Semin Thromb Hemost 2011; 37(3): 322-327
DOI: 10.1055/s-0031-1274515
© Thieme Medical Publishers

Low Molecular Weight Heparins Differ Substantially: Impact on Developing Biosimilar Drugs

Jeanine M. Walenga1 , Craig M. Jackson2 , Craig M. Kessler3
  • 1Loyola University Medical Center, Maywood, Illinois
  • 2Coagulation Diagnostics, San Diego, California
  • 3Georgetown University Medical Center, Washington, DC
Further Information

Publication History

Publication Date:
31 March 2011 (online)

ABSTRACT

Generic drugs are an important component for meaningful health-care reform currently being debated in the United States. Aside from defining the period of drug exclusivity, however, there is a critical need to ensure that generics of biologic medicines (biosimilars) are safe and effective. For low molecular weight heparins (LMWHs), the standard of care for management of venous thromboembolism, their complex structure and polypharmacological actions make producing a generic LMWH more challenging than a generic small molecule medicine. Because biosimilar LMWHs will be used interchangeably with their branded product, inherent variability between products could lead to important differences in potency, safety, or effectiveness, including unanticipated immune responses. Awareness of the specific problems associated with biosimilar LMWH development led to new recommendations from several expert bodies. This article discusses the implications of these differences for the production of biosimilar LMWHs and provides recommendations to address the limitations in the pending U.S. Congress legislation, a well-intentioned undertaking but one that must preserve the health and welfare of citizens who require these critical care medications.

REFERENCES

  • 1 Engelberg A B, Kesselheim A S, Avorn J. Balancing innovation, access, and profits—market exclusivity for biologics.  N Engl J Med. 2009;  361 (20) 1917-1919
  • 2 Casadevall N, Nataf J, Viron B et al.. Pure red-cell aplasia and antierythropoietin antibodies in patients treated with recombinant erythropoietin.  N Engl J Med. 2002;  346 (7) 469-475
  • 3 Rosendaal F R, Nieuwenhuis H K, van den Berg H M Dutch Hemophilia Study Group et al. A sudden increase in factor VIII inhibitor development in multitransfused hemophilia A patients in The Netherlands.  Blood. 1993;  81 (8) 2180-2186
  • 4 Li J L, Yang C, Xia Y et al.. Thrombocytopenia caused by the development of antibodies to thrombopoietin.  Blood. 2001;  98 (12) 3241-3248
  • 5 Harenberg J, Kakkar A, Bergqvist D Subcommittee on Control of Anticoagulation of the SSC of the ISTH et al. Recommendations on biosimilar low-molecular-weight heparins.  J Thromb Haemost. 2009;  7 (7) 1222-1225
  • 6 European Medicines Agency .Guideline on non-clinical and clinical development of similar biological medicinal products containing low-molecular-weight-heparins. European Medicines Agency, London, March 19, 2009, EMEA/CHMP/BMWP/118264/2007, 2007. Available at: http://www.emea.europa.eu/pdfs/human/biosimilar/11826407enfin.pdf Accessed September 6, 2009
  • 7 Casu B, Torri G. Structural characterization of low molecular weight heparins.  Semin Thromb Hemost. 1999;  25 (Suppl 3) 17-25
  • 8 Jeske W P, Walenga J M, Hoppensteadt D A et al.. Differentiating low-molecular-weight heparins based on chemical, biological, and pharmacologic properties: implications for the development of generic versions of low-molecular-weight heparins.  Semin Thromb Hemost. 2008;  34 (1) 74-85
  • 9 Jeske W P, Walenga J M, Fareed J. Differentiating between the low-molecular-weight heparins used for venous thromboembolism treatment and prophylaxis.  Thrombosis Clinic. 2008;  2 8-21
  • 10 Nicolau J C, Cohen M, Montalescot G. Differences among low-molecular-weight heparins: evidence in patients with acute coronary syndromes.  J Cardiovasc Pharmacol. 2009;  53 (6) 440-445
  • 11 Fareed J, Jeske W, Hoppensteadt D, Clarizio R, Walenga J M. Are the available low-molecular-weight heparin preparations the same?.  Semin Thromb Hemost. 1996;  22 (Suppl 1) 77-91
  • 12 Oosta G M, Gardner W T, Beeler D L, Rosenberg R D. Multiple functional domains of the heparin molecule.  Proc Natl Acad Sci U S A. 1981;  78 (2) 829-833
  • 13 Lindahl U, Thunberg L, Bäckström G, Riesenfeld J, Nordling K, Björk I. Extension and structural variability of the antithrombin-binding sequence in heparin.  J Biol Chem. 1984;  259 (20) 12368-12376
  • 14 Ofosu F A, Danishefsky I, Hirsh J. Heparin and related polysaccharides. Structure and activities.  Ann N Y Acad Sci. 1989;  556 1-501
  • 15 Rosenberg R D, Bauer K A. The heparin-antithrombin system: a natural anticoagulant mechanism. In: Colman R W, Hirsh J, Marder V J, et al, eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 3rd ed. Philadelphia, PA: JB Lippincott; 1994: 837-860
  • 16 Tollefsen D M, Majerus D W, Blank M K. Heparin cofactor II. Purification and properties of a heparin-dependent inhibitor of thrombin in human plasma.  J Biol Chem. 1982;  257 (5) 2162-2169
  • 17 Tobu M, Ma Q, Iqbal O et al.. Comparative tissue factor pathway inhibitor release potential of heparins.  Clin Appl Thromb Hemost. 2005;  11 (1) 37-47
  • 18 Friedel H A, Balfour J A. Tinzaparin. A review of its pharmacology and clinical potential in the prevention and treatment of thromboembolic disorders.  Drugs. 1994;  48 (4) 638-660
  • 19 Montalescot G, Collet J P, Lison L et al.. Effects of various anticoagulant treatments on von Willebrand factor release in unstable angina.  J Am Coll Cardiol. 2000;  36 (1) 110-114
  • 20 Salzman E W, Rosenberg R D, Smith M H, Lindon J N, Favreau L. Effect of heparin and heparin fractions on platelet aggregation.  J Clin Invest. 1980;  65 (1) 64-73
  • 21 Eika C. Inhibition of thrombin induced aggregation of human platelets by heparin.  Scand J Haematol. 1971;  8 (3) 216-222
  • 22 Guerrini M, Guglieri S, Casu B et al.. Antithrombin-binding octasaccharides and role of extensions of the active pentasaccharide sequence in the specificity and strength of interaction. Evidence for very high affinity induced by an unusual glucuronic acid residue.  J Biol Chem. 2008;  283 (39) 26662-26675
  • 23 Capila I, Linhardt R J. Heparin-protein interactions.  Angew Chem Int Ed Engl. 2002;  41 (3) 391-412
  • 24 Houbouyan L, Padilla A, Gray E, Longstaff C, Barrowcliffe T W. Inhibition of thrombin generation by heparin and LMW heparins: a comparison of chromogenic and clotting methods.  Blood Coagul Fibrinolysis. 1996;  7 (1) 24-30
  • 25 Hirsh J, Raschke R. Heparin and low-molecular-weight heparin: the Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy.  Chest. 2004;  126 (3, Suppl) 188S-203S
  • 26 Collignon F, Frydman A, Caplain H et al.. Comparison of the pharmacokinetic profiles of three low molecular mass heparins—dalteparin, enoxaparin and nadroparin—administered subcutaneously in healthy volunteers (doses for prevention of thromboembolism).  Thromb Haemost. 1995;  73 (4) 630-640
  • 27 Eriksson B I, Söderberg K, Widlund L, Wandeli B, Tengborn L, Risberg B. A comparative study of three low-molecular weight heparins (LMWH) and unfractionated heparin (UH) in healthy volunteers.  Thromb Haemost. 1995;  73 (3) 398-401
  • 28 Andrassy K, Eschenfelder V. Are the pharmacokinetic parameters of low molecular weight heparins predictive of their clinical efficacy?.  Thromb Res. 1996;  81 (2, Suppl) S29-S38
  • 29 Brieger D, Dawes J. Production method affects the pharmacokinetic and ex vivo biological properties of low molecular weight heparins.  Thromb Haemost. 1997;  77 (2) 317-322
  • 30 Nugent M A. Heparin sequencing brings structure to the function of complex oligosaccharides.  Proc Natl Acad Sci U S A. 2000;  97 (19) 10301-10303
  • 31 Turnbull J, Powell A, Guimond S. Heparan sulfate: decoding a dynamic multifunctional cell regulator.  Trends Cell Biol. 2001;  11 (2) 75-82
  • 32 Oelschläger C, Römisch J, Staubitz A et al.. Antithrombin III inhibits nuclear factor kappaB activation in human monocytes and vascular endothelial cells.  Blood. 2002;  99 (11) 4015-4020
  • 33 Elsayed E, Becker R C. The impact of heparin compounds on cellular inflammatory responses: a construct for future investigation and pharmaceutical development.  J Thromb Thrombolysis. 2003;  15 (1) 11-18
  • 34 Rao N V, Kennedy T P, Rao G, Ky N, Hoidal J R. Sulfated polysaccharides prevent human leukocyte elastase-induced acute lung injury and emphysema in hamsters.  Am Rev Respir Dis. 1990;  142 (2) 407-412
  • 35 Thourani V H, Brarr S S, Kennedy T P et al.. Nonanticoagulant heparin inhibits NF-κB activation and attenuates myocardial reperfusion injury.  Am J Physiol Heart Circ Physiol. 2000;  278 (Suppl H) 2084-2093
  • 36 Weiler J M, Edens R E, Linhardt R J, Kapelanski D P. Heparin and modified heparin inhibit complement activation in vivo.  J Immunol. 1992;  148 (10) 3210-3215
  • 37 Geiben-Lynn R, Brown N, Walker B D, Luster A D. Purification of a modified form of bovine antithrombin III as an HIV-1 CD8 + T-cell antiviral factor.  J Biol Chem. 2002;  277 (44) 42352-42357
  • 38 Ashikari-Hada S, Habuchi H, Kariya Y, Kimata K. Heparin regulates vascular endothelial growth factor 165-dependent mitogenic activity, tube formation, and its receptor phosphorylation of human endothelial cells. Comparison of the effects of heparin and modified heparins.  J Biol Chem. 2005;  280 (36) 31508-31515
  • 39 Ono K, Hattori H, Takeshita S, Kurita A, Ishihara M. Structural features in heparin that interact with VEGF165 and modulate its biological activity.  Glycobiology. 1999;  9 (7) 705-711
  • 40 Wang L, Brown J R, Varki A, Esko J D. Heparin's anti-inflammatory effects require glucosamine 6-O-sulfation and are mediated by blockade of L- and P-selectins.  J Clin Invest. 2002;  110 (1) 127-136
  • 41 Wei M, Tai G, Gao Y et al.. Modified heparin inhibits P-selectin-mediated cell adhesion of human colon carcinoma cells to immobilized platelets under dynamic flow conditions.  J Biol Chem. 2004;  279 (28) 29202-29210
  • 42 O'Reilly M S, Pirie-Shepherd S, Lane W S, Folkman J. Antiangiogenic activity of the cleaved conformation of the serpin antithrombin.  Science. 1999;  285 (5435) 1926-1928
  • 43 Clowes A W, Karnowsky M J. Suppression by heparin of smooth muscle cell proliferation in injured arteries.  Nature. 1977;  265 (5595) 625-626
  • 44 Castellot Jr J J, Favreau L V, Karnovsky M J, Rosenberg R D. Inhibition of vascular smooth muscle cell growth by endothelial cell-derived heparin. Possible role of a platelet endoglycosidase.  J Biol Chem. 1982;  257 (19) 11256-11260
  • 45 Gorelik E, Bere W W, Herberman R B. Role of NK cells in the antimetastatic effect of anticoagulant drugs.  Int J Cancer. 1984;  33 (1) 87-94
  • 46 Zhang W, Swanson R, Izaguirre G, Xiong Y, Lau L F, Olson S T. The heparin-binding site of antithrombin is crucial for antiangiogenic activity.  Blood. 2005;  106 (5) 1621-1628
  • 47 Ekman-Ordeberg G, Hellgren M, Akerud A et al.. Low molecular weight heparin stimulates myometrial contractility and cervical remodeling in vitro.  Acta Obstet Gynecol Scand. 2009;  88 (9) 984-989
  • 48 Nikkila E. Studies on the lipid-protein relationship in normal and pathological sera and the effect of heparin on serum lipoproteins.  Scand J Clin Lab Invest. 1953;  5 (Suppl 8) 9-100
  • 49 Warkentin T E, Greinacher A, Koster A, Lincoff A M. American College of Chest Physicians . Treatment and prevention of heparin-induced thrombocytopenia: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th edition).  Chest. 2008;  133 (6, Suppl) 340S-380S
  • 50 Walenga J M, Koza M J, Lewis B E, Pifarré R. Relative heparin-induced thrombocytopenic potential of low molecular weight heparins and new antithrombotic agents.  Clin Appl Thromb Hemost. 1996;  2 (Suppl 1) 21-27
  • 51 Walenga J M, Jeske W P, Prechel M M, Bacher P, Bakhos M. Decreased prevalence of heparin-induced thrombocytopenia with low-molecular-weight heparin and related drugs.  Semin Thromb Hemost. 2004;  30 (Suppl 1) 69-80
  • 52 Greinacher A, Alban S, Dummel V, Franz G, Mueller-Eckhardt C. Characterization of the structural requirements for a carbohydrate based anticoagulant with a reduced risk of inducing the immunological type of heparin-associated thrombocytopenia.  Thromb Haemost. 1995;  74 (3) 886-892
  • 53 Ahmad S, Haas S, Hoppensteadt D A et al.. Differential effects of clivarin and heparin in patients undergoing hip and knee surgery for the generation of anti-heparin-platelet factor 4 antibodies.  Thromb Res. 2002;  108 (1) 49-55
  • 54 Untch B, Ahmad S, Jeske W P et al.. Prevalence, isotype, and functionality of antiheparin-platelet factor 4 antibodies in patients treated with heparin and clinically suspected for heparin-induced thrombocytopenia. The pathogenic role of IgG.  Thromb Res. 2002;  105 (2) 117-123
  • 55 Walenga J M, Adiguzel C, Iqbal O, Hoppensteadt D, Cunanan J, Bakhos M. Fibrin clot formation differs in the presence of branded and generic enoxaparins.  J Thromb Haemost. 2009;  7 (Suppl 2) Abstract PP-WE-387
  • 56 Adiguzel C, Litinas E, Cunanan J, Hoppensteadt D, Walenga J M, Fareed J. Differential thrombin generation inhibition by branded and generic low molecular weight heparins (LMWHs) as studied by using fluorescence substrate based kinetic method.  J Thromb Haemost. 2009;  7 (Suppl 2) Abstract PP-MO-139
  • 57 Walenga J M, Hoppensteadt D, Cunanan J, Jeske W P, Adiguzel C, Bakhos M. Immunogenicity of low molecular weight heparins and biosimilars.  J Thromb Haemost. 2009;  7 (Suppl 2) Abstract PP-MO-399
  • 58 Kishimoto T K, Viswanathan K, Ganguly T et al.. Contaminated heparin associated with adverse clinical events and activation of the contact system.  N Engl J Med. 2008;  358 (23) 2457-2467

Jeanine M WalengaPh.D. 

Professor, Thoracic & Cardiovascular Surgery and Pathology, Cardiovascular Institute, Building 110, Room 5226, Loyola University Medical Center

2160 S. First Avenue, Maywood, IL 60153

Email: jwaleng@lumc.edu

    >