throbber
SEMINARS IN THROMBOSIS AND HEMOSTASIS-VOL. 25, SUPPL. 3, 1999
`
`Production and Chemical Processing of
`Low Molecular Weight Heparins
`
`ROBERT J. LINHARDT, PH.D. AND NUR SIBEL GUNAY, M.S.
`
`ABSTRACT Heparin is an animal tissue extract
`that is widely used as an anticoagulant drug. A num(cid:173)
`ber of low molecular weight heparins (LMWHs), in(cid:173)
`troduced in the past decade, are beginning to displace
`pharmaceutical (or compendial) grade heparins as
`clinical antithrombotic agents. This article desocibes
`the chemical properties of the glycosaminoglycan
`(GAG) heparin and how it is prepared and processed
`into pharmaceutical grade heparin. There are several
`commercially produced LMWHs that are prepared
`through the controlled depolymerization of pharma(cid:173)
`ceutical grade heparin. The chemistry of the com(cid:173)
`mercial processes used for manufacturing LMWHs is
`discussed. Structural differences are found in the
`LMWHs prepared using different commercial
`processes. Careful control of process variables has
`generally resulted in the reproducible preparation of
`LMWHs that are structurally uniform and of high
`quality. The specifications, however, remain different
`for each LMWH. Thus, LMWHs are a group of simi(cid:173)
`lar but different drug agents. As the structural prop(cid:173)
`erties of LMWHs vary significantly, the bio-equiva•
`lence or
`inequivalence of
`these agents must
`ultimately be established by the pharmacologists and
`the clinicians.
`
`Keywords: Low molecular weight heparin,
`analysis, structure, process, production
`
`Heparin, a clinical anticoagulant, has been one of
`the most effective and widely used drugs of this
`century. 1•2 As one of the oldest drugs currently stN.l in
`widespread clinical use, heparin is unique as it is among
`the first biopolymeric drugs and one of only a few carbo-
`
`Department of Medicinal and Natural Products Chemistry, University
`of Iowa, College of Pharmacy, Iowa City, Iowa.
`Reprint requests: Dr. Linhardt, Professor of Pharmacy and Chem(cid:173)
`ical Engineering, University of Iowa, College of Pharmacy, PHAR
`303A, University of Iowa, Iowa City, IA 52242.
`
`hydrate drugs. Indeed, heparin's introduction predates
`the establishment of the United States Food and Drug
`Administration. 2 Low molecular weight heparins
`(LMWHs ), also referred to as low molecular mass he(cid:173)
`parins (LMMHs ), are a group of heparin-derived antico(cid:173)
`agulant/antithrombotic agents that began their develop(cid:173)
`ment during the last quarter of this century. 3-7
`The introduction of LMWHs primarily resulted
`from an improved understanding of the molecular basis
`of the biochemistry associated with the coagulation cas(cid:173)
`cade. 8-10 The isolation of the serine protease inhibitor,
`antithrombin III (AT), and the characterization of coagu(cid:173)
`lation factors (serine proteases), such as thrombin and
`factor Xa (inhibited by AT), were critical in driving the
`development of LMWHsJ,8 Heparin accelerates the in(cid:173)
`hibition of these coagulation factors by AT, preventing
`the generation of a fibrin clot. In the coagulation cas(cid:173)
`cade, one factor activates the next until prothrombin
`(factor II) is converted to thrombin (factor Ila) by factor
`Xa. It is thrombin that acts on fibrinogen to form a fibrin
`clot.8 The very nature of this cascade suggested a thera(cid:173)
`peutic opportunity to develop an agent that was more
`specific than heparin (which acts at many points in the
`cascade) and that might provide more subtle regulation
`of coagulation and reduce the major hemorrhagic side
`effects associated with heparin. LMWHs were originally
`developed based on this rationale. Various laboratories
`observed that when heparin is fractionated based on size
`or broken down chemically or enzymatically, its activity
`against thrombin is decreased to a much greater extent
`than its activity against factor Xa. 11 - 15 The separation of
`activities result from differences in their molecular re(cid:173)
`quirements for inhibition. Factor Xa interacts directly
`with AT bound to a specific pentasaccharide sequence in
`heparin (the AT binding site) and requires a short heparin
`chain comprised only of these saccharide units for its in(cid:173)
`activation.5 In contrast, thrombin must also bind adja(cid:173)
`cent to AT on a flanking sequence in heparin, thus re(cid:173)
`quiring a longer heparin chain with 18 or more
`saccharide units for its inactivation.3,8 As factor Xa lies
`at the convergence of the extrinsic and intrinsic path(cid:173)
`ways of the coagulation cascade, it was speculated that a
`
`Copyright ©1999 by Thieme Medical Publishers, Inc. 333 Seventh Avenue, New York, NY 10001. Tel: + 1(212) 760-0888, ext. 132.
`0094-6176/1999/E1098-9064(1999)25:S3:0005-0016:STH00547X
`
`5
`
`Luitpold Pharmaceuticals, Inc., Ex. 2027, P. 1
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`6
`
`SEMINARS IN THROMBOSIS AND HEMOSTASIS-VOL. 25, SUPPL. 3, 1999
`
`LMWH having an enhanced anti-factor Xa/anti-factor
`Ila ratio would facilitate more subtle regulation of coag(cid:173)
`ulation and an improved therapeutic index.8-10
`Our understanding of the precise mechanism of ac(cid:173)
`tion of LMWHs through biochemical, pharmacological,
`and clinical studies have suggested that the initial ratio(cid:173)
`nale for their development may have been naive and not
`entirely correct.7·8 Nevertheless, LMWHs have been
`successfully introduced as new effective and improved
`antithrombotic/anticoagulant agents
`throughout
`the
`world. This review focuses on the chemical processing
`and production of LMWHs and how the various process(cid:173)
`ing routes result in structural differences among these
`pharmaceutical agents.
`
`WHAT IS HEPARIN?
`
`Within a decade of its discovery in 1916, heparin
`was identified as an anionic polysaccharide containing a
`uronic acid residue2 (Fig. 1). Early researchers showed
`that heparin also contained 0-sulfate esters and N-sul(cid:173)
`fated glucosamine residues. By 1970, iduronic acid was
`shown to be the major uronic acid component in heparin
`and a generalized structure of heparin could be drawn.
`Over the past two decades, the structure of the AT pen(cid:173)
`tasaccharide binding site has been discovered with much
`of heparin's fine structure elucidated, and an improved
`understanding of its conformation2l-23 and interaction
`with proteins was established.16-18,20,24-27
`Heparin is prepared by extraction from the tissue of
`slaughter house animals (i.e., porcine intestine, bovine
`lung). Like all other natural polysaccharides, heparin is a
`polydisperse mixture containing a large number of chains
`having different molecular weights (MWs).28,29 Heparin
`is composed of a major trisulfated disaccharide repeating
`unit (Fig. 1 ), but it also contains a number of additional
`disaccharide structures.18-20 It is these additional disac(cid:173)
`charide units that make heparin's structure complex and
`that also comprise the AT pentasaccharide binding site,
`important for heparin's anticoagulant activity.
`
`The heparin family of glycosaminoglycans (GAGs)
`includes both heparin and the related undersulfated poly(cid:173)
`saccharide heparan sulfate.30.31 While heparin and he(cid:173)
`paran sulfate GAGs are biosynthesized through a com(cid:173)
`mon pathway, structural studies clearly indicate that the
`structures of heparin and heparan sulfate are distinctly
`different.32,33 All the disaccharides found within heparin,
`including those comprising the AT pentasaccharide bind(cid:173)
`ing site34, are also found within heparan sulfate but in
`different proportions.30 Heparin and heparan sulfate,
`both found in tissues commonly used to prepare pharma(cid:173)
`ceutical grade heparin, differ substantially in their anti(cid:173)
`coagulant activity.3o Extraction methods that focus on
`the high specific anticoagulant activity required to meet
`United States Pharmacopeia (USP) specifications serve
`to eliminate much (but not all) of the heparan sulfate
`GAG from pharmaceutical grade heparin.
`Pharmaceutical grade heparin is a purified tissue
`extract comprised primarily of polydisperse GAGs con(cid:173)
`sisting primarily of heparin but containing other GAGs,
`such as heparan sulfate. Small amounts of dermatan sul(cid:173)
`fate, once present in some pharmaceutical grade he(cid:173)
`parins, have now been virtually eliminated.18,28,35 Chains
`of molecular weight from 5000 to over 40,000, making
`up polydisperse pharmaceutical grade heparin, also dis(cid:173)
`play significant sequence heterogeneity.' For example,
`many fully sulfated heparin chains are simply composed
`of uniform repeating sequences of trisulfated disaccha(cid:173)
`ride (Fig. 1). Alternatively, heparin chains having an in(cid:173)
`termediate level of sulfation are comprised of long seg(cid:173)
`ments of fully sulfated sequences with intervening
`undersulfated domains, such as that comprising the AT
`pentasaccharide binding site (Fig. 1). Finally, undersul(cid:173)
`fated heparin chains ( <2 sulfate groups/disaccharide)
`may simply be contaminating heparan sulfate.
`Not all heparin chains contain an AT pentasaccha(cid:173)
`ride binding site. Only 20 to 50% of the polysaccharide
`chains comprising pharmaceutical grade heparin contain
`an AT binding site and are called "high affinity hepa(cid:173)
`rin."1 No difference has been reported in the overall
`charge.or the average size of high affinity and low affin-
`
`OH
`
`Trisulfated disaccharide Disulfated disaccharide Antithrombln Pentasaccharide Binding Site
`
`Trisulfated disaccharide
`
`n+m= 16
`for MW 12,000
`FIG. 1. Chemical structure of a representative chain of pharmaceutical heparin. Clusters of trisulfated disaccharides (n and m, where
`n + m = 16 for MW 12,000), flank disulfated disaccharides and AT pentasaccharide binding site (ABCDE). Some structural variability
`both within and outside the AT binding site is indicated by multiple substituents. The attached substituents correspond to the major AT
`binding site structure found in porcine intestinal heparin.
`
`Luitpold Pharmaceuticals, Inc., Ex. 2027, P. 2
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`PRODUCTION AND CHEMICAL PROCESSING OF LMWHS-LINHARDT, GUNAY
`
`7
`
`TABLE 1. Composition of Heparins from Different Species and Tissues*
`
`Species
`
`Porcine
`Bovine
`Bovine
`Ovine
`Hen
`Clam
`
`Tissue
`
`intestine
`lung
`intestine
`intestine
`intestine
`
`N-acetylAT
`Binding Site
`
`0.5 (0.3-0.7)
`0.3
`0.3
`0.7
`0.3
`0.5
`
`Average Number in One Heparin Chaint
`
`N-sulfoAT
`Binding Site
`
`Trisulf ated
`Disaccharide
`
`Disulfated
`Disaccharide
`
`0.1
`0.3
`0.3
`0.4
`0.2
`0.4
`
`10 (10-15)
`14
`10
`11
`6.7
`5.0
`
`1.2(1-2)
`1.0
`1.7
`1.4
`1.7
`1.9
`
`*adapted from Loganathan et al.19
`tThe numbers shown in parentheses indicate a range of values typically observed.
`
`ity heparin. Some high MW heparin chains may contain
`more than a single AT binding site and thus display an
`enhanced level of anticoagulant activity. 36
`Heparins obtained from different tissues and differ(cid:173)
`ent species also differ structurally (Table 1).19 The most
`widely used tissue for the preparation of pharmaceutical
`grade heparin is porcine intestine. Heparin prepared from
`bovine lung differs substantially from porcine intestinal
`heparin. Bovine lung heparin has a higher sulfation level
`and slightly higher MW than porcine intestinal heparin,
`increasing its affinity for thrombin (factor Ila). Porcine in(cid:173)
`testinal heparin contains an AT binding site primarily hav(cid:173)
`ing an N-acetyl group in residue A (Fig. 1), while bovine
`lung heparin primarily has an N-sulfo group at residue A,
`resulting in their slightly different affinities for AT.19
`The disaccharide composition of porcine intestinal
`heparins can also differ substantially from each other.18
`The two mainly used raw materials (intestinal mucosa
`and whole intestine) contain differing amounts of conta(cid:173)
`minating heparan sulfate that can carry over into the fi(cid:173)
`nal pharmaceutical product. There are different sub(cid:173)
`species of hogs and the mast cell content of intestinal
`tissue can vary based on the diet and environment in
`which the animals are raised. These variables potentially
`contribute to the already complex structure of pharma(cid:173)
`ceutical grade heparin.
`
`HOW IS HEPARIN PREPARED?
`
`Methods of commercial production of pharmaceuti(cid:173)
`cal grade heparin are tightly guarded industrial· secrets
`and few publications or patents describe most commonly
`used pharmaceutical processes. The process of preparing
`pharmaceutical grade heparin has been altered some(cid:173)
`what over time as the primary tissue source has changed
`from dog liver to beef lung and finally to porcine intes(cid:173)
`tine.37 The methods used today for the commercial
`preparation of heparin involve five basic steps: (1)
`preparation of tissue; (2) extraction of heparin from tis(cid:173)
`sue; (3) recovery ofraw heparin; ( 4) purification of hep(cid:173)
`arin; and (5) recovery of purified heparin.
`
`The preparation of the tissue begins with the collec(cid:173)
`tion of the appropriate animal organ tissue at the slaugh(cid:173)
`terhouse and its preparation for processing. The whole
`intestine is either used to prepare "hashed pork guts" or
`processed into casings, which requires removal of the
`endothelial lining from the intestinal lumen. Crude hepa(cid:173)
`rin extraction typically takes place at the hog slaughter(cid:173)
`ing facility itself. Additional high potency heparin may
`be recovered by saving the waste brine solution of the
`hog casings operation.38
`In the second step, heparin is separated from the
`tissue. The use of elevated temperatures and pressures39
`and/or proteases ensures the solubilization of all GAGs.
`Currently, commercial crude heparin extraction processes
`involve hydrolysis at alkaline pH aided by proteolytic en(cid:173)
`zymes. Optionally, the digested tissue may be filtered or
`screened to remove any large particles yielding a deeply
`colored solution containing GAGs, peptides, and nucleic
`acids. At this point, the enzyme is often inactivated by
`heating the filtrate for 15 minutes at 90°C, at the same
`time serving as a sanitary step.
`The third step is the recovery of raw heparin. Cur(cid:173)
`rently, anion exchange resin is added, enabling the hepa(cid:173)
`rin-like GAGs to selectively adsorb onto the resin4o ac(cid:173)
`cording to the charge-density of the different GAGs.
`After complete adsorption of the heparin, the resin is de(cid:173)
`livered to a crude heparin manufacturing facility where
`it is washed and subsequently eluted. The concentrated
`crude heparin solution is usually filtered, precipitated,
`and vacuum dried (stage 12 heparin).
`The purification of crude heparin is typically per(cid:173)
`formed under good manufacturing practices conditions
`and deals with potential impurities originating from the
`starting material or introduced during crude heparin extrac(cid:173)
`tion. Generally, the crude heparin is dissolved in purified
`water, filtered at low pH to remove residual protein, and
`oxidized at alkaline pH to sanitize, decolorize, and depyro(cid:173)
`genate the material. This is often followed by cation ex(cid:173)
`change chromatography to remove extraneous cations,
`ethanol precipitation to reduce nucleotides, and sometimes
`by a chemical treatment to inactivate any viruses that
`might be present. The purified heparin is precipitated and
`
`Luitpold Pharmaceuticals, Inc., Ex. 2027, P. 3
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`8
`
`SEMINARS IN THROMBOSIS AND HEMOSTASIS-VOL. 25, SUPPL. 3, 1999
`
`either vacuum-dried or redissolved in purified water, fol(cid:173)
`lowed by various filtration steps and freeze-drying. The
`yield of porcine intestinal heparin is typically 10-25 mg/g
`wet tissue corresponding to 30,000 to 50,000 U/animal.
`
`WHAT ARE LMWHS?
`
`LMWHs are defined as salts of sulfated GAGs hav(cid:173)
`ing an average MW of less than 8000 Da and for which
`at least 60% of all molecules have a MW of less than
`8000 Da. These are obtained by fractionation or depoly(cid:173)
`merization of heparin and have a potency of greater than
`70 units/mg of anti-factor Xa activity and a ratio of anti(cid:173)
`factor Xa activity to anti-factor Ila activity of::=:: 1.5.41
`Before any LMWHs had been approved for human
`use, the implicit goal of pharmaceutical scientists was
`that the composition of these LMWHs should closely re(cid:173)
`semble the structure of heparin in all aspects except MW
`and ratio of anti-factor Xa to anti-factor Ila activity. An
`ideal LMWH might be simply a LMW subfraction of
`heparin prepared by sizing, using gel permeation chro(cid:173)
`matography (GPC). Direct size fractionation has been
`used to prepare a LMWH on a laboratory bench scale
`that exhibits the appropriate MW and activity properties,
`and contains the same disaccharide composition and se(cid:173)
`quences as heparin. Such methods, however, are rarely
`used on the scale required for the commercial manufac(cid:173)
`ture of heparin.42 Pharmaceutical chemists have relied
`on a number of chemical or enzymatic depolymeriza(cid:173)
`tion methods to manufacture commercial quantities of
`LMWHs. These depolymerization methods were se(cid:173)
`lected to give a product with: (1) suitable average MW
`and low polydispersity; (2) anti-factor Xa/anti-factor Ila
`activity >1; (3) structure similar to a LMWH prepared
`through fractionation and with few structural artifacts
`resulting from the depolymerization method used; ( 4) no
`residual toxic reagents;43 and (5) a cost-effective repro(cid:173)
`ducible and scalable process having a minimum number
`of process steps, little if any required purification, nei(cid:173)
`ther labor, reagent nor capital intensive, and high yield(cid:173)
`ing. While no current manufacturing process meets all of
`these goals, the currently used processes have afforded a
`first generation of clinically useful LMWHs.
`
`METHODS FOR PREPARING LMWHS
`
`Many years of experience in the manufacture of
`pharmaceutical grade heparin have shown that it exhibits
`a surprisingly high level of physical and chemical stability
`with a shelf life approaching a decade. Numerous
`processes have been used to prepare pharmaceutical grade
`heparins involving the use of harsh conditions including
`elevated temperature, pressure, shear, high ionic strength,
`acid, base, and organic solvents. These processes have re(cid:173)
`producibly afforded a uniformly high quality product. The
`
`success of the manufacturers represented the only avail(cid:173)
`able data demonstrating the physical and chemical stabil(cid:173)
`ity of heparin. Recently, an accelerated stability study un(cid:173)
`der elevated temperatures and under acidic and basic
`conditions confirmed the surprising stability of heparin.44
`A decomposition pathway for heparin under these stressed
`conditions has been proposed. Elevated temperatures can
`result in substantial damage to functionality within the
`heparin molecule (i.e., loss of sulfation) that occurs con(cid:173)
`currently with depolymerization. Neutral and acidic path(cid:173)
`ways result in a similar formation of small desulfated
`products, while the basic pathway terminates in a Canniz(cid:173)
`zaro reaction and de Bruyn van Eckenstein rearrange(cid:173)
`ment.42 Other physical parameters such as agitation result
`in no structural alterations, as the heparin molecule is not
`sufficiently large to be shear-sensitive.
`The oxidative instability of heparin had been
`widely observed by heparin manufacturers. Indeed, an(cid:173)
`tioxidants (bisulfite and metal chelators) have been
`added at various stages in heparin's manufacture to en(cid:173)
`hance stability.42 These observations suggested the pos(cid:173)
`sibility of utilizing oxidative methods
`to prepare
`LMWHs. Manufacturers have also observed microbial
`degradation of heparin. A bacterial enzyme, heparin
`lyase I (heparinase), is known to act on heparin.45,46 This
`enzyme acts in a random endolytic fashion through a 13-
`eliminative cleavage mechanism.46 This enzymatic reac(cid:173)
`tion can be mimicked chemically by esterifying the car(cid:173)
`boxyl group of the uronic acid residue and treating the
`resulting heparin ester with base.47 Thus, enzymatic or
`chemical 13-eliminative cleavage offers a second possi(cid:173)
`ble method for heparin depolymerization and the manu(cid:173)
`facture of LMWHs. It is interesting to note that many of
`the processes to prepare LMWHs started out as analyti(cid:173)
`cal tools to degrade heparin in an effort to understand its
`structure.
`Heparin can be oxidatively broken down using a
`variety of oxygen containing reagents like hydrogen per(cid:173)
`oxide or by ionizing -y-irradiation (Fig. 2).11.48-50 Each of
`these methods relies on the generation of oxygen radi(cid:173)
`cals that !ife believed to act by oxidizing sensitive sac(cid:173)
`charide residues within the heparin polymer. Nonreduc(cid:173)
`ing sugars are essentially inert to aqueous hydrogen
`peroxide except in the presence of alkali or in the pres(cid:173)
`ence of a metal catalyst. Both of these conditions lead to
`the generation of the hydroxy 1 radical that will react with
`sugar residues and degrade them to 1-, 2- and 3-carbon
`fragments without modifying the residues on either side
`of the point of attack. The most susceptible residues ap(cid:173)
`pear to be those that are unsubstituted at positions 2 and
`3 in the sugar ring. Studies of the composition of disac(cid:173)
`charides that result from oxidative depolymerization of
`LMWHs suggest that nonsulfated uronic acid residues in
`heparin are selectively oxidized to volatile acids (i.e.,
`formic acid).51 Under controlled conditions (tempera(cid:173)
`ture, pressure, time, oxidant), LMWHs having appropri(cid:173)
`ate MWs and activity can be obtained in reasonable
`
`Luitpold Pharmaceuticals, Inc., Ex. 2027, P. 4
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`PRODUCTION AND CHEMICAL PROCESSING OF LMWHS-LINHARDT, GUNAY
`
`9
`
`HO
`
`OX
`
`O
`
`o
`
`~HY
`
`0
`
`H
`
`OH
`
`OH
`
`OX
`
`m
`
`NHS03-
`
`f:{-
`~2ox Q }2 - CH,OS03-
`0xid~
`co:;on ~
`
`Chemic~) - _
`/
`~-elimin/
`
`~ ~-~1hmatic
`~mination
`
`CH,OS03-
`
`~0"
`
`NHS03-
`
`£;-0"
`
`NHS03-
`
`m
`m
`FIG. 2. Depolymerization of heparin to prepare LMWHs. The heparin chain in the center can undergo depolymerization by each of
`the four processes shown. Heparin chain size is reduced (n > m), affording a LMWH.
`
`yields. 11 •4s,s2 Of these oxidative methods, only hydrogen
`peroxide has been utilized to commercially prepare
`LMWHs (ardeparin sodium and parnaparin sodium) for
`clinical use (Table 2).
`In addition to oxygen radical processes, it is possi(cid:173)
`ble to oxidatively depolymerize heparin through deami(cid:173)
`nation (Fig. 2). In these reactions, heparin is N-ni(cid:173)
`trosated, using either nitrous acid or another nitrosating
`reagent such as isoamyl nitrite, at the amino group of its
`N-sulfoglucosamine residues. The resulting unstable N-
`
`nitrososulfamide loses nitrogen and sulfate and gener(cid:173)
`ates a carbocation at C-2 of the saccharide residue. Sub(cid:173)
`sequent ring contraction of this residue and hydrolysis of
`the adjacent glycosidic bond affords a LMWH. Each
`product chain resulting from this process contains an an(cid:173)
`hydromannose residue (bearing a terminal aldehyde) at
`the reducing terminus. This residue can subsequently be
`converted to anhydromannitol using a reducing agent,
`such as sodium borohydride. Controlled deaminative
`cleavage is possible by controlling the process condi-
`
`TABLE 2. Commercially Avaifable LMWHs
`
`LMWH
`
`Trade Name
`
`Manufacturer
`
`Preparation Method
`
`Ardeparin sodium
`Certoparin sodium
`Dalteparin sodium
`
`Normiflo
`Sandoparin
`Fragmin
`
`Enoxaparin sodium
`
`Lovenox
`Clexane
`
`Wyeth-Ayerst
`Novartis
`Pharmacia-U pjohn
`Kissei
`Rhone-Poulenc Rorer
`Avantis
`
`Oxidative depolymerization with H20 2
`Deaminative cleavage with isoamyl nitrite
`Deaminative cleavage with nitrous acid
`
`B-eliminative cleavage of the benzyl ester of heparin
`by alkaline treatment
`
`Nadroparin calcium
`
`Fraxiparin
`
`Sanofi-Winthrop
`
`Deaminative cleavage with nitrous acid
`
`Parnaparin sodium
`Reviparin sodium
`
`Fluxum
`Clivarin
`
`Alfa Wassermann
`Knoll
`
`Oxidative depolymerization with Cu+ and Hz02
`Deaminative cleavage with nitrous acid
`
`Tinzaparin sodium
`
`Innohep
`Logiparin
`
`Braun
`Novo/Leo/Dupont
`
`B-eliminative cleavage by heparinase
`
`Approved
`Markets
`
`USA
`Germany
`USA, Japan
`UK, Germany
`USA
`Germany
`Spain
`France
`Germany
`Italy
`Canada
`Germany
`Germany
`Denmark,
`USA
`
`Luitpold Pharmaceuticals, Inc., Ex. 2027, P. 5
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`10
`
`SEMINARS IN THROMBOSIS AND HEMOSTASIS-VOL. 25, SUPPL. 3, 1999
`
`tions (temperature, pH, time) or by limiting the amount
`of nitrosation reagent.53,54 The LMWH product formed
`using these controlled conditions is obtained in high
`yield and has the appropriate chemical and biological
`properties. Several LMWHs prepared through deamina(cid:173)
`tive cleavage are currently used clinically (Table 2).
`Two 13-eliminative methods, one enzymatic and the
`other chemical, are used to commercially prepare
`LMWHs. In the enzymatic method, heparin lyase I (or
`heparinase) is used to depolymerize heparin.is The ex(cid:173)
`tent of this reaction can be conveniently monitored by
`measuring the change in absorbance associated with the
`unsaturated uronate residue formed in each product mol(cid:173)
`ecule.46,55 Cleavage takes place only at the 2-0-sul(cid:173)
`foiduronic acid residue.46 The depolymerization is
`stopped by removing or inactivating the enzyme. After
`recovery of the GAG from the enzyme and very low
`MW byproducts (i.e., disaccharides and tetrasaccha(cid:173)
`rides), a LMWH is obtained that has the desired MW
`and activity properties. This method is used to prepare
`the clinically used LMWH product, tinzaparin sodium
`(Table 2). Chemical 13-elimination can involve the direct
`treatment of heparin or its quaternary ammonium salt
`with base.56,57 Alternatively, the benzyl ester can be pre(cid:173)
`pared by treatment of the benzethonium salt of heparin
`with benzyl chloride and base with heating.58 Under
`these conditions, chemical 13-elimination takes place, af(cid:173)
`fording LMWH that contains an unsaturated uronate
`residue in the nonreducing end. Cleavage occurs specifi(cid:173)
`cally at iduronic acid without preference for the presence
`or absence of a 2-0-sulfo group. Proper control of these
`process conditions affords the clinically used LMWH,
`enoxaparin sodium (Table 2).
`
`ANALYSIS OF HEPARIN AND LMWHS
`
`The analysis of pharmaceutical grade heparin is
`governed by the USP and other national pharmacopeia.
`Bioanalyses include a routine in vitro coagulation assay
`and a test for bacterial endotoxins (pyrogens). The hepa(cid:173)
`rin assay, when expressed in units/mg or international
`units/mg, is the amount of heparin that will cause 1 ml of
`sheep plasma to half-clot when kept for 1 hour at 20°C
`compared to a USP reference standard (K-4) or the Fifth
`International Standard for Unfractionated Heparin
`(WH0-5), respectively. The analysis of LMWHs is' gov(cid:173)
`erned by the European Pharmacopeia and proposals
`have also been made to amend the USP monograph to
`cover LMWHs.41 Such analyses include in vitro ami(cid:173)
`dolytic assays in a purified system for anti-factor Xa and
`anti-factor Ila activities, and structural identification by
`nuclear magnetic resonance and size exclusion chro(cid:173)
`matography.
`Less routine analyses rely on in vitro protein (i.e.,
`AT) binding assays and protein interaction studies, so(cid:173)
`phisticated ex vivo or in vivo animal assays and finally,
`
`human clinical evaluation.3-7,36,59 Refined chemical,
`chromatographic, electrophoretic, and spectroscopic as(cid:173)
`says have been developed that are often used by research
`laboratories studying heparin. The presence of unsubsti(cid:173)
`tuted glucosamine can be measured in heparin and
`LMWHs by chemical modification with an amine reac(cid:173)
`tive fluorescent reagent.60 While chemical assays for
`uronic acid glucosamine have been used in heparin
`analysis, these have been largely displaced by rapid and
`more reliable spectroscopic analysis.61 Chromatographic
`analysis of heparin and LMWHs primarily involves
`high-pressure liquid chromatography (HPLC), such as
`gel-permeation chromatography (GPC) to determine
`MW and polydispersity.28,29 This method, once requiring
`the tedious production of MW standards, can now be
`performed using a heparinase-prepared LMWH standard
`through the clever use of dual mass and absorbance de(cid:173)
`tection. 20,62 Ion exchange chromatography, following
`sample pyrolysis, can be accurately used to determine
`the level of sulfation or the presence of contaminating
`metals or anions. 63 Disaccharide compositional analysis
`(Table 3) and oligosaccharide mapping of heparin and
`LMWHs have in the past relied on HPLC analysis fol(cid:173)
`lowing the complete or partial heparinase catalysis of ni(cid:173)
`trous acid depolymerization of heparin or LMWHs.51,64
`Capillary electrophoresis (CE) has gained popularity,
`making disaccharide analysis and oligosaccharide map(cid:173)
`ping extremely rapid and sensitive.65,66 Oligosaccharide
`mapping by CE can provide data on the content of AT
`binding sites in different lots of a given LMWH.67 Poly(cid:173)
`acrylamide gel electrophoresis has been used to deter(cid:173)
`mine the MW and polydispersity of heparin and
`LMWHs.68 Spectroscopic analysis, while requiring ex(cid:173)
`pensive instrumentation, can facilitate the high resolu(cid:173)
`tion analysis of heparin and LMWHs, often providing
`information that is not available using any other tech(cid:173)
`niques. Proton nuclear magnetic resonance (NMR) spec(cid:173)
`troscopy has been used to determine the ratio of iduronic
`to glucuronic acid, the content of an N-acetylglu(cid:173)
`cosarnine, and position of sulfation.20,3o,59,60 Since this
`method is· quantitative and nondestructive, it can be con(cid:173)
`veniently used to afford a disaccharide analysis.30 Proton
`NMR has also replaced more tedious methods for the de(cid:173)
`termination of contaminating GAGs, such as dermatan
`sulfate,35 or the contaminating organic molecules that
`might be used in the manufacturing of a pharmaceutical
`grade heparin or LMWH.69 Finally, proton NMR is ex(cid:173)
`tremely valuable in the conformational analysis of hepa(cid:173)
`rin.21,22 Carbon NMR spectroscopy has also been used in
`heparin and LMWH analysis. While requiring substan(cid:173)
`tially more sample than proton NMR, this method re(cid:173)
`sults in higher signal resolution and offers abundant
`structural information.69 By measuring the intensity of
`signals resulting from the reducing end anomeric carbon
`and all other anomeric carbons, this method offers a so(cid:173)
`phisticated standard-free determination of the number(cid:173)
`averaged MW ofheparin7o and LMWH.71 While modern
`
`Luitpold Pharmaceuticals, Inc., Ex. 2027, P. 6
`
`Pharmacosmos A/S v. Luitpold Ex. Pharmaceuticals, Inc., IPR2015-01490
`
`

`
`PRODUCTION AND CHEMICAL PROCESSING OF LMWHS-LINHARDT, GUNAY
`
`11
`
`TABLE3. Disaccharide Composition of LMWHss1
`
`Trisulfated Disaccharides
`
`Disulfated Disaccharides
`
`Monosulfated Disaccharide
`
`Sample
`
`Heparin
`Fractionated
`LMWH
`Ardeparin
`Sodium
`Dalteparin
`Sodium
`Enoxaparin
`Sodium
`Nadroparin
`Calcium
`Parnaparin
`Sodium
`Tinzaparin
`Sodium
`
`2SNS6S
`
`NS3S6S
`
`NS6S
`
`2SNS
`
`51.9
`34.7
`
`86.1
`
`49.3
`
`71.3
`
`17.2
`
`48.4
`
`88.9
`
`1.4
`1.2
`
`1.7
`
`2.6
`
`1.9
`
`0.8
`
`0.4
`
`1.9
`
`4.1
`4.0
`
`7.0
`
`3.2
`
`11.4
`
`1.2
`
`3.6
`
`7.5
`
`2.6
`1.6
`
`6.1
`
`3.1
`
`2.5
`
`1.8
`
`4.4
`
`5.4
`
`6S
`
`1.1
`0.9
`
`1.3
`
`2.0
`
`1.4
`
`0.7
`
`0.3
`
`1.4
`
`soft ionization techniques available in mass spectrome(cid:173)
`try (MS) offer some intriguing possibilities for heparin
`and LMWH analysis, the only successful application of
`MS has been the characterization of heparin-derived
`oligosaccharides. Fast atom bombardment (FAB),72
`MS/MS-FAB,73 electrospray ionization (ESI),74 and ma(cid:173)
`trix-assisted laser desorption ionization (MALDI)75 have
`all proved effective in such analyses. CE or HPLC/MS
`interfaces also offer possibilities for improved, standard(cid:173)
`free analysis of heparin disaccharides and oligosaccha(cid:173)
`rides. The primary limitation for the direct MS analysis
`of heparin or LMWHs remains their high polydispersity
`and microheterogeneity. Other spectral techniques, such
`as las

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket