., Protease activation of α2-macroglobulin modulates a chaperone-like action with broad speciicity, Biochemistry 47, 1176–1185, 2008. 313. Webb, D.J., Weaver, A.M., Atlkins-Brady, R.L., and Gonias, S.L., Proteinases are isoform-speciic regulators of the binding of transforming growth factor beta to α2-macroglobulin, Biochem. J. 320, 551–555, 1996. 314. Krimbou, L., Tremblay, M., Davignon, J., and Cohn, J.S., Association of apolipoprotein E with α2-macroglobulin in human plasma, J. Lipid Res. 39, 2373–2386, 1998. 315. Gavish, H., Bab, I., Tartakovsky, A., et al., Human α2-macroglobulin is an osteogenic growth peptide-binding protein, Biochemistry 36, 14883–14888, 1997. 316. Cianciolo, G.J., Enghild, J.J., and Pizzo, S.V., Covalent complexes of antigen and α2-macroglobulin: Evidence for dramatically-increased immunogenicity, Vaccine 20, 554–562, 2001. 317. Bowers, E.V., Horvath, J.J., Bond, J.E., et al., Antigen delivery by α2-macroglobulin enhances the cytotoxic T lymphocyte response, J. Leukoc. Biol. 86, 1259–1268, 2009. 318. Jochheck-Clark, A.R., Bowers, E.V., Totonchy, M.B., et al., Re-examination of CD91 (Glycoprotein 96) surface binding, uptake, and peptide-cross presentation, J. Immunol. 185, 6819–6830, 2010. 319. Binder, R.J., Karimeddini, D., and Srivastava, P.K., Adjuvanticity of α2-macroglobulin, an independent ligand for the heat shock protein receptor CD91, J. Immunol. 166, 4968– 4972, 2001.

Plasma Proteinase Inhibitors

339

320. Binder, R.J., Kumar, S.K., and Srivastava, P.K., Naturally formed or artiicially reconstituted non-covalent α2-macroglobulin–peptide complexes elicit CD91-dependent cellular immunity, Cancer Immun. 2, 16, 2002. 321. Binder, R.J., Puriication of α2-macroglobulin and the construction of immunogenic α2-macroglobulin–peptide complexes for use as cancer vaccines, Methods 32, 29–31, 2004. 322. Baumgart, Y., Otto, A., Schäfer, A., et al., Characterization of novel monoclonal antibodies for prostate-speciic antigen (PSA) with potency to recognized PSA bound to α2-macroglobulin, Clin. Chem. 51, 84–92, 2005. 323. Obiezu, C.V., Michael, I.P., Levesque, M.A., and Diamandis, E.P., Human kalikrein 4: Enzymatic activity, inhibition, and degradation of extracellular matrix proteins, Biol. Chem. 387, 749–759, 2006. 324. Misra, U.K., Payne, S., and Pizzo, S.V., Ligation of prostate cancer cell surface GRP78 activates a proproliferative and antiapoptotic feed back loop. A role for secreted prostatespeciic antigen, J. Biol. Chem. 286, 1248–1259, 2011. 325. Sinnreich, O., Kratzsch, J., Reichenbach, A., et al., Plasma levels of transforming growth factor-1β and α2-macroglobulin before and after radical prostatecomy: Association to clinicopathological parameters, Prostate 61, 201–208, 2004. 326. Poynard, T., Lebray, P., Ingiliz, P., et al., Prevalence of liver ibrosis and risk factors in a general population using non-invasive biomarkers (Fibrotest), BMC Gastroenterol. 10, 40, 2010. 327. Calès, P., Halfon, P., Batisse, D., et al., Comparison of liver ibrosis blood tests developed for HCV with new speciic tests in HIV/HCV co-infection, J. Hepatol. 53, 238– 244, 2010. 328. Cheung, K.J., Tilleman, K., Deforce, D., et al., Usefulness of a novel serum proteomederived index FI-PRO (ibrosis protein) in the prediction of ibrosis in chronic hepatitis C, Eur. J. Gastroenterol. Hepatol. 23, 701–710, 2011. 329. Ho, A.S., Cheng, C.C., Lee, S.C., et al., Novel biomarkers predict liver ibrosis in hepatitis C patients: α2-Macroglobulin, vitamin D binding protein and apolipoprotein AI, J. BIomed. Sci. 17, 58, 2010. 330. Rajmanickam, C., Sakthivel, S., Babu, G.J., et al., Cardiac isoform of α2-macroglobulin, novel serum protein, may induce cardiac hypertrophy, Basic Res. Cardiol. 96, 23–33, 2001. 331. Rajan, S., Radhakrishnan, J., and Rajamanickam, C., Direct injection and expression in vivo of full-length cDNA of the cardiac isoform of α2-macroglobulin induces cardiac hypertrophy in the rat heart, Basic Res. Cardiol. 98, 39–49, 2003. 332. Rajmanickam, C. and Jeejabai, R., Evaluation of the cardiac isoform of α2-macroglobulin as a factor inducing cardiac hypertrophy, Meth. Mol. Med. 112, 261–275, 2005. 333. Annapoorani, P., Dhandapany, P.S., Sadayappan, S., et al., Cardiac isoform of α2 macroglobulin—A new biomarker for myocardial infarcted diabetic patients, Atherosclerosis 186, 173–176, 2006. 334. Subbiah, R., Chengar, V. Clifton, J.D., et al., Cardiac isoform of α2-macroglobulin and its reliability as a cardiac marker in HIV patients, Heart Lung Circ. 19, 93–95, 2010. 335. Bai, Y., Sivori, D., Woo, S.B., et al., During glaucoma α2-macroglobulin accumulates in aqueous humor, and binds to nerve growth factor neutralizing neuroprotection, Invest. Ophthalmol. Vis. Sci. 52, 5260–5265, 2011. 336. Oh, J.H., Craft, J.M., Townsend, R., et al., A bioinformatics approach for biomarker identiication in radiation-induced lung inlammation from limited proteomics data, J. Proteome Res. 10, 1406–1415, 2011. 337. Kovacs, D.M., α2-Macroglobulin in late-onset Alzheimer’s disease, Exp. Gerontol. 35, 473–479, 2000.

340

Biotechnology of Plasma Proteins

338. McGeer, P.L. and McGeer, E.G., Polymorphisms in inlammatory genes and the risk of Alzheimer disease, Arch. Neurol. 58, 1790–1792, 2001. 339. McGeer, P.L. and McGeer, E.G., Inlammation, autotoxicity and Alzheimer disease, Neurobiol. Aging 22, 799–809, 2001. 340. Mocchegiani, E. and Malavolta, M., Zinc dyshomeostasis, ageing and neurogeneration: Implicatios of A2M and inlammatory gene polymorphisms, J. Alzheimers Dis. 12, 101– 109, 2007. 341. Song, R., Poljak, A., Smythe, G.A., and Sachdev, P., Plasma biomarkers for mild cognitive impairment and Alzheimer’s disease, Brain Res. Rev. 61, 69–80, 2009. 342. Bauer, J., Strauss, S., Schreiter-Gasser, U., et al., Interleukin-6 and α2-macroglobulin indicate an acute-phase state in Alzheimer’s disease cortices, FEBS Lett. 285, 111–114, 1991. 343. Giometto, B., Argentiero, V., Sanson, F., et al., Acute-phase proteins in Alzheimer’s disease, Eur. Neurol. 28, 30–33, 1988. 344. Gao, F., Bales, K.R., Dodel, R.C., et al., NF-κB mediates IL-1beta-induced synthesis/ release of α2-macroglobulin in a human glial cell line, Brain Res. Mol. Brain Res. 105, 108–114, 2002. 345. Zetterberg, H., Mattsson, N., Shaw, L.M., and Blennow, K., Biochemical markers in Alzheimer’s disease clinical trials, Biomark. Med. 4, 91–98, 2010. 346. Yerbury, J.J. and Wilson, M.R., Extracellular chaperones modulate the effects of Alzheimer’s patient cerebrospinal luid on Aβ(1–42) toxicity and uptake, Cell. Stress Chaperon. 15, 115–121, 2010. 347. Thambisetty, M., Do extracellular chaperone proteins in plasma have potential as Alzheimer’s disease biomarkers?, Biomark. Med. 4, 831–834, 2010. 348. Ozawa, D., Hasegawa, K., Lee, Y.H., et al., Inhibition of β2-microglobulin amyloid ibril formation by α2-macroglobulin, J. Biol. Chem. 286, 9668–9676, 2011. 349. Ito, S., Ohtsuki, S., Kamiie, J., et al., Cerebral clearance of human amyloid-β peptide (1–40) across the blood brain barrier is reduced by self-aggregation and formation lf low-density lipoprotein receptor-related protein-1 ligand complexes, J. Neurochem. 103, 2482–2890, 2007. 350. Mackiewicz, A., Kushner, I., and Baumann, H. (eds.), Acute Phase Proteins Molecular Biology, Biochemistry, and Clinical Applications, CRC Press, Boca Raton, FL, 1993. 351. Steel, D.M. and Whitehead, A.S., The acute phase response, in Humeral Factors, ed. E. Sim, Chapter 1, pp. 1–29, IRL Press at Oxford University Press, Oxford, UK, 1993. 352. Lundblad, R.L., Development and Application of Biomarkers, CRC Press, Boca Raton, FL, 2010. 353. Furie, B. and Furie, B.C., Molecular basis of blood coagulation, in Hematology Basic Principles and Practice, 5th edn., eds. R. Hoffman, E.J. Benz, Jr., S.J. Shattil, B. Furie, L.E. Silberstein, P.McGlare, H.E. Heslop, and J. Anastasi, Chapter 118, pp. 1819–1836, 2009. 354. Butenas, S., Ofeo, T., and Mann, K.G., Tissue factor in coagulation: Which? where? when?, Arterioscler. Thromb. Vasc. Biol. 29, 1989–1996, 2009. 355. Rodman, N.F., Jr., Barrow, E.M., and Graham, J.B., Diagnosis and control of the hemophilioid states with the partial thromboplastin (PTT) test, Am. J. Clin. Pathol. 29, 525– 538, 1958. 356. White II, G.C., The partial thromboplastin time: Deining an era in coagulation, J. Thromb. Haemost. 1, 2267–2270, 2003. 357. Morin, R.J. and Willoughby, D., Comparison of several activated partial thromboplastin time methods, Am. J. Clin. Pathol. 64, 241–247, 1975. 358. Christensen, R.L. and Triplett, D.A., Factor assay (VIII and IX) results in the College of American Pathologists Survey Program (1980–1982), Am. J. Clin. Pathol. 80(Suppl.) 633–642, 1983.

Plasma Proteinase Inhibitors

341

359. Lawrie, A.S., Kitchen, S., Purdy, G., et al., Assessment of ActinFS and ActinFSC sensitivity to speciic clotting factor deiciencies, Clin. Lab. Hematol. 20, 179–186, 1998. 360. van Den Besselaar, A.H., Evatt, B.L., Brogan, D.R., and Triplett, D.A., Proiciency testing and standardization of prothrombin time: Effect of thromboplastin, instrumentation, and plasma, Am. J. Clin. Pathol. 82, 688–699, 1984. 361. Carter, C.J., Griswold, D.J., Louis, P.A., et al., A comparison of seven prothrombin time reagents—Development of an evaluation strategy, Mod. Pathol. 1, 284–287, 1988. 362. Riley, R.S., Rowe, D., and Fisher, L.M., Clinical utilization of the international normalized ratio (INR), J. Clin. Lab. Anal. 14, 101–114, 2000. 363. Dahlbäck, B. and Stenlo, J., Regulatory mechanisms in hemostasis: Natural anticoagulants, in Hematology Basic Principles and Practice, 5th edn., eds. R. Hoffman, E.J. Benz, Jr., S.J. Shattil, B. Furie, L.E. Silberstein, P. McGlare, H.E. Heslop, and J. Anastasi, Chapter 120, pp. 1843–1849, 2009. 364. Seested, T., Appa, R.S., Chrisstensen, E.J., et al., In vivo clearance and metabolism of recombinant activated factor VII (rFVIIa) and its complexes with plasma protease inhibitors in the liver, Thromb. Res. 127, 356–362, 2011. 365. Seligsohn, U., Kasper, C.K., Osterud, B., and Rapaport, S.I., Activated factor VII: Presence in factor IX concentrates and persistence in the circulation after infusion, Blood 53, 828–837, 1979. 366. Kasthuri, R.S., Glover, S.L., Boles, J., and Mackman, N., Tissue factor and tissue factor pathway inhibitor as key regulators of global hemostasis: Measurement of their levels in coagulation assays, Semin. Thromb. Hemost. 36, 764–771, 2010. 367. Bauer, K., Hypercoagulable states, in Hematology Basic Principles and Practice, 5th edn., eds. R. Hoffman, E.J. Benz, Jr., S.J. Shattil, B. Furie, L.E. Silberstein, P. McGlare, H.E. Heslop, and J. Anastasi, Chapter 134, pp. 2021–2041, 2009. 368. Zakai, N.A., Lutsey, P.L, Folsom, A.R., et al., Total tissue factor pathway inhibitor and venous thrombosis. The Longitudinal Investigation of Thromboembolism Etiology, Thromb. Haemost. 104, 207–212, 2010. 369. Bajaj, M., Rama, S., Wysolmerski, R., et al., Inhibition of the factor VII–tissue factor complex is reduced in patients with disseminated intravascular coagulation but not in patients with severe hepatocellular disease, J. Clin. Invest. 79, 1874–1878, 1987. 370. Maroney, S.A. and Mast, A.E., Expression of tissue factor pathway inhibitor by endothelial cells and platelets, Transfus. Apher. Sci. 38, 9–14, 2008. 371. Dhainaut, J.F., Marin, N., Mignon, A., and Vinsonneau, C., Hepatic response to sepsis: Interaction between coagulation and inlammatory processes, Crit. Care Med. 29(7 Suppl.), S42–S47, 2001. 372. Tang, H., Ivanciu, L., Popsecu, N., et al., Sepsis-induced coagulation in the baboon lung is associated with decreased tissue factor pathway inhibitor, Am. J. Pathol. 171, 1066–1077, 2007. 373. Levi, M. and van der Poll, T., Inlammation and coagulation, Crit. Care Med. 38(2 Suppl.), S26–S34, 2010. 374. Levi, M. and Marder, V.J., Coagulation abnormalities in sepsis, in Hemostasis and Thrombosis, 5th edn., eds. R.W. Colman, V.J. Marder, A.W. Clowes, J.N. George, and S.Z. Goldhaber, Chapter 110, pp. 1601–1611, Lippincott, Williams & Wilkins, Philadelphia, 2006. 375. Bützow, R., Huhtala, M.L., Bohn, H., et al., Puriication and characterization of placental protein 5, Biochem. Biophys. Res. Commun. 150, 483–490, 1988. 376. Warn-Cramer, B.J., Rao, L.V., Maki, S.L., and Rapoport, S.I., Modiication of extrinsic pathway inhibitor (EPI) and factor X that affect their ability to interact and to inhibit factor VIIa/tissue factor: Evidence for a two-step model of inhibition, Thromb. Haemost. 60, 453–456, 1988.

342

Biotechnology of Plasma Proteins

377. Broze, G.J., Jr., Warren, L.A., Girard, J.J., and Miletich, J.P., Isolation of the lipoproteinassociated coagulation inhibitor produced by HepG2 (human hepatoma) cells using bovine factor Xa afinity chromatography, Thromb. Res. 40, 253–259, 1987. 378. Sprecher, C.A., Kisiel, W., Matthews, S., and Foster, D.C., Molecular cloning, expression, and partial characterization of a second human tissue-factor-pathway inhibitor, Proc. Natl. Acad. Sci. USA 91, 3353–3357, 1994. 379. Jin, M., Udagawa, K., Miyagi, E., et al., Expression of serine proteinase inhibitor PP5/ TFPI-2/MSPI decreases the invasive potential of human choriocarcinoma cells in vitro and in vivo, Gynecol. Oncol. 83, 325–333, 2001. 380. Chand, H.S., Foster, D.C., and Kisiel, W., Structure, function and biology of tissue factor pathway inhibitor-2, Thromb. Haemost. 94, 1122–1130, 2005. 381. Nisbet, A.D., Brenner, R.D., Horne, C.H., and Bohn, H., Placental protein 5 (PP5) in pregnancy and malignant disease: The inluence of heparin binding, Clin. Chim. Acta 119, 21–29, 1982. 382. Siiteri, J.E., Koistinen, R., Salem, H.T., et al., Placental protein 5 is related to blood coagulation and ibrinolytic systems, Life Sci. 30, 1885–1891, 1982. 383. Salem, H.T., Seppälä, M., Ranta, T., et al., The effect of protamine on serum levels of placental protein 5 (PP5) in normal and abnormal pregnancy: A possible relation to coagulation abnormalities, Br. J. Obstet. Gynaecol. 88, 367–370, 1981. 384. Salem, H.T., Obiekwe, B.C., Al-Ani, A.T., et al., Molecular heterogeneity of placental protein 5 (PP5) in late pregnancy serum and plasma: Evidence for a heparin–PP5 polymer, Clin. Chim. Acta 107, 211–215, 1980. 385. Jones, G.R., Davey, M.W., Sinosich, M., and Grudzinskas, J.G., Speciic interaction between placental protein 5 and heparin, Clin. Chim. Acta 110, 65–70, 1981. 386. Bremner, R.D., Nisbet, A.D., Herriott, R., et al., Detection of placental protein ive (PP5) and pregnancy-speciic glycoprotein (SP1) in benign and malignant breast disease, Oncodev. Biol. Med. 2, 55–62, 1981. 387. Nisbet, A.D., Bremner, R.D., Horne, C.H., et al., Placental protein 5 in gestational trophoblastic disease: Localization and circulating levels, Am. J. Obstet. Gynecol. 144, 396–401, 1982. 388. Carson, S.D., Plasma high density lipoproteins inhibit the activation of coagulation factor X by factor VIIa and tissue factor, FEBS Lett. 132, 37–40, 1981. 389. Morrison, S.A. and Jesty, J., Tissue factor—Dependent activation of trititum-labeled factor IX and factor X in human plasma, Blood 63, 1338–1347, 1984. 390. Sanders, N.L., Bajaj, S.P., Zivelin, A., and Rapaport, S.I., Inhibition of tissue factor/ VIIa activity requires factor X and an additional plasma component, Blood 66, 204–212, 1985. 391. Rao, L.V. and Rapaport, S.I., Studies of a mechanism inhibiting the initiation of the extrinsic pathway of coagulation, Blood 69, 645–651, 1987. 392. Warn-Cramer, B.J., Rao, L.V.M., Maki, S., and Rapaport, S.I., Modiication of extrinsic pathway inhibitor (EPI) and factor X that affect their ability to interact and to inhibit factor VIIa/tissue factor: Evidence for a two-step model of inhibition, Thromb. Haemost. 60, 453–456, 1988. 393. Huang, Z.-F., Wun, T.-C., and Broze, G.J., Jr., Kinetics of factor X inhibition by tissue factor pathway inhibitor, J. Biol. Chem. 268, 26950–26955, 1983. 394. Baugh, R.J., Broze, G.J., Jr., and Krishnaswamy, S., Regulation of extrinsic pathway factor Xa formation by tissue factor pathway inhibitor, J. Biol. Chem. 273, 4378–4386, 1998. 395. Broze, G.L., Jr., Warren, L.A., Girard, J.J., and Miletich, J.P., Isolation of the lipoprotein associated coagulation inhibitor produced by HepG2 (human hepatoma) cells using bovine factor Xa afinity chromatography, Thromb. Res. 48, 253–259, 1987.

Plasma Proteinase Inhibitors

343

396. Novotny, W.F., Girard, T.J., Miletich, J.P., and Broze, G.J., Jr., Puriication and characterization of the lipoprotein-associated coagulation inhibitor for human plasma, J. Biol. Chem. 264, 18832–18837, 1989. 397. Broze, G.L., Jr., Tissue factor pathway inhibitor and the revised hypothesis of blood coagulation, Trends Cardiovasc. Med. 2, 72–77, 1992. 398. Garantziotis, S., Hollingsworth, J.W., Ghanayam, R.B., et al., Inter-alpha-trypsin inhibitor attenuates complement activation and complement-induced lung injury, J. Immunol. 179, 4187–4192, 2007. 399. Opal, S.M., Lim, Y.P., Cristofaro, P., et al., Inter-α inhibitor proteins: A novel therapeutic strategy for experimental anthrax infection, Shock 35, 42–44, 2011. 400. Rao, C.N., Liu, Y.Y., Peavey, C.L., and Woodley, D.T., Novel extracellular matrixassociated serine proteinase inhibitors from human skin ibroblasts, Arch. Biochem. Biophys. 317, 311–314, 1995. 401. Fries, E. and Blom, A.M., Bikunin—Not just a plasma proteinase inhibitor, Int. J. Biochem. Cell Biol. 32, 125–137, 2000. 402. Kobayashi, H., Endogenous anti-inlammatory substances, inter-alpha-inhibitor and bikunin, Biol. Chem. 387, 1545–1549, 2006. 403. Delaria, K.A., Muller, D.K., Marlor, C.W., et al., Characterization of placental bikunin, a novel human serine protease inhibitor, J. Biol. Chem. 272, 12209–12214, 1997. 404. Wun, T.C., Kretzmer, K.K., Girard, T.J., et al., Cloning and characterization of a cDNA coding for the lipoprotein-associated coagulation inhibitor shows that it consists of three tandem Kunitz-type inhibitory domains, J. Biol. Chem. 263, 6001–6004, 1988. 405. Nakahara, Y., Miyata, T., Hamura, T., et al., Amino acid sequence and carbohydrate structure of a recombinant human tissue factor pathway inhibitor expressed in Chinese hamster ovary cells: One N-linked and two O-linked carbohydrate chains are located between Kunitz domains 2 and 3 and one N-linked carbohydrate chains is in Kunitz domain 2, Biochemistry 35, 6450–6459, 1996. 406. Girard, T.J., Warren, L.A., Novotny, W.F., et al., Identiication of the 1.4 kb and 4.0 kb messages for the lipoprotein associated coagulation inhibitor and expression of the encoded protein, Thromb. Res. 55, 37–50, 1989. 407. Abumiya, T., Enjyoji, K., Kokawa, T., et al., An anti-tissue factor pathway inhibitor (TFPI) monoclonal antibody recognized the third Kunitz domain (K3) of free-form TFPI but lipoprotein-associated forms in plasma, J. Biochem. 118, 178–182, 1995. 408. Piro, O. and Broze, G.J., Jr., Role for the Kunitz-3 domain of tissue factor pathway inhibitor-α in cell surface binding, Circulation 110, 3567–3572, 2004. 409. Lin, Y.F., Zhang, N., Guo, H.S., et al., Recombinant tissue factor pathway inhibitor induces apoptosis in cultured rat mesangial cells via its Kunitz-3 domain and C-terminal through inhibiting PI3-kinase/AKT pathway, Apoptosis 12, 2163–2173, 2007. 410. Warshawsky, I., Bu., G., Mast, A., et al., The carboxy terminus of tissue factor pathway inhibitor is required for interacting with hepatoma cells in vitro and in vivo, J. Clin. Invest. 95, 1777–1781, 1995. 411. Ndonwi, M., Tuley, E.A., and Broze, G.J., Jr., The Kunitz-3-domain of TFPI-α is required for protein S-dependent enhancement of factor Xa inhibition, Blood 116, 1344–1351, 2010. 412. Kunitz, M. and Northrop, J.H., Isolation from beef pancreas of crystalline trypsinogen, trypsin, a trypsin inhibitor, and an inhibitor–trypsin compound, J. Gen. Physiol. 19, 991–1007, 1936. 413. Kunitz, M., Crystalline soybean trypsin inhibitor, J. Gen. Physiol. 29, 149–154, 1946. 414. Johnstone, E.M., Chaney, M.O., Moore, R.E., et al., Alzheimer’s disease amyloid peptide is encoded by two exons and shows similarity to soybean trypsin inhibitor, Biochem. Biophys. Res. Commun. 163, 1248–1255, 1989.

344

Biotechnology of Plasma Proteins

415. Markland, W. and Ladner, R.C., Kallikrein-inhibiting “Kunitz domain” proteins and analogues thereof, U.S. Patent 5, 795, 865, 1998. 416. Hochstasser, K. and Wachter, E., Kunitz-type proteinase inhibitors derived by limited proteolysis of inter-α-trypsin inhibitor I. Determination of the antitryptic domain by solid-phase Edman degradation, Hoppe-Seyler’s Zeit. Physiol. Chem. 360, 1285–1296, 1979. 417. Salamed, M.A., Robinson, J.L., Naraneetham, D., et al., The amyloid precursor protein/ protease nexin 2 Kunitz inhibibitor domain is a highly speciic substrate of mesotrypsin, J. Biol. Chem. 285, 1939–1949, 2010. 418. Shijetomi, H., Chagi, A., Kajiwara, H., et al., Anti-inflammatory action of serine protease inhibitors containing the Kunitz domain, Inflamm. Res. 59, 679–687, 2010. 419. Lazrak, A., Nita, I., Subramaniyam, D., et al., α1-Antitrypsin inhibits epithelial Na+ in vitro and in vivo, Am. J. Resp. Cell Mol. Biol. 41, 261–270, 2009. 420. Lwaleed, B.A. and Bass, P.S., Tissue factor pathway inhibitor: Structure, biology, and involvement in disease, J. Pathol. 208, 327–339, 2006. 421. Broze, G.J., Jr., Lange, G.W., Dufin, K.L., and MacPhail, L., Heterogeniety of plasma tissue factor pathway inhibitor, Blood Coag. Fibrin. 5, 551–559, 1994. 422. Novotny, W.F., Girard, T.J., Miletich, J.P., and Broze, G.J., Jr., Platelets secrete a coagulation inhibitor functionally and antigenically similar to the lipoprotein associated coagulation inhibitor, Blood 72, 2020–2025, 1988. 423. Li, Y., Spencer, F.A., and Becker, R.C., Plasmin-mediated proteolysis of vascular endothelial cell heparin releasable tissue factor pathway inhibitor, J. Thromb. Thrombolysis 15, 19–23, 2003. 424. Hamuro, T., Kido, H., Asada, Y., et al., Tissue factor pathway inhibitor is highly susceptible to chymase-mediated proteolysis, FEBS J. 274, 3065–3077, 2007. 425. Kothari, H., Kaur, G., Sahoo, S., et al., Plasmin enhances cell surface tissue factor activity in mesothelial and endothelial cells, J. Thromb. Haemost. 7, 121–131, 2009. 426. Piro, O. and Broze, G.J., Jr., Comparison of cell-surface TFPIα and β, J. Thromb. Haemost. 3, 2677–2683, 2005. 427. Maroney, S.A. and Mast, A.E., Expression of tissue factor pathway inhibitor by endothelial cells and platelets, Transfus. Aphresis Sci. 38, 9–14, 2008. 428. Pou, J., Rebollo, A., Piera, L., et al., Tissue factor pathway inhibitor 2 is induced by thrombin in human macrophages, Biochim. Biophys. Acta 1813, 1254–1260, 2011. 429. Schuepbach, R.A., Velez, K., and Riewald, M., Activated protein C up-regulates procoagulant tissue factor activity on endothelial cells by shedding the TFPI Kunitz 1 domain, Blood 117, 6338–6346, 2011. 430. Lindahl, A.K., Abildgaard, U., and Stokke, G., Release of extrinsic pathway inhibitor after heparin injuection: Increased response in cancer patients, Thromb. Res. 59, 651– 656, 1990. 431. Lindahl, A.K., Abildgaard, U., Larsen, M.L., et al., Extrinsic pathway inhibitor (EPI) and the post-heparin anticoagulant effect in tissue thromboplastin induced coagulation, Thromb. Res. Suppl. 14, 39–48, 1991. 432. Abildgaard, U., Tissue factor pathway inhibitor and heparin, Adv. Exp. Med. Biol. 313, 199–204, 1992. 433. Warn-Cramer, B.J., Maki, S.L., and Rapaport, S.I., Heparin-releasable and platelet pools of tissue factor pathway inhibitor in rabbits, Thromb. Haemost. 69, 221–226, 1993. 434. Kragh, M. and Loechel, F., Non-anti-coagulant heparins: A promising approach for prevention of tumor metastasis, Int. J. Oncol. 27, 1159–1167, 2005. 435. Mouse, S.A. and Petersen, L.J., Anti-cancer properties of low-molecular-weight heparin: Preclinical evidence, Thromb. Haemost. 102, 258–267, 2009.

Plasma Proteinase Inhibitors

345

436. Hiremath, M., Newall, F., Ignjatovic, V., et al., The contribution of tissue factor pathway inhibitor to thrombin generation in children receiving unfractionated heparin, Thromb. Res. 127, 275–276. 2011. 437. Harenberg, J., Malsch, R., and Heene, D.L., Tissue factor pathway inhibitor: Proposed heparin recognition region, Blood Coagul. Fibrinol. 6, 550–556, 1996. 438. Valentin, S., Østergaard, P., Kristensen, H., and Nordfang, O., Simultaneous presence of tissue factor pathway inhibitor (TFPI) and low molecular weight heparin has a synergistic effect in different coagulation assays, Blood Coagul. Fibrinol. 2, 629–635, 1991. 439. Nadir, Y., Vlodavsky, I., and Brenner, B., Heparanse, tissue factor, and cancer, Semin. Thromb. Hemost. 34, 187–194, 2008. 440. Nadir, Y. and Brenner, B., Heparanase procoagulant effects and inhibition by heparins, Thromb. Res. 125(Suppl. 2), S72–S76, 2010. 441. Holroyd, E.W. and Simari, R.D., Interdependent biological systems, multi-functional molecules: The evolving role of tissue factor pathway inhibitor beyond anti-coagulation, Thromb. Res. 125(Suppl. 1), S57–S59, 2010. 442. Amirkhosravi, A., Meyer, T., Amaya, M., et al., The role of tissue factor pathway inhibitor in tumor growth and metastasis, Semin. Thromb. Hemost. 33, 643–652, 2007. 443. Wojtokiewicz, M.Z., Sierka, E., and Rak, J., Contribution of the hemostatic system to angiogenesis in cancer, Semin. Thromb. Hemost. 30, 5–20, 2004. 444. Yu, J.L., May, L., Klement, P., et al., Oncogenes as regulators of tissue factor expression and anti-cancer therapy, Semin. Thromb. Hemost. 30, 21–30, 2004. 445. Hengel, L.G. and Versteeg, H.H., Tissue factor signaling: A multi-faceted function in biological proceses, Front. Biosci. 3, 1500–1510, 2011. 446. Cho, Y., Cao, X., Shen, D., et al., Evidence for enhanced tissue factor expression in agerelated macular degeneration, Lab. Invest. 91, 519–526, 2011. 447. Dvorek, H.E. and Rickles, F.R., Malignancy and hemostasis, in Hemostasis and Thrombosis, 5th edn., eds. R.W. Colman, V.J. Marder, A.W. Clowes, J.N. George, and S.Z. Goldhaber, Chapter 57, pp. 851–873, Lippincott, Williams & Wilkins, Philadelphia, 2006. 448. Wojtukiewicz, M.Z., Sierko, E., and Kisiel, W., The role of hemostatic system inhibitors in malignancy, Semin. Thromb. Hemost. 33, 621–642, 2007. 449. Ruf, W., Yokota, N., and Schaffner, F., Tissue factor in cancer progression and angiogenesis, Thromb. Res. 125(Suppl. 2), S36–S38, 2010. 450. Sierko, E., Wojtukiewicz, M.Z., and Kisiel, W., The role of tissue factor pathway inhibitor-2 in cancer biology, Semin. Thromb. Hemost. 33, 653–659, 2007. 451. Piro, O. and Broze, G.J., Jr., Comparison of cell-surface TFPIalpha and beta, J. Thromb. Hemost. 3, 2677–2683, 2005. 452. Stavik, B., Skretting, G., Sletten, M., et al., Overexpression of both TFPIα and TFPIβ induces apoptosis and expression of genes involved in the death receptor pathway in breast cancer cells, Mol. Carcinog. 49, 951–963, 2010. 453. Maroney, S.A., Ferrel, J.P., Pan, S., et al., Temporal expression of alternatively spliced forms for tissue factor pathway inhibitor in mice, J. Thromb. Haemost. 7, 1106–1113, 2009. 454. Van Den Boorgaard, F.E., Brands, X., Schultz, M.J., et al., Tissue factor pathway inhibition and bacterial infection, J. Thromb. Haemost. 9, 122–132, 2011. 455. Schirm, S., Liu, X., Jennings, L.L., et al., Fragmented tissue factor pathway inhibitor (TFPI) and TFPI C-terminal peptides eliminate serum-resistance Echerichia coli from bloood cultures, J. Infect. Dis. 199, 1807–1815, 2009. 456. Regnault, V., Perret-Guillaume, C., Kearny-Schwartz, A., et al., Tissue factor pathway inhibitor. A new link among arterial stiffness, pulse pressure, and coagulation in postmenopausal women, Arterioscler. Thromb. Vasc. Biol. 31, 1226–1232, 2011.

346

Biotechnology of Plasma Proteins

457. Wunderlink, R.G., Laterre, P.-F., Francois, B., et al., Recombinant tissue factor pathway inhibitor in severe community-acquired pneumonia, Am. J. Resp. Crit. Care Med. 183, 1561–1568, 2011. 458. Abraham, E., Tissue factor inhibion and clinical trial results of tissue factor pathway inhibitor in sepsis, Crit. Care Med. 28(9 Suppl.), S31–S33, 2000. 459. Abraham, E., Reinhart, K., Opal, S., et al., Eficacy and safety of Tifacogin (recombinant tissue factor pathway inhibitor) in severe sepsis. A randomized controlled trial, JAMA 290, 238–247, 2003. 460. Morange, P.E., Blankenberg, S., Alessi, M.C., et al., Prognostic value of plasma tissue factor and tissue factor pathway inhibitor for cardiovascular death in patients with coronary artery disease: The AtheroGene study, J. Thromb. Haemost. 5, 475– 482, 2007. 461. Kamikura, Y., Wada, H., Yamada, A., et al., Increased tissue factor pathway inhibitor in patients with acute myocardial infarction, Am. J. Hematol. 55, 183–187, 1997. 462. Shimura, M., Wada, H., Wakita, Y., et al., Plasma tissue factor and tissue pathway inhibitor levels in patients with disseminated intravascular coagulation, Am. J. Hematol. 55, 169–174, 1997. 463. Mitchell, C.T., Kamineni, A., Palmas, W., and Cushman, M., Tissue factor pathway inhibitor, vascular risk factors and subclinical atherosclerosis: The Multi-Ethnic Study of Atherosclerosis, Atherosclerosis 207, 277–283, 2009. 464. Yasuda, T., Ueda, T., Kamei, K., et al., Plasma tissue factor pathway inhibitor levels in patients with acute pancreatitis, J. Gastroenterol. 44, 1071–1079, 2009. 465. Undas, A., Jankoski, M., Kaczmark, P., et al., Thrombin generation in chronic obstructive pulmonary disease: Dependence on plasma factor composition, Thromb. Res. 125, e24–e28, 2011. 466. Golino, P., Ravera, A., Ragni, M., et al., Involvement of tissue factor pathway inhibitor in the coronary circulation of patients with acute coronary syndrome, Circulation 108, 2864–2869, 2003. 467. Ignjatovic, V., Than, J., Summerhaves, R., et al., Hemostatic reponse in paediatric patients undergoing cardiopulmonary bypass surgery, Pediatr. Cardiol. 32, 621–627, 2011. 468. Van Dreden, P., Grosly, M., and Cost, H., Total and free levels of tissue factor pathway inhibitor: A risk factor in patients with factor V Leiden?, Blood Coagul. Fibrinol. 10, 115–116, 1999. 469. Donahue, B.S., Gailiani, D., and Mast, A.E., Disposition of tissue factor pathway inhibitor during cardiopulmonary bypass, J. Thromb. Haemost. 4, 1011–1116, 2006. 470. Kemme, M.J.B., Burggraaf, J., Schomaker, R.C., et al., Quantiication of heparininduced TFPI release: A maximum release at low heparin dose, Br. J. Clin. Pharmacol. 54, 627–634, 2002. 471. Palmer, M.O., Hall, L.J., Reisch, C.M., et al., Clearance of recombinant tissue factor pathway inhibitor (TFPI) is rabbits, Thromb. Haemost. 68, 33–36, 1992. 472. Bregengaard, P., Nordfang, O., Ostergaard, P., et al., Pharmacokinetics of full length and two-domain tissue factor pathway inhibitor in combination with heparin, Thromb. Haemost. 70, 454–457, 1993. 473. Holst, J., Lindblad, B., Westerland, G., et al., Pharmacokinetics and delayed experimental anti-thrombotic effect of two domain non-glycosylated tissue factor pathway inhibitor, Thromb. Res. 81, 461–470, 1996. 474. Kamikubo, Y., Hamuro, T., Matsuda, J., et al., The clearance of proteoglycan-associated human recombinant tissue factor pathway inhibitor (h-rTFPI) in rabbits: A complex formation of h-rTFPI with factor Xa promote a clearance rate of h-rTFPI, Thromb. Res. 83, 161–173, 1996.

Plasma Proteinase Inhibitors

347

475. van’t Veer, C., Hackeng, T.M., Delahaye, C., et al., Activated factor X and thrombin formation triggered by tissue factor on endothelial cell matrix in a low model: Effect of the tissue factor pathway inhibitor, Blood 84, 1132–1142, 1994. 476. Erhardtsen, E., Ezban, M., Madsen, M.T., et al., Blocking of tissue factor pathway inhibitor (TFPI) shortens the bleeding time in rabbits with antibody induced haemophilia A, Blood Coagul. Fibrinolysis 6, 388–395, 1995. 477. Liu, T., Scallan, C.D., Broze, G.J., Jr., et al., Improved coagulation in bleeding disorders by non-anticoagulant sulfated polysaccharides (NASP), Thromb. Haemost. 95, 68–76, 2006. 478. Waters. E.K., Genga, R.M., Schwartz, M.C., et al., Aptamer ARC19499 mediates a procoagulant hemostatic effect by inhibiting tissue factor pathway inhibitor, Blood 117, 5514–5522, 2011. 479. Parunov, L.A., Fadeeva, O.A., Balandina, A.N., et al., Improvement of spatial ibrin formation by the anti-TFPI aptamer BAX499: Changing clot size by targeting extrinsic pathway initiation, J. Thromb. Haemost. 9, 1825–1834, 2011. 480. Kisiel, W., Recollections on the discovery of factor VIIa as a novel therapeutic agent for hemophiliacs with inhibitors, J. Thromb. Haemost. 7, 1053–1056, 2009. 481. Bidwell, E., Booth, J.M., Dike, G.W., and Denson, K.W., The preparation for therapeutic use of a concentrate of factor IX containing also factors II, VII, and X, Br. J. Haematol. 13, 568–579, 1967. 482. Kelly, P. and Penner, J.A., Antihemophilic factor inhibitors. Management with prothrombin complex concentrates, JAMA 236, 2061–2064, 1976. 483. Blatt, P.M., Lundblad, R.L., Kingdon, H.S., et al., Thrombogenic materials in prothrombin complex concentrates, Ann. Intern. Med. 81, 766–770, 1974. 484. Köhler, M., Heiden, M., Harbauer, G., et al., Comparison of different prothrombin complex concentrates—In vitro and in vivo studies, Thromb. Res. 60, 63–70, 1990. 485. Fekete, L.F., Holst, S.L., Peeton, F., et al., “Auto”-factor IX concentrate: A new therapeutic approach to treatment of hemophilia A patients with inhibitors, Fourteenth International Congress of Hematology, Sao Paulo, Brazil, 1972 (cited in Kelly, P. and Penner, J.A., Antihemophilic factor inhibitors. Management with prothrombin complex concentrates, JAMA 236, 2061–2064, 1976). 486. Kurcynski, K.M. and Penner, J.A., Activated prothrombin complex concentrate for patients with factor VIII inhibitors, New Eng. J. Med. 291, 164–167, 1974. 487. Abildgaard, C.F., Penner, J.A., and Watson-Williams, E.J., Anti-inhibitor coagulant complex (Autoplex) for treatment of factor VIII inhibitors in hemophilia, Blood 56, 978–984, 1980. 488. Collen, D., On the regulation and control of ibrinolysis, Thromb. Haemost. 43, 77–89, 1980. 489. Lijnen, H.R. and Collen, D., Interaction of plasminogen activators and inhibitors with plasminogen and ibrin, Sem. Thromb. Haemost. 8, 2–10, 1982. 490. Francis, C.W. and Marder, V.J., A molecular model of plasmic degradation of crosslinked ibrin, Sem. Thromb. Haemost. 8, 25–35, 1982. 491. Booth, N.A. and Bachmann, F., Plasminogen–plasmin system, in Hemostasis and Thrombosis. Basic Principles and Clinical Practice, 5th edn., eds. R.W. Colman, V.J. Marder, A.W. Clowes, J.N. George, and S.Z. Goldhaber, Chapter 18, pp. 335–365, Lippincott, Williams & Wilkins, Philadelphia, 2006. 492. Marder, V.J. and Francis, C.W., Physiologic regulation of ibrinolysis, in Hemostasis and Thrombosis. Basic Principles and Clinical Practice, 5th edn., eds. R.W. Colman, V.J. Marder, A.W. Clowes, J.N. George, and S.Z. Goldhaber, Chapter 23, pp. 419–436, Lippincott, Williams & Wilkins, Philadelphia, 2006. 493. Schaller, J. and Gerber, S.S., The plasmin–antiplasmin system: Structural and functional aspects, Cell. Mol. Life Sci. 68, 785–801, 2011.

348

Biotechnology of Plasma Proteins

494. Collen, D., Identiication and some properties of a new fast-reacting plasmin inhibitor in human plasma, Eur. J. Biochem. 69, 205–216, 1976. 495. Collen, D., De Cock, F., and Verstrate, M., Immunochemical distinction between antiplasmin and α1-antitrypsin, Thromb. Res. 7, 245–249, 1975. 496. Shieh, B.-H. and Travis, J., The reactive site of human α2-antiplasmin, J. Biol. Chem. 262, 6055–6059, 1987. 497. Nobar, S.M., Guy-Crotte, O., Rabaud, M., and Bieth, J.G., Inhibition of human pancreatic proteinases by human α2-antiplasmin and antithrombin, Biol. Chem. 385, 423–427, 2004. 498. Levi, M., Roem, D., Kamp, A.M., et al., Assessment of the relative contribution of different protease inhibitors to the inhibition of plasmin in vivo, Thromb. Haemst. 69, 141– 146, 1993. 499. Banbula, A., Zimmerman, T.P., and Novokhatny, V.V., Blood inhibitory capacity toward exogenous plasmin, Blood Coagul. Fibrinol. 18, 241–246, 2007. 500. Brown, E.W., Ravindran, S., and Patston, P.A., The reaction between plasmin and C1-inhibitor results in plasmin inhibition by the serpin mechanism, Blood Coagul. Fibrinol. 13, 711–714, 2002. 501. Wallace, E.M., Perkins, S.J., Sim, R.B., et al., Degradation of C1-inhibitor by plasmin: Implications for control of inlammatory processes, Mol. Med. 3, 385–396, 1997. 502. Brower, M.S. and Harpel, P.C., Proteolytic cleavage and inactivation of α2-plasmin inhibitor and C1 inactivator by human polymorphonuclear leukocyte elastase, J. Biol. Chem. 257, 9849–9854, 1982. 503. Kang, U.B., Baek, J.H., Ryu, S.H., et al., Kinetic mechanism of protease inhibition by α1-antitrypsin, BIochem. Biophys. Res. Commun. 323, 409–415, 2004. 504. Sazonova, I.Y., Thomas, B.M., Gladysheva, I.C., et al., Fibrinolysis is ampliied by converting α2-antiplasmin from a plasmin inhibitor to a substrate, J. Thromb. Haemost. 5, 2087–2094, 2007. 505. Carpenter, S.L. and Mathew, P., α2-Antiplasmin and its deiciency: Fibrinolysis out of balance, Haemophilia 14, 1250–1254, 2008. 506. Francis, R.B., Jr., Clinical disorders of ibrinolysis: A critical review, Blut 59, 1–14, 1989. 507. Williams, E.C., Plasma alpha-2-antiplasmin activity. Role in the evaluation and management of ibrinolytic states and other bleed disorders, Arch. Intern. Med. 149, 1769–1772, 1989. 508. Miles, L.A., Plow, E.F., Donnelly, K.J., et al., A bleeding disorder due to deiciency of α2-antiplasmin, Blood 59, 1246–1251, 1982. 509. Towne, J.B., Bandyk, D.F., Hussey, C.V., and Tollack, V.T., Abnormal plasminogen: A genetically determined cause of hypercoagulability, J. Vasc. Surg. 1, 896–902, 1984. 510. Collen, D. and Verstrate, M., α2-Antiplasmin consumption and ibrinogen breakdown during ibrinolytic therapy, Thromb. Res. 14, 631–639, 1979. 511. Simpson, M.L., Goldenberg, N.A., Jacobson, L.J., et al., Simultaneous thrombin and plasma generation capacities in normal and abnormal states of coagulation in children and adults, Thromb. Res. 17, 317–323, 2011. 512. Wassenaar, T., Black, J., Kahl, B., et al., Acute promyelocytic leukemia and acquired alpha-2-antiplasmin inhibits deiciency: A retrospective look at the use of epsilonaminocaproic acid (Amicar) in 30 patients, Hematol. Oncol. 26, 241–246, 2008. 513. Henry, D., Carless, P., Ferguson, D., and Laupacis, A., The safety of aprotinin and lysine-derived antiibrinolytic drugs in cardiac surgery: A meta-analysis, CMAJ 180, 183–193, 2009.

Plasma Proteinase Inhibitors

349

514. Roberts, I., Shaker, H., Ker, K., and Coats, T., Antiibrinolytic drugs for acute traumatic injury, Cochrane Database Syst. Rev. 19(1), CD004896, 2011. 515. Koster, A. and Schimer, U., Re-evaluation of the role of antiibrinolytic therapy with lysine analogues during cardiac surgery during the post-aprotinin era, Curr. Opin. Anaesthesiol. 24, 92–97, 2011. 516. Kang, H.-M., Kalnoski, M.H., Frederick, M., and Chandler, W.L., The kinetics of plasmin inhibition by aprotinin in vivo, Thromb. Res. 115, 327–340, 2005. 517. Pipan, C.M., Glasheen, W.P., Matthew, T.L., et al., Effects of ibrinolytic agents on the life span of ibrin sealant, J. Surg. Res. 53, 402–407, 1992. 518. Ngaage, D.L. and Bland, J.M., Lessons from aprotinin: Is the routine use of and inconsistent dosing of transexamic acid prudent? Meta-analysis of randomized and large matched observational studies, Eur. J. Cardiothorac. Surg. 37, 1375–1383, 2010. 519. Dietrich, W., Aprotinin—1 year on, Curr. Opin. Anaesthesiol. 22, 121–127, 2009. 520. Lee, K.N., Jackson, C.W., Christiansen, V.J., et al., A novel plasma proteinase potentiates α2-antiplasmin inhibition of ibrin digestion, Blood 103, 3783–3788, 2004. 521. Sakata, Y. and Aoki, N., Cross-linking of α2-plasmin inhibitor to ibrin by ibrinstabilizing factor, J. Clin. Invest. 65, 290–297, 1980. 522. Hortin, G., Fok, K.F., Toven, P.C., and Strauss, A.W., Sulfation of a tyrosine residue in the plasmin binding domain of α2-antiplasmin, J. Biol. Chem. 262, 3082–3085, 1988. 523. Holmes, E., Nelles, L., Lijnen, H.R., and Collen, D., Primary structure of human α2-antiplasmin, a serine protease inhibitor/serpin, J. Biol. Chem. 262, 1659–1664, 1987. 524. Tone, M., Kikuno, R., Kume-Iwaki, A., and Hashimoto-Gotoh, T., Structure of human α2-plasmin inhibitor derived from the cDNA sequence, J. Biochem. 102, 1033–1041, 1987. 525. Lijnen, H.R., Holmes, W.E., van Hoef, B., et al., Amino-acid sequence of human α2-antiplasmin, Eur. J. Biochem. 166, 565–574, 1987. 526. Högstorp, H. and Carlin, G., Studies on biosynthesis of alpha-2-antiplasmin in rat liver cells, Thromb. Res. 49, 307–318, 1988. 527. Koyama, T., Kake, Y., Toyota, S., et al., Different NH2-terminal form with 12 additional residues of α2-plasmin inhibitor from human plasma and culture media, of Hep G2 cells, Biochem. Biophys. Res. Commun. 200, 417–422, 1994. 528. Law, R.H.P., Soian, T., Kan, W.-T., et al., X-ray crystal structure of the ibrinolysis inhibitor α2-antiplasmin, Blood 111, 2049–2052, 2008. 529. Frank, P.S., Douglas, J.T., Locher, M., et al., Structural/functional characterization of the α2-plasmin inhibitor C-terminal peptide, Biochemistry 42, 1078–1085, 2003. 530. Rossi, V., Balby, I., Ancelet, S., et al., Functional characterization of the recombinant human C1 inhibitor serpin domain: Insights into heparin binding, J. Immunol. 184, 4982–4989, 2010. 531. Lee, K.N., Lee, C.S., Tae, W.-C., et al., Crosslinking of α2-antiplasmin to ibrin, Ann. N. Y. Acad. Sci. 936, 335–339, 2001. 532. Lee, K.N., Jackson, K.W., Christiansen, V.J., et al., Why α-antiplasmin must be converted to a derivative form for optimal function, J. Thromb. Haemost. 5, 2095–2104, 2007. 533. Mosesson, M.W., Siebenlist, K.R., Hernandez, I., et al., Evidence that α2-antiplasmin becomes covalently ligated to plasma ibrinogen in the circulation: A new role for plasma factor XIII in ibrinolysis regulation, J. Thromb. Haemost. 6, 1565–1570, 2008. 534. Mosesson, M.W. and Finlayson, J.S., Biochemical and chromatographic studies of certain activities associated with human ibrinogen preparation, J. Clin. Invest. 42, 747– 755, 1963.

350

Biotechnology of Plasma Proteins

535. Jámbor, C. Reul, V., Schnider, T.W., et al., In vitro Inhibition of factor XIII retards clot formation, reduces clot irmness, and increases ibrinolytic effects in whole blood, Anaesthes. Analg. 109, 1023–1028, 2009. 536. Nair, S.C., Dargaud, Y., Chitlur, M., and Srivastava, A., Tests of global haemostasis and their applications in bleeding disorders, Haemophilia 16(Suppl. 5), 85–92, 2010. 537. De Pietri, L., Montalti, R., Begliomini, B., et al., Thromboeleastographic changes in liver and pancreatic cancer surgery: Hypercoagulability, hypocoagulability or normocoagulability, Eur. J. Anaesthesiol. 27, 608–616, 2010. 538. Mutch, N.J., Koikkalainen, J.S., Fraser, S.R., et al., Model thrombi formed under low reveals the role of factor XIII-mediated cross-linking in resistance to ibrinolysis, J. Thromb. Haemost. 8, 2017–2024, 2010. 539. Chandler, A.B., In vitro thrombotic coagulation of the blood: A method for producing a thrombus, Lab. Invest. 7, 110–114, 1958. 540. Mutch, N.J., Moore, N.R., Mattsson, C., et al., The use of the Chandler loop to examine the interaction potential of NXY-059 or the thrombolytic properties of rtPA on human thrombin in vitro, Br. J. Pharmacol. 153, 124–131, 2008. 541. Tsurupa, G., Yakovlev, S., McKee, P., and Medved, L., Noncovalant interaction of α2-antiplasmin with ibrin(ogen): Localization of α2-antiplasmin-binding sites, Biochemistry 49, 7643–7651, 2010. 542. Zamarron, C., Ginsberg, M.H., and Plow, E.F., Monoclonal antibodies speciic for a conformation different state of ibrinogen, Thromb. Haemost. 64, 41–46, 1990. 543. Morris, T.A., Marsh, J.J., Chiles, P.G., et al., Fibrin derived from patients with chronic thromboembolic pulmonary hyptertension is resistant to lysis, Am. J. Resp. Crit. Care Med. 173, 1270–1275, 2006. 544. Stone, S.R., Whisstock, J.C., Bottomley, S.P., and Hopkins, P.C.R., Serpins: A mechanistic class of their own, Adv. Exp. Med. Biol. 425, 5–15, 1997. 545. Faraday, C.J. and Craik, C.S., Mechanisms of macromolecular protease inhibitors, ChemBioChem 11, 2341–2346, 2010. 546. Wiman, B. and Collen, D., On the mechanism of the reaction between α2-antiplasmin and plasmin, J. Biol. Chem. 254, 9291–9297, 1979. 547. Shieh, B.-H. and Travis, J., The reaction site of human α2-antiplasmin, J. Biol. Chem. 262, 6055–6059, 1987. 548. Christensen, S., Valnickova, Z., Thøgersen, I.B., et al., Assignment of a single disulide bridge in human α2-antiplasmin: Implications for the structural and functional properties, Biochem. J. 323, 847–852, 1997. 549. Christensen, S., Berglund, L., and Sottrup-Jensen, L., Primary structure of bovine α2-antiplasmin, FEBS Lett. 343, 223–228, 1994. 550. Jobse, B.N., Sutherland, J.S., Vaz, D., et al., Molecular cloning and functional expression of rabbit α2-antiplasmin, Blood Coagul. Fibrinol. 17, 283–291, 2006. 551. Creighton, T.E., Kinetic study of protein unfolding and refolding using urea gradient electrophoresis, J. Mol. Biol. 137, 61–80, 1980. 552. Attanasio, R., Stunz, G.W., and Kennedy, R.C., Folding patterns of immunoglobulin molecules identiied by urea gradient electrophoresis, J. Biol. Chem. 269, 1834–1838, 1994. 553. Knaupp, A.S., Levina, V., Robertson, A.L., et al., Kinetic instability of the serpin Z α1-antitrypsin promotes aggregation, J. Mol. Biol. 396, 375–383, 2010. 554. Riordan, J.F., Bier, M., and Nord, F.F., On the mechanism of enzyme action. 70. Urea denaturation of trypsin and acyltrypsins, Arch. Biochem. Biophys. 90, 125–131, 1960. 555. Delaage, M. and Lazdunski, M., Trypsinogen, trypsin, trypsin–substrate and trypsin– inhibitor complexes in urea solutions, Eur. J. Biochem. 4, 378–384, 1968.

Plasma Proteinase Inhibitors

351

556. Stevens, F.C. and Doskoch, E., Lima bean trypsin inhibitor: Reduction and reoxidation of the disulide bonds and their reactivity in the trypsin–inhibitor complex, Canad. J. Biochem. 51, 1021–1028, 1973. 557. Irving, J.A., Pike, R.N., Lesk, A.M., and Whissock, J.C., Phylogeny of the serpin superfamily: Implications of patterns of amino acid conservation for structure and function, Genome Res. 10, 1845–1864, 2000. 558. Plow, E.F., Wiman, B., and Collen, D., Changes in antigenic structure and conformation of α2-antiplasmin induced by interaction with plasmin, J. Biol. Chem. 255, 2902–2906, 1980. 559. Nilsson, T., Sjöholm, I., and Wiman, B., Circular dichroism studies on α2-antiplasmin and its interactions with plasmin and plasminogen, Biochim. Biophys. Acta 705, 264– 270, 1982. 560. Piët, M.P., Chin, S., Prince, A.M., et al., The use of tri (n-butyl)phosphate detergent mixtures to inactivate hepatitis viruses and human immunodeiciency virus in plasma and plasma’s subsequent fractionation, Transfusion 30, 591–598, 1990. 561. Hellstern, P. and Solheim, B.G., The use of solvent/detergent treatment in pathogen reduction of plasma, Transfus. Med. Hemother. 38, 65–70, 2011. 562. Mast, A.E., Stadanlick, J.E., Lockett, M., and Dietzen, D.J., Solvent/detergent-treated plasma has decreased antitrypsin activity and absent antiplasmin activity, Blood 94, 3922–3927, 1999. 563. Burnouf, T., Goubran, H.A., Radosevich, M., et al., Impact of Triton X-100 on alpha 2-antiplasmin (SERPINF2) activity in solvent/detergent-treated plasma, Biologicals 35, 349–353, 2007. 564. Edmunds, L.H., Jr., Managing ibrinolysis without aprotinin, Ann. Thorac. Surg. 89, 324–331, 2010. 565. Manjunath, G., Fozailoff, A., Mitcheson, D., and Sarnak, M.J., Epsilon-aminocaproic acid and renal complications: Case report and review of the literature, Clin. Nephrol. 58, 63–67, 2002. 566. Tavenner, R.W., Use of transexamic acid in control of haemorrhage after extraction of teeth in haemophilia and Christmas disease, Br. Med. J. 2, 314–315, 1972. 567. Ghosh, K., Shetty, S., and Kulkarni, B., Correlation of thromboelastographic patterns with severe haemophilia patients, Haemophilia 13, 734–739, 2007. 568. Sweeney, W.M., Aminocaproic acid, an inhibitor of ibrinolysis, Am. J. Med. Sci. 249, 576–589, 1965. 569. Brown, N.J., Therapeutic potential of plasminogen activator inhibitor-1 inhibitors, Ther. Adv. Cardiovasc. Dis. 4, 315–324, 2010. 570. Izuhara, Y., Yamaoka, N., Kodama, H., et al., A novel inhibitor of plasminogen activator inhibitor-1 provides antithrombotic beneits devoid of bleeding effect in nonhuman primates, J. Cereb. Blood Flow Metab. 30, 904–912, 2010. 571. Gramling, M.W. and Church, F.C., Plasminogen activator inhibitor-1 is an aggregate response factor with pleiotropic effects on cell signaling in vascular disease and the tumor microenvironment, Thromb. Res. 125, 377–381, 2010. 572. Dawley, B., Alpha II antiplasmin deiciency complicating pregnancy: A case report, Obstet. Gynecol. Int. 2011, 698648, 2011. 573. Schaefer, B.M., Maier, K., Eickhoff, U., et al., Alpha-2-antiplasmin and plasminogen activator inhibitors in healing human skin wounds, Arch. Dermatol. Res. 288, 122–128, 1996. 574. Kanno, Y., Hirade, K., Ishisaki, A., et al., Lack of α2-antiplasmin improves cutaneous would healing via over-released endothelial growth factor-induced angiogenesis in wound lesions, J. Thromb. Haemost. 4, 1602–1610, 2006. 575. Hatziapostolou, M., Katsoris, P., and Papadimitriou, E., Different inhibitors of plasmin differentially affect angiostatin production and angiogenesis, Eur. J. Pharmacol. 460, 1–8, 2003.

352

Biotechnology of Plasma Proteins

576. Van Leer, C., Stutz, M., Haeberli, A., and Geiser, T., Urokinase plasminogen activator released by alveolar epithelial cells modulates alveolar epithelial repair in vitro, Thromb. Haemost. 94, 1257–1264, 2005. 577. Caillot, F., Hiron, M., Goria, O., et al., Novel serum markers of ibrosis progression for the follow-up of hepatitis C virus-infected patients, Am. J. Pathol. 175, 46–53, 2009. 578. Chan, K.Y., Lai, P.B., Squire, J.A., et al., Positional expression proiling indicates candidate genes in deletion hotspots of hepatocellular carcinoma, Mod. Pathol. 19, 1546–1554, 2006. 579. Lei, H., Velez, G., Hovland, P., et al., Plasmin is the major protease responsible for processing PDGF-C in the vitreous of patients with proliferative vitreoretinopathy, Invest. Ophthalmol. Vis. Sci. 49, 42–48, 2008. 580. Michael, I.P., Sotiropoulou, G., Pampalakis, G., et al., Biochemical and enzymatic characterization of human kallikrein 5(hK5), a novel serine protease potentially involved in cancer progression, J. Biol. Chem. 280, 14628–14635, 2005. 581. Beinrohr, L., Murray-Rust, T.A., Dyksterhuis, L., et al., Serpins and the complement system, Meth. Enzymol. 499, 55–75, 2011. 582. Borsos, T. and Circolo, A., Binding of activation of C1 by cell bound IgG: Activition depends on cell surface hapten density, Mol. Immunol. 20, 433–438, 1983. 583. Kratz, H.J., Borsos, T., and Isliker, H., Mouse monoclonal antibodies at the red cell surface. II: Effect of hapten density on complement ixation and activation, Mol. Immunol. 22, 229–235, 1985. 584. Wong, J.T. and Colvin, R.B., Bi-speciic monoclonal antibodies: Selective binding and complement ixation to cells that express two different surface antigens, J. Immunol. 139, 1369–1374, 1987. 585. Borsos, T. and Rapp, H.J., Complement ixation on cell surfaces by 19S and 7S antibodies, Science 150, 505–506, 1965. 586. Volanakis, J.E., Overview of the complement system, in The Human Complement System in Health and Disease, eds. J.E. Volanakis and M.M. Frank, Chapter 2, pp. 9–32, Marcel Dekker, New York, 1998. 587. Nagaki, K. and Stroud, R.M., Speciic antisera to C1s: Detection of different electrophoretic species of C1s, J. Immunol. 103, 141–145, 1969. 588. Ratnoff, O.D. and Lepow, I.H., Some properties of a serine esterase derived from preparations of the irst component of complement, J. Exp. Med. 106, 327–343, 1957. 589. Pensky, J., Levy, R.L., and Lepow, I.H., Partial puriication of a serum inhibitor of C1 esterase, J. Biol. Chem. 236, 1674–1679, 1961. 590. Frank, M.M., Introduction and historical notes, in The Human Complement System in Health and Disease, eds. J.E. Volanakis and M.M. Frank, Chapter 1, pp. 1–8, Marcel Dekker, New York, 1998. 591. Morgan, B.P. and Harris, C.L., Complement Regulatory Proteins, Academic Press, San Diego, CA, 1999. 592. Osborn, T.B., Complement or Alexin, Oxford University Press, London, 1937. 593. Kabat, E.A. and Mayer, M.M., Experimental Immunology, 2nd edn., C.C. Thomas, Springield, IL, 1961. 594. Wolstenholme, G.E.W. and Knight, J. (eds.), Complement, Little, Brown, and Company, Boston, MA, 1965. 595. Osler, A.G., Complement. Mechanisms and Functions, Prentice-Hall, Englewood Cliffs, NJ, 1964. 596. Whaley, K. (ed.), Methods in Complement for Clinical Immunologists, Churchill Livingstone, Edinburgh, Scotland, UK, 1985. 597. Career, F.M., The C1 inhibitor deiciency. A review, Eur. J. Clin. Chem. Clin. Biochem. 30, 793–807, 1992.

Plasma Proteinase Inhibitors

353

598. Davis, A.E., C1 inhibitor gene and hereditary angioedema, in The Human Complement System in Health and Disease, eds. J.E. Volanakis and M.M. Frank, Chapter 21, pp. 455– 480, Marcel Dekker, New York, 1998. 599. Weis, M., Clinical review of hereditary angioedema: Diagnosis and management, Postgrad. Med. 121, 113–120, 2009. 600. Nagy, N., Grattan, C.E., and McGrath, J.A., New insights into hereditary angio-oedema: Molecular diagnosis and therapy, Australas. J. Dermatol. 51, 157–162, 2010. 601. Keating, G.M., Human C1-esterase inhibitor concentrate (Berinert®), BioDrugs 23, 399–406, 2009. 602. Wouters, D., Wagenaar-Bos, I., van Ham, H., and Zeerleder, S., C1 inhibitor: Just a serin protease inhibitor? New and old considerations on therapeutic applications of C1 inhibitor, Expert Opin. Biol. Ther. 8, 1225–1240, 2008. 603. Riedl, M., Gower, R.G., and Chrvala, C.A., Current medical management of hereditary angioedema: Results from a large survey of US physicians, Ann. Allergy Asthma Immunol. 106, 316–322, 2011. 604. Breitbart, S.I. and Bielory, L., Acquired angioedema: Autoantibody association and C1q utility as a diagnostic tool, Allergy Asthma Proc. 31, 428–434, 2010. 605. Farkas, H. and Varga, L., Ecallantide is a novel treatment for attacks of hereditary angioedema due to C1 inhibitor deiciency, Clin. Cosmet. Investig. Dermatol. 4, 61–68, 2011. 606. Banta, E., Horn, P., and Craig, T.J., Response to ecallantide treatment of acute attacks of hereditary angioedema based on time to intervention: Results from EDEMA clinical trials, Allergy Asthma Proc. 32, 319–324, 2011. 607. Frank, M.M., Hereditary angioedema, J. Allergy Clin. Immunol. 121(2 Suppl.), S398– S401, 2008. 608. Maurer, M. and Magerl, M., Long-term prophylaxis of hereditary angioedema with androgen derivatives: A critical appraisal and potential alternatives, J. Dtsch. Dermatol. Ges. 9, 99–107, 2011. 609. Davis, A.E., Mejia, P., and Lu, F., Biological activities of C1 inhibitor, Mol. Immunol. 45, 4057–4063, 2008. 610. Davis III, A.E., Lu, F., and Mejia, P., C1 inhibitor, a multi-functional serine protease inhibitor, Thromb. Haemost. 104, 886–893, 2010. 611. Bossi, F., Bulla, R., and Tedesco, F., Endothelial cells are a target of both complement and kinin system, Int. Immunopharmacol. 8, 143–147, 2008. 612. Dorresteijn, M.J., Visser, T., Cox, L.A., et al., C1-esterase inhibitor attenuates the inlammatory response during human endotoxemia, Crit. Care. Med. 38, 2139–2145, 2010. 613. Thorgersen, E.B., Ludviksen, J.K., Lambris, J.D., et al., Anti-inlammatory effects of C1-inhibitor in porcine and human whole blood are independent of its protease inhibition activity, Innate Immun. 16, 254–264, 2010. 614. Wagnenaar-Bos, I.G. and Hack, C.E., Structure and function of C1-inhibitor, Immunol. Allergy Clin. North Am. 26, 615–632, 2006. 615. Nilsson, T. and Wiman, B., Puriication and characterization of human C1-esterase inhibitor, Biochim. Biophys. Acta 705, 271–276, 1982. 616. Harrison, R.A., Human C1 inhibitor: Improved isolation and preliminary structural characterization, Biochemistry 22, 5001–5007, 1983. 617. Bock, S.C., Skriver, K., Nielsen, E., et al., Human C1 inhibitor: Preliminary structure, cDNA cloning and chromosomal locatization, Biochemistry 25, 4292–4301, 1986. 618. Perkins, S.J., Smith, K.F., Amatayakul, S., et al., Two-domain structure of the native and reactive centre cleaved forms of C1 inhibitor of human complement by neutron scattering, J. Mol. Biol. 214, 751–763, 1990.

354

Biotechnology of Plasma Proteins

619. Kyte, J., Structure in Protein Chemistry, 2nd edn., Chapter 12, Physical measurements of structure, Garland Science, New York, 2007. 620. Ward, D.N. and Arnott, M.S., Gel iltration of proteins with particular reference to the glycoprotein luteinizing hormone, Anal. Biochem. 12, 296–302, 1965. 621. Andrews, P., Estimation of the molecular weight of proteins by Sephadex gel iltration, Biochem. J. 91, 222–233, 1964. 622. Ackers, G.K., Molecular sieve methods of analysis, in The Proteins, 3rd edn., eds. H. Neurath and R.L. Hill, Vol. 1, Chapter 1, pp. 1–94, Academic Press, New York, 1965. 623. Pitt-Rivers, R. and Impiombato, F.S.A., The binding of sodium dodecyl sulphate to various proteins, Biochem. J. 109, 825–830, 1968. 624. Weber, K. and Osborn, M., Proteins and sodium dodecyl sulfate: Molecular weight determination on polyacrylamide gels and related procedures, in The Proteins, 3rd edn., Vol. 1, pp. 179–223, Academic Press, New York, 1968. 625. Fairman, R., Fenderson, W. Hail, M.E., et al., Molecular weights of CTLA-4 and CD-80 by sedimentation equilibrium ultracentrifugation, Anal. Biochem. 270, 286– 295, 1999. 626. Ye, H., Simultaneous determination of protein aggregation, degradation, and absolute molecular weight by size exclusion chromatography-multiangle laser light scattering, Anal. Biochem. 356, 76–85, 2006. 627. Seyfried, B.K., Siekmann, J., Belgacem, O., et al., MALDI linear TOF mass spectrometry of PEGylated (glyco)proteins, J. Mass Spectrom. 45, 612–617, 2010. 628. Müller, R., Marchetti-Deschmann, M., Elgass, H., et al., Molecular weight determination of high molecular mass (glyco)proteins using CGE-on-a-chip, planar SDS-PAGE and MALDI-TOF-MS, Electrophoresis 31, 3850–3862, 2010. 629. Tosi, M., Duponchel, C., Bourgarel, P., et al., Molecular cloning of human C1 inhibitor: Sequence homologies with alpha-1-antitypsin and other members of the serpins super family, Gene 42, 265–272, 1986. 630. Davies III, A.E., Whitehead, A.S., Harrison, R.A., et al., Human inhibitor of the irst component of complement, C1: Characterization of cDNA clones and localization of the gene to chromosome 11, Proc. Natl. Acad. Sci. USA 83, 3161–3165, 1986. 631. Verpy, E., Couture-Tosi, E., Eldering, E., et al., Crucial residues in the carboxy-terminal end of C1 inhibitor revealed by pathogenic mutants impaired in secretion or function, J. Clin. Invest. 95, 350–359, 1995. 632. Bos, I.G., Lubbers, Y.T., Eldering, E., et al., Effect of reactive site loop elongation on the inhibitory activity of C1-inhibitor, Biochim. Biophys. Acta 1699, 139–144, 2004. 633. Harpel, P.C. and Cooper, N.R., Studies on human plasma C1 inhibitor–enzyme interactions. I: Mechanisms of interaction with C1s, plasmin, and trypsin, J. Clin. Invest. 55, 593–604, 1975. 634. Chesne, S., Villiers, C.L., Arlaud, G.J., et al., Fluid-phase interaction of C1 inhibitor (C1 Inh) and the subcomponents C1r and C1s of the irst of component of complement, C1, Biochem. J. 201, 61–70, 1982. 635. Lennick, M., Brew, S.A., and Ingham, K.C., Kinetics of interaction of C1 inhibitor with complement C1s, Biochemistry 25, 3890–3898, 1986. 636. Murray-Rust, T.A., Kerr, F.K., Thomas, A.R., et al., Modulation of the proteolytic activity of the complement protease C1s by polyanions: Implications for polyanionmediated accelerated of interaction between C1s and SERPING1, Biochem. J. 422, 295–303, 2009. 637. Rossi, V., Bally, I., Ancelet, S., et al., Functional characterization of the recombinant human C1 inhibitor serpin domain: Insights into heparin binding, J. Immunol. 184, 4982–4989, 2010.

Plasma Proteinase Inhibitors

355

638. Gozzo, A.J., Nunes, V.A., Cruz-Silva, I., et al., Heparin modulation of human plasma kallikrein on different substrates and inhibitors, Biol. Chem. 387, 1129–1138, 2006. 639. Pixley, R.A., Schapira, M., and Colman, R.W., The regulation of human factor XIIa by plasma proteinase inhibitors, J. Biol. Chem. 260, 1723–1729, 1985. 640. Kaplan, A.P., Enzymatic pathways in the pathogenesis of hereditary angioedema: The role of C1 inhibitor therapy, J. Allergy Clin. Immunol. 126, 918–925, 2010. 641. Van Nostrand, W.E. and Cunningham, D.D., Puriication of a proteinase inhibitor from bovine serum with C1-inhibitor activity, Biochim. Biophys. Acta 923, 167–175, 1987. 642. Sanchez, J., ELgue, G., Riesenfeld, J., et al., Studies of adsorption, activation, and inhibition of factor XII on immobilized heparin, Thromb. Res. 89, 41–50, 1998. 643. Minnema, M.C., Peters, R.J., de Winter, R., et al., Activation of clotting factors XI and IX in patients with acute myocardial infarction, Arterioscler. Thromb. Vasc. Biol. 20, 2489–2493, 2000. 644. Coppola, L., Guastaierro, S., Verrazzo, G., et al., C1 inhibitor infusion modiies platelet activity in hereditary angioedema patients, Arch. Pathol. Lab. Med. 126, 842–845, 2002. 645. Bäck, J., Sanchez, J., Elgue, G., et al., Activated human platelets induce factor XIIa-mediated contact activation, Biochem. Biophys. Res. Commun. 391, 11–17, 2010. 646. Kaplan, A.P. and Ghebrehiwet, B., The plasma bradykinin-forming pathways and its interrelationships with complement, Mol. Immunol. 47, 2161–2169, 2010. 647. Yang, L., Sun, M.F., Gailani, D., and Rezaie, A.R., Characterization of a heparinbinding site on catalytic domain of factor XIa: Mechanism of heparin acceleration of the factor XIa inhibition by the serpins antithrombin and C1-inhibitor, Biochemistry 48, 1517–1524, 2009. 648. Zuraw, B.L. and Christiansen, S.C., Pathogenesis and laboratory diagnosis of hereditary angioedema, Allergy Asthma Proc. 30, 487–492, 2009. 649. Kaplan, A.P., Kinins, airway obstruction, and anaphylaxis, Chem. Immunol. Allergy 95, 67–84, 2010. 650. Schamier, A.H., The elusive physiologic role of factor XII, J. Clin. Invest. 118, 3006– 3009, 2008. 651. Vogler, E.A. and Siedlecki, C.A., Contact activation of blood-plasma coagulation, Biomaterials 30, 1857–1869, 2009. 652. Ebo, D.G., Verweij, M.M., and De Knop, K.J., Hereditary angioedema in childhood: An approach to management, Paediatr. Drugs 12, 257–268, 2010. 653. Rip, J., Nierman, M.C., Ross, C.J., et al., Lipoprotein lipase S447X: A naturally occuring gain-of-function mutation, Arterioscler. Thromb. Vasc. Biol. 26, 1236–1245, 2006. 654. Woulters, D., Wagenaar-Bos, I., van Ham, M., and Zeerleder, S., C1 inhibitor: Just a protease inhibitor? New and old considerations on therapeutic applications of C1 inhibitor, Expert Opin. Biol. Ther. 8, 1225–1240, 2008. 655. Davis III, A.E., Lu, F., and Mejia, P., C1 inhibitor, a multi-functional serine protease inhibitor, Thromb. Haemost. 104, 886–893, 2010. 656. Cheng, Z.-D., Liu, M.-Y., Chen, G., et al., Anti-vascular permeability of the cleaved reactive center loop within the carboxyl-terminal domain of C1 inhibitor, Mol. Immunol. 45, 1743–1751, 2008. 657. Liu, D., Gu, X., Scaidi, J., and Davies III, A.E., N-Linked glycosylation is required for C1 inhibitor-mediated protection from endotoxin shock in mice, Infect. Immun. 72, 1946–1955, 2004. 658. Cai, S., Dole, V.S., Bergmeier, W., et al., A direct role for C1 inibitor in regulation of leukocyte adhesion, J. Immunol. 174, 6462–6466, 2005.

356

Biotechnology of Plasma Proteins

659. Gesuete, R., Storini, C., Fantin, A., et al., Recombinant C1 inhibitor in brain ischemia injury, Ann. Neurol. 66, 332–342, 2009. 660. Kerr, F.K., Thomas, A.R., Wijeyewickrema, L.C., et al., Elucidation of the substrate speciicity of the MASP-2 protease of the lectin complement pathway and identiication of the enzyme as a major physiological target of the serpin C1-inhibitor, Mol. Immunol. 45, 670–677, 2008. 661. Osthoff, M., Katan, M., Fluri, F., et al., Mannose-binding lectin deiciency is associated with smaller infarction size and favorable outcome in ischemic stroke patients, PLoS ONE 6(6), e21338, 2011. 662. Cervera, A., Planas, A.M., Justicia, C., et al., Genetically-deined deiciency of mannosebinding lectin is associated with protection after experimental stroke in mice and outcome in human stroke, PLoS One 5(2), e8433, 2010. 663. Morrison, H., Frye, J., Davis-Gorman, G., et al., The contribution of mannose binding lectin to reperfusion injury after ischemic stroke, Curr. Neurovasc. Res. 8, 52–63, 2011. 664. Tomasi, S., Sarmientos, P., Giorda, G., et al., Mutant prourokinase with adjunctive C1-inhibitor is an effective and safer alternative to tPA in rat stroke, PloS One 6(7), e21999, 2011. 665. Banz, Y. and Rieban, R., Role of complement and perspectives for intervention in ischemia-reperfusion damage, Ann. Med.(in press). 666. Koles, K., van Berkel, P.H., Pieper, F.R., et al., N- and O-glycans of recombinant human C1 inhibitor expressed in the milk of transgenic rabbits, Glycobiology 14, 51–64, 2004. 667. Koles, K., van Berkel, P.H., Mannesse, M.L., et al., Inluence of lactation parameters on the N-glycosylation of recombinant human C1 inhibitor isolated from the milk of transgenic rabbits, Glycobiology 14, 979–986, 2004. 668. Wolff, M.W., Zhang, F., Roberg, J.J., et al., Expression of C1 esterase inhibitor by the baculovirus expression vector system: Preparation, puriication, and characterization, Protein Expr. Purif. 22, 414–421, 2001. 669. Bos, I.G., de Bruin, E.C., Karuntu, Y.A., et al., Recombinant human C1-inhibitor produced in Pichia pastoris has the same inhibitory capacity as plasma C1-inhibitor, Biochim. Biophys. Acta 1648, 75–83, 2003. 670. Lamark, T., Ingebrigtsen, M., Bjørnstad, C., et al., Expression of active human C1 inhibitor serpin domain in Escherichia coli, Protein Expr. Purif. 22, 349–358, 2001. 671. Cai, S. and Davis III, A.E., Complement regulatory protein C1 inhibitor binds to selectins and interferes with endothelial-leukocyte adhesion, J. Immunol. 171, 4785–4791, 2003. 672. Solá, R.J. and Griebenow, K., Glycosylation of therapeutic proteins: An effective strategy to optimize eficacy, BioDrugs 24, 9–21, 2010. 673. Carter, P.J., Introduction to current and future protein therapeutics: A protein engineering perspective, Exp. Cell Res. 317, 1261–1269, 2011. 674. Varga, L. and Farkas, H., rhC1INH: A new drug for the treatment of attacks in hereditary angioedema caused by C1-inhibitor deiciency, Expert Rev. Clin. Immunol. 7, 143–153, 2011. 675. Zuraw, B., Cicardi, M., Levy, R.J., et al., Recombinant human C1-inhibitor for the treatment of acute angioedema attacks in patients with hereditary angioedema, J. Allergy Clin. Immunol. 126, 821–827, 2010. 676. Lunn, M. and Banta, E., Ecallantide for the treatment of hereditary angioedema in adults, Clin. Med. Insights Cardiol. 4, 49–54, 2011. 677. Nicholl, S.M., Roztocil, E., and Davies, M.G., Plasminogen activator system and vascular disease, Curr. Vasc. Pharmacol. 4, 101–116, 2006.

Plasma Proteinase Inhibitors

357

678. Croucher, D.R., Saunders, D.N., Lobov, S., et al., Revisiting the biological roles of PAI2 (SERPINB2) in cancer, Nat. Rev. Cancer. 8, 535–545, 2008. 679. Stief, T., Radtke, K.P., and Heimburger, N., Inhibition of urokinase by protein C-inhibitor (PCI). Evidence for identity of PCI and plasminogen activator inhibitor 3, Biol. Chem. Hoppe Seyler 368, 1427–1433, 1987. 680. Heeb, M.J., España, F., Geiger, M., et al., Immunological identify of heparin-dependent plasma and urinary protein C inhibitor and plasminogen activator inhibitor-3, J. Biol. Chem. 262, 15813–15816, 1987. 681. Schroder, W.A., Major, L., and Suhrbier, A., The role of serpinB2 in immunity, Crit. Rev. Immunol. 31, 15–30, 2011. 682. Yepes, M., Loskutoff, D.J., and Lawrence, D.A., Plasminogen activator inhibitor-1, in Hemostasis and Thrombosis Basic Principles and Clinical Practice, eds. R.W. Colman, V.J. Marder, A.W., Clowes, J.N. George, and S.L. Goldhaber, Chapter 19 pp. 365–380, Lippincott, Williams & Wilkins, Philadelphia, 2006. 683. Cale, J.M. and Lawrence, D.A., Structure–function relationships of plasminogen activator inhibitor-1 and its potential as a therapeutic agent, Curr. Drug Targets 8, 971–981, 2007. 684. Garg, N. and Fay, W.P., Plasminogen activator inhibitor-1 and restenosis, Curr. Drug Targets 8, 1003–1006, 2007. 685. Tsantes, A.E., Nikolopoulos, G.K., Bagos, P.G., et al., The effect of the plasminogen activator inhibitor-1 4G/5G polymorphism on the thrombotic risk, Thromb. Res. 122, 736–742, 2008. 686. Wang, L., Bastarache, J.A., and Ware, L.B., The coagulation cascade in sepsis, Curr. Pharm. Des. 14, 1860–1869, 2008. 687. Mehta, R. and Shapiro, A.D., Plasminogen activator inhibitor type 1 deiciency, Haemophilia 14, 1255–1260, 2008. 688. Dupont, D.M., Madsen, J.B., Kristensen, T., et al., Biochemical properties of plasminogen activator inhibitor-1, Front. Biosci. 14, 1337–1361, 2009. 689. Eddy, A.A., Serine proteases, inhibitors and receptors in renal ibrosis, Thromb. Haemost. 101, 656–664, 2009. 690. Gettins, P.G. and Olson, S.T., Exosite determinants of serpin speciicity, J. Biol. Chem. 284, 20441–20445, 2009. 691. Jankun, J. and Skrzypczak-Jankun, E., Yin and yang of the plasminogen activator inhibitor, Pol. Arch. Med. Wewn. 119, 410–417, 2009. 692. Alessi, M.C., Declerck, P.J., De Mol, M., et al., Puriication and characterization of natural and recombinant human plasminogen activator inhibitor-1 (PAI-1), Eur. J. Biochem. 175, 531–540, 1988. 693. Jackman, R.P., Utter, G.H., Heitman, J.W., et al., Effects of blood sample age at time of separation on measured cytokine concentrations in human plasma, Clin. Vaccine Immunol. 18, 318–326, 2011. 694. Kruithof, E.K.O., Gudinchet, A., and Bachmann, F., Plasminogen activator inhibitor 1 and plasminogen activator inhibitor 2 in various disease states, Thromb. Haemost. 59, 7–12, 1988. 695. Schoenhard, J.A., Smith, L.H., Painter, C.A., et al., Regulation of the PAI-1 promoter by circadian clock components: Differential activation by BMAL1 and BMAL2, J. Mol. Cell Cardiol. 35, 473–481, 2003. 696. Oishi, K., Plasminogen activator inhibitor-1 and the circadian clock in metabolic disorders, Clin. Exp. Hypterten. 31, 208–219, 2009. 697. Bergheanu, S.C., Pons, D., Jukema, J.W., et al., Myocardial infarction occurs with a similar 25 h pattern in the 4G/5G versions of plasminogen activator inhibitor-1, Chronobiol. Int. 26, 637–652, 2009.

358

Biotechnology of Plasma Proteins

698. Oishi, K., Koyanagi, S., Matsunaga, N., et al., Bezaibrate induces plasminogen activator inhibitor-1 gene expression in a CLOCK-dependent circadian manner, Mol. Pharmacol. 78, 135–141, 2010. 699. Ehrlich, H.J., Gebbink, R.K., Keijer, J., et al., Alteration of serpin speciicity by a protein cofactor. Vitronectin endows plasminogen activator inhibitor 1 with thrombin inhibitory properties, J. Biol. Chem. 265, 13029–13035, 1990. 700. van Meijer, M., Smilde, A., Tans, G., et al., The suicide substrate reaction between plasminogen activator inhibitor 1 and thrombin is regulated by the cofactors vitronectin and heparin, Blood 90, 1874–1882, 1997. 701. Rezaie, A.R., Role of exosites 1 and 2 in thrombin reaction with plasminogen activator inhibitor-1 in the absence and presence of cofactors, Biochemistry 38, 14592–14599, 1999. 702. Stoop, A.A., Lupu, F., and Pannekoek, H., Colocalization of thrombin, PAI-1, and vitronectin in the atherosclerotic vessel wall: A potential regulatory mechanism of thrombin activity by PAI-1/vitronectin complexes, Arterioscler. Thromb. Vasc. Biol. 20, 1143– 1149, 2000. 703. Dekker, R.J., Pannekoek, H., and Horrevoets, A.J., A steady-state competition model describes the modulating the effects of thrombomodulin on thrombin inhibition by plasminogen activator inhibitor-1 in the absence and presence of vitronectin, Eur. J. Biochem. 270, 1942–1951, 2003. 704. Seetharam, R., Dwivedi, A.M., Duke, J.L., et al., Puriication and characterization of active and latent forms of recombinant plasminogen activator inhibitor 1 produced in Escherichia coli, Biochemistry 31, 9877–9882, 1992. 705. Gibson, A., Baburaj, K., Day, D.E., et al., The use of luorescent probes to characterize conformational changes in the interaction between vitronectin and plasminogen activator inhibitor-1, J. Biol. Chem. 272, 5112–5121, 1997. 706. Lawrence, D.A., Palaniappan, S., Stefansson, S., et al., Characterization of the binding of different conformational forms of plasminogen activator inhibitor-1 to vitronectin. Implications for the pericellular proteolysis, J. Biol. Chem. 272, 7676–7680, 1997. 707. Cabrita, L.D. and Bottomley, S.P., How do proteins avoid becoming too stable? Biophysical studies into metastable proteins, Eur. Biophys. J. 33, 83–88, 2004. 708. Baldwin, A.J., Knowles, T.P., Tartaglia, G., et al., Metastability of native proteins and the phenomenon of amyloid formation, J. Am. Chem. Soc. 133, 14160–14163, 2011. 709. Im, H. and Yu, M.H., Role of Lys335 in the metastability and function of inhibitory serpins, Protein Sci. 9, 934–941, 2000. 710. Sarkar, A. and Wintrode, P.L., Effects of glycosylation on the stability and lexibility of a metastable protein: The human serpin α1-antitrypsin, Int. J. Mass Spectrom. 302, 69–75, 2011. 711. Stout, T.J., Graham, H., Buckley, D.I., and Matthews, D.J., Structures of active and latent PAI-1: A stabilizing role for chloride ions, Biochemistry 39, 8460–8469, 2000. 712. Thompson, L.C., Goswami, S., Ginsberg, D.S., et al., Metals affect the structure and activity of human plasminogen activator-1. I. Modulation of stability and protease inhibition, Protein Sci. 20, 353–365, 2011. 713. Agren, A., Wiman, B., Stiller, V., et al., Evaluation of low2 PAI-1 activity as a risk factor for hemorrhagic diathesis, J. Thromb. Haemost. 4, 201–208, 2006. 714. Mehta, R. and Shapiro, A.D., Plasminogen activator inhibitor-type 1 deiciency, Haemophilia 14, 1255–1260, 2008. 715. Binder, B.R., Christ, G., Gruber, F., et al., Plasminogen activator inhibitor 1: Physiological and pathophysiological roles, News Physiol. Sci. 17, 56–61, 2002. 716. Shen, G.X., Impact and mechanism for oxidized and glycated lipoproteins on generation of ibrinolytic regulators from vascular endothelial cells, Mol. Cell. Biochem. 246, 69–74, 2003.

Plasma Proteinase Inhibitors

359

717. Oliver, J.J., Webb, D.J., and Newby, D.E., Stimulated tissue plasminogen activator release as a marker of endothelial function in humans, Arterioscler. Thromb. Vasc. Biol. 25, 2470–2479, 2005. 718. Fay, W.P., Garg, N., and Sunkar, M., Vascular functions of the plasminogen activation system, Arterioscler. Thromb. Vasc. Biol. 27, 1231–1237, 2007. 719. Chrusciel, P., Goch, A., Banach, M., et al., Circadian changes in the hemostatic system in healthy men and patients with cardiovascular diseases, Med. Sci. Monit. 15, RA203– RA208, 2009. 720. Wiman, B., Andersson, T., Hallqviust, J., et al., Plasma levels of tissue plasminogen activator/plasminogen activator inhibitor-1 complex and von Willebrand factor are signiicant risk markers for recurrent myocardial infarction in the Stockholm Heart Epidemiology Program (SHEEP) study, Arterioscler. Thromb. Vasc. Biol. 20, 2019– 2023, 2000. 721. Hernestål-Boman, J., Jansson, J.H., Nilsso, T.K., et al., Long-term stability of ibrinolytic factors stored at −80°C, Thromb. Res. 125, 451–456, 2010. 722. Nordenbem, A., Leander, K., Hallquist, J., et al., The complex between tPA and PAI-1: Risk factor for myocardial infarction as study in the SHEEP project, Thromb. Res. 116, 223–232, 2005. 723. Sahli, D., Eriksson, J.W., Boman, K., and Svensson, M.K., Tissue plasminogen activator (tPA) activity is a novel and early marker of assymptomatic LEAD in type 2 diabetes, Thromb. Res. 123, 701–706, 2009. 724. Christ, G., Nikfardjam, M., Huber-Beckmann, R., et al., Predictive value of plasma plasminogen activator-1 for coronary restenosis: Dependence on stent implantation and antithrombotic medication, J. Thromb. Haemost. 3, 233–239, 2005. 725. Mytnik, M. and Staskjo, J., D-dimer, plasminogen activator inhibitor-1, prothrombin fragements and protein C—Role in prothrombotic state of colorectal cancer, Neoplasma 58, 235–238, 2011. 726. van Zaane, B., Squizzato, A., Debeij, J., et al., Alterations in coagulation and ibrinolysis after Levothyroxine exposure in healthy volunteers: A controlled randomized crossover study, J. Thromb. Haemost. 9, 1816–1824, 2011. 727. Suehiro, A., Wakabayashi, I., Uchida, K., et al., Impaired spontaneous thrombolytic activity measured by global thrombosis test in males with metabolic syndrome, Thromb. Res. 129, 499–501, 2012. 728. Folkes, A., Brown, S.D., Canne, L.E., et al., Design, synthesis and in vitro evaluation of potent, novel, small molecule inhibitors of plasminogen activator-1, Bioorg. Med. Chem. Lett. 12, 1063–1066, 2002. 729. Brown, N.J., Therapeutic potential of plasminogen activator inhibitor-1 inhibitors, Ther. Adv. Cardiovasc. Dis. 4, 315–324, 2010. 730. Suzuki, J., Ogawa, M., Muto, S., et al., Effects of speciic chemical suppresssors of plasminogen activator inhibitor-1 in cardiovascular diseases, Expert Opin. Investig. Drugs 20, 255–264, 2011. 731. Ploplis, V.A., Effects of altered plasminogen activator inhibitor-1 on cardiovascular disease, Curr. Drug. Targets 12, 1782–1789, 2011. 732. van der Heide, Y.T., Morriën, W., and van den Ende, A., Evaluation of blood sampling for measurement of tissue-type plasminogen activator (t-PA) and plasminogen activator inhibitor-1 (PAI-1), Fibrinolysis 8(Suppl. 2), 152–153, 1994. 733. Seifried, E., Tanswell, P., Rikjen, D.C., et al., Pharmacokinetics of antigen and activity of recombinant tissue-type plasminogen activator after infusion in healthy volunteers, Arzneim. Forsch. 38, 418–422, 1988. 734. Verheijen, J.H., Mullaart, E., Chang, G.T., et al., A simple, sensitive spectrophotometric assay for extrinsic (tissue-type) plasminogen activator applicable to measurements in plasma, Thromb. Haemost. 48, 266–269, 1982.

360

Biotechnology of Plasma Proteins

735. Tanswell, P., Tissue-type plasminogen activator, in Therapeutic Proteins. Pharmacokinetics and Pharmacodynamics, Chapter 12, pp. 225–281, W.H. Freeman, New York, 1993. 736. Astedt, B., Casslén, B., Lecander, I., et al., Rebound increase of PAI-1 following local intra-arterial rt-PA infusion, a possible cause of reocclusion, Blood Coagul. Fibrinolysis 4, 563–567, 1993. 737. Paganelli, F., Alessi, M.C., Morange, P., et al., Relationship of plasminogen activator inhibitor-1 levels following thrombolytic therapy with rt-PA as compared to streptokinase and patency of infarct related coronary artery, Thromb. Haemost. 82, 104–108, 1999. 738. Mlynaska, A., Waszyrowski, T., and Kasprzak, J.D., Increase in plasma plaminogen activators inhibitor type 1 concentration after ibrinolytic treatment in patients with acute myocardial infarction is associated with 4G/5G polymorphism of PAI-1 gene, J. Thromb. Haemost. 4, 1361–1366, 2006. 739. Lindahl, T.L., Ohlsson, P.-I., and Wiman, B., The mechanism of the reaction between human plasminogen-activator 1 and tissue plasminogen activator, Biochem. J. 265, 109– 113, 1990. 740. Takada, Y. and Takada, A., The activation of plasminogen by T-PA-PAI-1 complex in the absence of ibrin, Thromb. Res. 63, 169–177, 1991. 741. Yano, D., Urano, T., Takada, Y., and Takada, A., Dissociation of α2-plasmin-inhibitor– plasmin complex and regeneration of plasmin activity by SDS treatment, Thromb. Res. 69, 461–469, 1993. 742. Shore, J.D., Day, D.E., Francis-Chumra, A.M., et al., A luorescent probe study of plasminogen activator inhibitor-1. Evidence for reactive center loop insertion and its role in the inhibitory mechanism, J. Biol. Chem. 270, 5395–5398, 1995. 743. Olson, S.T., Swanson, R., Day, D., et al., Resolution of Michaelis complex, acylation, and conformational change steps in the reactions of serpin, plasminogen activator inhibitor-1, with tissue plasminogen activator and trypsin, Biochemistry 40, 11742–11756, 2001. 744. Nilsson, T.K., Boman, K., Jansson, J.-H., et al., Comparison of soluble thrombomodulin, von Willebrand factor, tPA/PAI-1 complex, and high-sensitivity CRP concentrations in serum, EDTA plasma, citrated plasma, and acidiied citrated plasma (Stabilyte™) stored at −70°C for 8–11 years, Thromb. Res. 116, 249–254, 2005. 745. Hernestål-Boman, J., Jansson, J.H., Nilsson, T.K., et al., Long-term stability of ibrinolytic factors stored at −80° C, Thromb. Res. 125, 451–456, 2010. 746. Samarakoon, R., Goppelt-Strube, M., and Higgins, P.J., Linking cell structure to gene regulation: Signaling events and expression controls on the model genes PAI-1 and CTGF, Cell Signal. 22, 1413–1419, 2010. 747. Ma, Z., Paek, D., and Oh, C.K., Plasminogen activator inhibitor-1 and asthma: Role in the pathogenesis and regulation, Clin. Exp. Allergy 39, 1136–1144, 2009. 748. Ma, Z., Kwong, K.Y., Tovar, J.P., and Paek, D., Cyclic adenosine monophosphate induces plasminogen activator inhibitor-1 expression in human mass cells, Biochem. Biophys. Res. Commun. 400, 569–574, 2010. 749. Soeda, S., Koyanagi, S., Kuramoto, Y., et al., Anti-apoptotic roles of plasminogen activator inhibitor-1 as a neurotrophic factor in the central nervous system, Thromb. Haemost. 100, 1014–1020, 2008. 750. Ha, H., Oh, E.Y., and Lee, H.B., The role of plasminogen activator inhibitor-1 in renal and cardiovascular disease, Nat. Rev. Nephrol. 5, 203–211, 2009. 751. Kruithof, E.K., Regulation of plasminogen activator inhibitor 1 gene expression by inlammatory mediators and statins, Thromb. Haemost. 100, 969–975, 2008. 752. Sakamoto, K., Osaki, M., Hozumi, A., et al., Simvastatin suppresses dexamethasoneinduced secretion of plasminogen activator inhibitor-1 in human bone marrow adipocytes, BMC Musculoskelet. Disord. 12, 82, 2011.

Plasma Proteinase Inhibitors

361

753. Schmitt, M., Mengele, K., Napieralski, R., et al., Clinical evidence of level-of-evidence-1 disease forcast cancer biomarkers uPA and its inhibitor PAI-1, Exp. Rev. Med. Diagn. 10, 1051–1067, 2010. 754. Mengele, K., Napieralski, R., Magdolen, V., et al., Characteristics of the level-ofevidence-1 disease forcast cancer biomarkers uPA and its inhibitor PAI-1, Expert Rev. Mol. Diagn. 10, 947–962, 2010. 755. Kantelhardt, E.J., Vetter, M., Schmidt, M., et al., Prospective evaluation of prognostic factors uPA/PAI-1 in node-negative breast cancer: Phase III NNBC3-Europe trial (AGO,GBG, EORTC-PBG) comparing 6xFEC versus 3xFEC/3xDocetaxel, BMC Cancer 11, 140, 2011. 756. Malinowsky, K., Böllner, C., Hipp, S., et al., UPA and PAI-1 analysis from ixed tissues—New perspectives for a known set of predictive markers, Curr. Med. Chem. 17, 4370–4377, 2010. 757. Ulisse, S., Baldini, E., Sorrenti, S., and D’Armiento, M., The urokinase plasminogen activator system: A target for anti-cancer therapy, Curr. Cancer Drug Targets 9, 32–71, 2009. 758. Hillon, P., Guiu, B., Vincent, J., and Petit, J.M., Obesity, type 2 diabetes, and risk of digestive cancer, Gastroenterol. Clin. Biol. 34, 529–533, 2010. 759. van Kruijsdijk, R.C., van der Wall, E., and Visseren, F.L., Obesity and cancer: The role of dysfunctional adipose tissue, Cancer Epidermiol. Biomarkers Prev. 18, 2569–2578, 2009. 760. Matzuzawa, Y., Adiponectin: A key player in obesity related disorders, Curr. Pharm. Des. 16, 1896–1908, 2010. 761. van Zaane, B., Nur, E., and Squizzata, A., Systematic review on the effect of glucocorticoid use on procoagulant, anti-coagulant and ibrinolytic factors, J. Thromb. Haemost. 8, 2483–2493, 2010. 762. Shen, G.X., Oxidative stress and diabetic cardiovascular disorders: Role of mitochondria and NADPH oxidase, Can. J. Physiol. Pharmacol. 88, 241–248, 2010. 763. Cesari, M., Pahor, M., and Incalzi, R.A., Plasminogen activator inhibitor-1: A key factor linking ibrinolysis and age-related subclinical and clinical conditions, Cardiovasc. Ther. 28, e72–e91, 2010. 764. Canield, W.M. and Kisiel, W., Evidence of normal functional levels of activated protein C inhibitor in combined factor V/VIII deiciency disease, J. Clin. Invest. 70, 1260–1272, 1982. 765. Laurell, M., Christensson, A., Abrahamsson, P.-A., et al., Protein C inhibitor in human body luids. Seminal plasma is rich in inhibitor antigen deriving from cells throughout the male reproductive system, J. Clin. Invest. 89, 1094–1101, 1992. 766. Laurell, M., Stenlo, J., and Carlson, T.H., Turnover of *I-protein C inhibitor and *I-α1antitrypsin and their complexes with activated protein C, Blood 76, 2290–2296, 1990. 767. Suzuki, K., Nishioka, J., Kusumoto, H., and Hashimoto, S., Mechanism of inhibition of activated protein C by protein C inhibitors, J. Biochem. 95, 187–195, 1984. 768. Cooper, S.T. and Church, F.C., PCI: Protein C inhibitor?, Adv. Exp. Med. Biol. 425, 45–54, 1997. 769. España, F., Berrettini, M., and Grifin, J.H., Puriication and characterization of plasma protein C inhibitor, Thromb. Res. 55, 369–384, 1989. 770. Pratt, C.W., Macik, B.G., and Church, F.C., Protein C inhibitors: Puriication and proteinase reactivity, Thromb. Res. 53, 595–603, 1989. 771. Stief, T.W., Radtke, K.P., and Heimburger, N., Inhibition of urokinase by protein C-inhibitor (PCI). Evidence for identity of PCI and plasminogen activator inhibitor-3, Biol. Chem. Hoppe Seyler 368, 1427–1433, 1987. 772. Suzuki, K., The multi-functional serpin, protein C inhibitor: Beyond thrombosis and hemostasis, J. Thromb. Haemost. 6, 2017–2026, 2008.

362

Biotechnology of Plasma Proteins

773. Meijers, J.C.M. and Herwald, H., Protein C inhibitor, Semin. Thromb. Hemost. 37, 149– 154, 2011. 774. Bijsmans, I.T., Smits, K.M., de Graeff, P., et al., Loss of serpinA5 protein expression is associated with advanced-stage serous ovarian tumors, Mod. Pathol. 24, 463–470, 2011. 775. España, F., Sanchez-Curenca, J., Vera, C.D., et al., A quantitative ELISA for the measurement for the measurement of complexes of prostate-speciic antigen with protein C inhibitor when using a puriied standard, J. Lab. Clin. Med. 122, 711–719, 1993. 776. Uhrin, P., Dewerchin, M., Hilpert, M., et al., Disruption of the protein C inhibitor gene results in impaired spermatogenesis and male infertility, J. Clin. Invest. 106, 1531–1539, 2000. 777. Uhrin, P., Schöfer, C., Zaujec, J., et al., Male fertility and protein C inhibitor/plasminogen activator inhibitor-3 (PCI): Localization of PCI in the mouse testis and failure of single plasminogen activator knockout to restore spermatogenesis in PCI-deicient mice, Fertil. Steril. 88, 1049–1057, 2007. 778. Medcalf, R.L. and Stasinopoulos, S.J., The undecided serpin. The ins and outs of plasminogen activator inhibitor type 2, FEBS J. 272, 4858–4867, 2005. 779. Lobov, S. and Ranson, M., Molecular competition between plasminogen activator inhibitors type 1 and 2 for urokinase: Implications for cellular proteolysis and adhesion in cancer, Cancer Lett. 303, 118–127, 2011. 780. Wang, Y.X., Zhao, L., Nagashima, M., et al., A novel inhibitor of activated thrombinactivatable ibrinolysis inhibitor (TAFIa)—Part I: Pharmacological characterization, Thromb. Haemost. 97, 45–53, 2007. 781. Bunnage, M.E., Blagg, J., Steele, J., et al., Discovery of potent & selective inhibitors of activated thrombin-activatable ibrinolysis inhibitor for the treatment of thrombosis, J. Med. Chem. 50, 6095–6103, 2007. 782. Owen, D.R., Bull, D.J., Bunnage, M.E., et al., Oxygenated analogues of UK-396082 as inhibitors of activated thrombin activatable ibrinolysis inhibitor, Bioorg. Med. Chem. Lett. 20, 92–96, 2010. 783. LeWitt, P.A., Rezai, A.R., Leehey, M.A., et al., AAV2-GAD gene therapy for advanced Parkinson’s disease: A double-blind, sham-surgery controlled, randomized trial, Lancet Neurol. 10, 309–319, 2011. 784. Edelman, G.M. and Gally, J.A., Degeneracy and complexity in biological systems, Proc. Natl. Acad. Sci. USA 98, 13763–13768, 2001. 785. O’Reilly, M.S., Antiangiogenicantithrombin, Semin. Thromb. Hemost. 33, 660–666, 2007. 786. Khor, B. and Van Cott, E.M., Laboratory tests for antithromin deiciency, Am. J. Hematol. 85, 947–950, 2010. 787. Tuder, R.M., Janciauskiene, S.M., and Petrache, I., Lung disease associated with α1-antitrypsin deiciency, Proc. Am. Thorac. Soc. 7, 381–386, 2010. 788. Bouchecareilh, M., Conkright, J.J., and Balch, W.E., Proteostasis strategies for restoring α1-antitrypsin deiciency, Proc. Am. Thorac. Soci. 7, 415–422, 2010. 789. Thelwell, C., Marszal, E., Rigsby, P., and Longstaff, C., An international collaborative study to establish the WHO 1st international standard for alpha-1-antitrypsin, Vox Sang. 101, 83–89, 2011. 790. Flotte, T.R., Trapnell, B.C., Humphries, M., et al., Phase 2 clinical trial of a recombinant adeno-associated virus vector expressing alpha-1-antitrypsin: Interim results, Hum. Gene Ther. 22, 1239–1247, 2011. 791. Grimstein, C., Choi, Y.K., Wasserfall, C.H., et al., Alpha-1-antitrypsin protein and gene therapies decrease autoimmunity and delay arthritis development in mouse model, J. Transl. Med. 9, 21, 2011.

Plasma Proteinase Inhibitors

363

792. Blanchard, V., Liu, X., Eigel, S., et al., N-Glycosylation and biological activity of recombinant human α1-antitrypsin expressed in a novel human neuronal cell line, Biotechnol. Bioeng. 108, 2118–2128, 2011. 793. Janciauskiene, S., Nita, I., Subramaniyam, D., et al., α1-Antitrypsin inhibits the activity of the matriptase catalytic domain in vitro, Am. J. Resp. Cell Mol. Biol. 39, 631–637, 2008. 794. Feldman, S.R., Ney, K.A., Gonias, S.L., and Pizzo, S.L., In vitro binding and in vivo clearance of human α2-macroglobulin after reaction with endoproteases from four different classes, Biochem. Biophys. Res. Commun. 114, 757–762, 1983. 795. Kluthe, R., Kleine, N., and Hagemann, U., Bestimmung des α-2-Makroglobulinumsatzes beim Menschen, Experientia 22, 385–386, 1966. 796. Balajthy, Z., Synthesis and evaluation of macromolecule-bound derivatives of a peptidyl1-β-D-arabinofuranosylcytosine prodrug, Drug Metabol. Lett. 2, 83–89, 2008. 797. Blatrix, C., Amouch, P., Drouet, J., et al., Plasmatic elimination of the α2-macroglobulin– proteinase complexes, Pathologie Biologie 21(Suppl.), 11–14, 1973. 798. Cáceres, L.C., Bonacci, G.R., Sánchez, M.C., et al., Activated α2-macroglobulin induces matrix metalloproteinase 9 expression by low-density lipoprotein receptor-related protein 1 through MAPK-ERK1/2 and NF-κB activation in macrophage-derived cell lines, J. Cell Biochem. 111, 607–617, 2010. 799. Moroi, M. and Aoki, N., Isolation and characterization of α2-plasmin inhibitor from human plasma. A novel proteinase inhibitor which inhibits activator-induced clot lysis, J. Biol. Chem. 251, 5956–5965, 1976. 800. Collen, D. and Lijnen, H.R., Molecular basis of ibrinolysis, as relevant for thrombolytic therapy, Thromb. Haemost. 74, 167–171, 1995. 801. Coughlin, P.B., Antiplasmin: The forgotten serpin?, FEBS J. 272, 4852–4857, 2005. 802. Matsuno, H., α2-Antiplasmin on cardiovascular diseases, Curr. Pharm. Des. 12, 841– 847, 2006. 803. Sarilla, S., Habib, S.Y., Kravtsov, D.V., et al., Sucrose octasulfate selectively accelerates thrombin inactivation by heparin cofactor II, J. Biol. Chem. 285, 8278–8289, 2010. 804. Langford-Smith, K.J., Mercer, J., Petty, J., et al., Heparin cofactor II–thrombin complex and dermatansulphate: Chondroitin sulphate ratio are markers of short and long-term treatment effects in mucopolysacchride diseases, J. Inherit. Metab. Dis. 34, 499–508, 2011. 805. Rau, J.C., Mitchell, J.W., Fortenberry, Y.M., and Church, F.C., Heparin cofactor II: Discovery, properties, and role in controlling vascular homeostasis, Semin. Thromb. Hemost. 37, 339–348, 2011. 806. Singer, M. and Jones, A.M., Bench-to-bedside review: The role of C1-esterase inhibitor in sepsis and other critical illnesses, Crit. Care 15, 203, 2011. 807. Bernstein, J.A., Ritchie, B., Levy, R.J., et al., Population pharmacokinetics of plasmaderived C1 esterase inhibitor concentrate used to treat acute hereditary angioedema attacks, Ann. Allergy Asthma Immunol. 105, 149–154, 2010. 808. Brown, T., Cicardi, M., Farkas, H., et al., International consensus algorithm for the diagnosis, therapy and management of hereditary angioedema, Allergy Asthma Clin. Immunol. 6, 24, 2010. 809. Zeerleder, S., C1-inhibitor: More than a serine protease inhibitor, Semin. Thromb. Hemost. 37, 362–374, 2011. 810. Drouet, C., Bouillet, L., Csopaki, F., and Colomb, M.G., Hepatitis C virus NS3 serine protease interacts with the serpin C1 inhibitor, FEBS Lett. 458, 415–418, 1999. 811. Boncela, J., Przygodzka, P., Papiewska-Pajak, I., et al., Plasminogen activator inhibitor type 1 interacts with α3 subunit of proteosome and modulates its activity, J. Biol. Chem. 286, 6820–6831, 2011.

364

Biotechnology of Plasma Proteins

812. Güngör, Y., Kayatas, M., Yildiz, G., et al., The presence of PAI-1 4G/5G and ACE DD genotypes increases the risk of early-stage AVF thrombosis in hemodialysis patients, Ren. Fail. 33, 169–175, 2011. 813. D’Elia, A.V., Fabbor, D., Driul, L., et al., Plasminogen activator inhibitor-1 gene polymorphisms in pre-eclapsia, Semin. Thromb. Hemost. 37, 97–105, 2011. 814. Cho, S., Kang, J., Lyttle, C., et al., Association of elevated plasminogen activator inhibitor 1 levels with diminished lung function in patients with asthma, Ann. Allergy Asthma Immunol. 106, 371–377, 2011. 815. Mazouni, C., Bonnier, P., Romain, S., and Martin, B.M., A nomogram predicting the probability of primary breast cancer survival at 2- and 5-years using pathological and biological tumor parameters, J. Surg. Oncol. 103, 746–750, 2011. 816. Malmström, E., Mörgelin, M., Malmsten, M., et al., Protein C inhibitor—A novel antimicrobial agent, PLoS Pathog. 5, e1000698, 2009. 817. Fortenberry, Y.M., Brandal, S., Bialas, R.C., and Church, F.C., Protein C inhibitor regulates both cathepsin L activity and cell-mediated tumor cell migration, Biochim. Biophys. Acta 1800, 580–590, 2010. 818. Suzuki, K., Hepatocyte growth factor activator (HGFA): Its regulation by protein C inhibitor, FEBS J. 277, 2223–2229, 2010. 819. Bijsmans, I.T., Smits, K.M., de Graeff, P., et al., Loss of serpinA5 protein expression is associated with advanced-stage serous ovarian tumors, Mod. Pathol. 24, 463–470, 2011. 820. Radtke, K.P., Wenz, K.H., and Heimburger, N., Isolation of plasminogen activator inhibitor-2 (PAI-2) from human placenta. Evidence for vitronectin/PAI-2 complexes in human placenta extract, Biol. Chem. Hoppe Seyler 371, 1119–1127, 1990. 821. Scherrer, A., Kruithof, E.K., and Grob, J.P., Plasminogen activator inhibitor-2 in patients with monocytic leukemia, Leukemia 5, 479–486, 1991. 822. Jensen, P.H., Lorand, L., Ebbesen, P., and Gliemann, J., Type-2 plasminogenactivator inhibitor is a substrate for trophoblast transglutaminase and Factor XIII. Transglutaminase-catalyzed cross-linking to cellular and extracellular structures, Eur. J. Biochem. 214, 141–146, 1993. 823. Kruithof, E.K.O., Baker, M.S., and Bunn, C.L., Biological and clinical aspects of plasminogen activator inhibitor type 2, Blood 86, 4007–4024, 1995. 824. Astedt, B., Lindoff, C., and Lecander, I., Signiicance of the plasminogen activator inhibitor of placental type (PAI-2) in pregnancy, Semin. Thromb. Hemost. 24, 431–435, 1998. 825. Robbie, L.A., Dummer, S., Booth, N.A., et al., Plasminogen activator inhibitor 2 and urokinase-type plasminogen activator in plasma and leukocytes in patients with severe sepsis, Br. J. Haematol. 109, 342–348, 2000. 826. Hellgren, M., Hemostasis during normal pregnancy and puerperium, Semin. Thromb. Hemost. 29, 125–130, 2003. 827. Syrjänen, S., Naud, P., Sarian, L., et al., Up-regulation of plasminogen activator inhibitor-2 is associated with high-risk HPV and grade of cervical lesion at baseline but does not predict outcome of high-risk HPV infections or incident CIN, Am. J. Clin. Pathol. 132, 883–892, 2009. 828. Wang, W., Hendriks, D.F., and Sharpé, S.S., Carboxypeptidase U, a plasma carboxypeptidase with high afinity for plasminogen, J. Biol. Chem. 269, 15937–15944, 1994. 829. Foley, J.H., Cook, P.F., and Nesheim, M.E., Kinetics of activated thrombin-activatable ibrinolysis inhibitor (TAFIa)-catalyzed cleavage of C-terminal lysine residues of ibrin degradation products and removal of plasminogen-binding sites, J. Biol. Chem. 286, 19280–19286, 2011.

Plasma Proteinase Inhibitors

365

830. Nesheim, M., Wang, W., Boffa, M., et al., Thrombin, thrombomodulin and TAFI in the molecular link between coagulation and ibrinolysis, Thromb. Haemost. 78, 386–391, 1997. 831. Heylen, E., Van Goethem, S., Willemse, J. et al., Development of a sensitive and selective assay for the determination of procarboxypeptidase U (thrombin-activatable ibrinolysis inhibitor) in plasma, Anal. Biochem. 396, 152–154, 2010. 832. Julian-Vague, I., Morange, P.E., Aubert, H., et al., Plasma thrombin-activatable ibrinolysis inhibitor antigen concentration and genotype in relation to myocardial infarction in the North and South of Europe, Arteriocler. Thromb. Vasc. Biol. 22, 867–873, 2002. 833. Willemse, J.L., Heylen, E., Nesheim, M.E., and Hendriks, D.F., Carboxypeptidase U (TAFIa): A new drug target for ibrinolytic therapy?, J. Thromb. Haemost. 7, 1962– 1971, 2009.

8

Vitamin K–Dependent Proteins

The vitamin K–dependent proteins are characterized by (1) their dependence on vitamin K for the synthesis of a mature, functional protein and (2) the presence of γ-carboxyglutamic acid (GLA) in the amino-terminal portion of the mature protein. The vitamin K–dependent proteins, with the exception of prothrombin, are modest, globular proteins with molecular weights between 50 and 70 kDa and are present in the plasma at low concentrations. A list of the characterized vitamin K– dependent proteins of biotechnological interest is presented in Table 8.1. In addition to the plasma proteins, γ-carboxylation occurs in the proteins found in the bones and cartilage.1–3 There has been recent interest in the role of Vitamin K in hard tissue function4–7 with an emphasis on osteoporosis,8,9 which may be associated with γ-carboxylation.9 As Vermeer and Theuwissen note, inadequate dietary Vitamin K is a modiiable risk factor for osteoporosis. The synthesis of the vitamin K–dependent proteins, as the name suggests, requires the presence of vitamin K, originally described by Henrik Dam in 1935.10 At the same time, dicoumarol was identiied by Karl Paul Link, at the University of Wisconsin, as the hemorrhagic agent in spoiled sweet clover. The story of the discovery of dicoumarol and the subsequent development of warfarin as an anticoagulant was provided by Link in 1959.11 Warfarin derives its name from the Wisconsin Alumni Research Foundation. Warfarin and a number of its derivatives are still widely used, despite the advent of several new direct enzyme inhibitors of thrombin and factor Xa.12 These drugs are discussed in the following text in greater detail and may impact the use of warfarin. Warfarin derivatives have a long record of use and physicians are mostly comfortable with their use. Vitamin K antagonists require continued laboratory monitoring and are sensitive to diet. Genetic polymorphism (warfarin resistance) can also result in a variable response to warfarin in the target population,13–15 suggesting that genotyping might be useful in determining a patient’s suitability for warfarin therapy.16 Proteins induced by the absence of vitamin K (PIVKA) are the uncarboxylated forms of the vitamin K–dependent proteins, which are found in the circulation17 in the presence of the vitamin K antagonist or in the absence of the dietary vitamin K, and were observed to be inhibitors of coagulation.18,19 More recently, PIVKA-II (uncarboxylated prothrombin) has been used as a biomarker for liver function.20–23 PIVKA has also been used as a biomarker for hard tissue disorders.24–26 The use of PIVKA-II has been suggested as an alternative marker for vitamin K antagonist

367

368

Biotechnology of Plasma Proteins

TABLE 8.1 Vitamin K–Dependent Plasma Proteins Protein Factor II (Prothrombin) Factor VII Factor IX (Christmas factor) Factor X (Stuart–Prower factor) Protein C Protein S Protein Z

Plasma Concentration

Molecular Weight (kDa)b

Mg/L

nM

Half-Life

72

100–150

1400–2000

2–3 days

50 57

0.5 4.5–5

10 80–90

3–6 h 18–24 h

57

8–10

140–170

30–40 h

62 69 62

4–5 21–25 2.2

60–80 300–360 35

6h 40 h