throbber
New strategy for chemical modification of hyaluronic acid:
`Preparation of functionalized derivatives and their use in
`the formation of novel biocompatible hydrogels
`
`Paul Bulpitt, Daniel Aeschlimann
`Division of Orthopedic Surgery, University of Wisconsin, H5/301 Clinical Science Center, 600 Highland Avenue,
`Madison, Wisconsin 53792
`
`Received 2 September 1998; revised 4 January 1999; accepted 3 March 1999
`
`Abstract: Biodegradable materials for spatially and tempo-
`rally controlled delivery of bioactive agents such as drugs,
`growth factors, or cytokines are key to facilitating tissue
`repair. We have developed a versatile method for chemical
`crosslinking high-molecular-weight hyaluronic acid under
`physiological conditions yielding biocompatible and biode-
`gradable hydrogels. The method is based on the introduc-
`tion of functional groups onto hyaluronic acid by formation
`of an active ester at the carboxylate of the glucuronic acid
`moiety and subsequent substitution with a side chain con-
`taining a nucleophilic group on one end and a (protected)
`functional group on the other. We have formed hyaluronic
`acid with amino or aldehyde functionality, and subse-
`quently hydrogels with these hyaluronic acid derivatives
`and bifunctional crosslinkers or mixtures of the hyaluronic
`acid derivatives carrying different functionalities using ac-
`tive ester- or aldehyde-mediated reactions. Size analysis of
`the hyaluronic acid derivatives showed that the chemical
`
`modification did not lead to fragmentation of the polysac-
`charide. Hydrogels formed with hyaluronic acid derivatized
`to a varying degree and crosslinked with low- or high-
`molecular-weight crosslinkers were evaluated for biode-
`gradability by digestion with hyaluronidase and for biocom-
`patibility and ectopic bone formation by subcutaneous im-
`plantation in rats. Several hydrogel formulations showed
`excellent cell infiltration and chondro-osseous differentia-
`tion when loaded with bone morphogenetic protein-2 (BMP-
`2). Synergistic action of insulin-like growth factor-1 with
`BMP-2 promoted cartilage formation in this model, while
`addition of transforming growth factor-b and BMP-2 led to
`rapid replacement of the matrix by bone. © 1999 John Wiley
`& Sons, Inc. J Biomed Mater Res, 47, 152–169, 1999.
`
`Key words: hyaluronic acid; crosslinking; tissue repair; cy-
`tokine delivery; bone formation
`
`INTRODUCTION
`
`In orthopedic surgery, defects in articular cartilage
`present a very complicated treatment problem be-
`cause of the very limited capacity of cartilage to repair
`spontaneously. The failure of regenerating persistent
`hyaline cartilage by surgical procedures prompted in-
`vestigators to attempt repair using biological strate-
`gies.1 Repair has been induced by transplantation of
`culture-expanded autologous chondrocytes2 or by re-
`cruitment of mesenchymal stem cells from the syno-
`vium using chemotactic and mitogenic factors.3 The
`shortcoming of both strategies is the difficulty in sta-
`bly anchoring the repair-inducing factors, whether
`cells or growth factors, within the lesion. Hyaluronic
`acid (HA) is a good candidate for the development of
`
`Correspondence to: D. Aeschlimann
`Contract grant sponsor: Orthogene, Inc.
`
`© 1999 John Wiley & Sons, Inc.
`
`CCC 0021-9304/99/020152-18
`
`novel biomaterials for local delivery of cells and bio-
`active factors because of its unique physicochemical
`properties and its excellent biocompatibility and bio-
`degradability.
`Hyaluronic acid, or hyaluronan, is a natural poly-
`saccharide that is most abundant in cartilage and in
`the vitreous.4 HA plays a key structural role in the
`organization of the cartilage extracellular matrix as an
`organizing structure for the assembly of aggrecan, the
`large cartilage proteoglycan.5 The highly negatively
`charged aggrecan/HA assemblies are largely respon-
`sible for the viscoelastic properties of cartilage by im-
`mobilizing water molecules. HA is unique among gly-
`cosaminoglycans with respect to not being covalently
`bound to a polypeptide. HA is also unique in having
`a relatively simple structure of repeating nonsulfated
`disaccharide units composed of D-glucuronic acid and
`N-acetyl-D-glucosamine (Fig. 1). The molecular mass
`of HA in extracellular matrices is typically several mil-
`lion Daltons. Besides its structural function in extra-
`cellular matrix assembly, HA plays pivotal roles in
`
`ALL 2093
`PROLLENIUM V. ALLERGAN
`IPR2019-01505 et al.
`
`

`

`IN SITU POLYMERIZABLE HYALURONIC ACID HYDROGEL MATERIALS
`
`153
`
`Figure 1. Structure of HA.
`
`promoting cell motility and differentiation in devel-
`opment and wound healing.4,6 Blocking the interac-
`tion of HA with cell-surface receptors, i.e., isoforms of
`CD44, in prechondrogenic micromass cultures from
`embryonic limb bud mesoderm inhibits chondrogen-
`esis, indicating that the establishment and mainte-
`nance of a differentiated chondrocyte phenotype is at
`least in part dependent on HA and HA–receptor in-
`teractions.7
`Based on its unique rheological properties, HA is
`currently being used clinically in viscosupplementa-
`tion and viscosurgery.4,8 Viscous solutions of high-
`molecular-weight HA and its salts are being used in
`therapy for arthropathies by intraarticular injection,9
`to promote wound healing in various tissues, and as a
`surgical aid in eye and middle-ear surgery. More re-
`cently, HA has been used in derivatized and/or
`crosslinked form to manufacture thin films which are
`used to create tissue separations.10 Extensive efforts
`have been made by various laboratories to produce
`derivatives of HA with unique properties for specific
`biomedical applications. Most of the developments
`have been focusing on the production of materials
`such as films or sponges for implantation and the sub-
`stitution of HA with therapeutic agents11–13 for de-
`layed release and/or prolonged effect. In fact, the half-
`life of pharmacological compounds has been shown to
`be drastically increased when delivered systemically
`as a HA conjugate.11,12 Strategies for modification of
`HA have included esterification of HA,14,15 acrylation
`of HA,16 and crosslinking of HA using divinyl sul-
`fone17 or glycidyl ether.18 However, the modified HA
`molecules show altered physical characteristics such
`as decreased solubility in water, and/or the chemical
`reaction strategies used are not designed for crosslink-
`ing of HA under physiological conditions, and thus
`cannot be used to polymerize a biodegradable matrix
`in situ which would be desirable in resurfacing of ar-
`ticular cartilage.
`
`The introduction of functional groups on HA, e.g.,
`amino groups, which could be used for further con-
`venient coupling or crosslinking reactions under mild
`physiological conditions, is a subject of great interest.
`Previous methods have produced a free amino group
`on high-molecular-weight HA by alkaline N-deace-
`tylation of its glucosamine moiety.19,20 However, con-
`comitant degradation of HA via b-elimination in the
`glucuronic acid moiety was observed under the harsh
`reaction conditions needed. An early report claimed
`that carbodiimide-catalyzed reaction of HA with gly-
`cine methyl ester, a monofunctional amine, led to the
`formation of an amide linkage.21 This, however, has
`been proven in a number of studies not to be the
`case.22,23 Under mildly acidic conditions the unstable
`intermediate O-acylisourea is readily formed, which
`in the absence of nucleophiles, rearranges by a cyclic
`electronic displacement to a stable N-acylurea24 (Fig.
`2). This O fi N migration of the O-acylisourea also
`occurs when the nucleophile is a primary amine23 and
`any amide formation that does occur is insignificant,
`as reported by Ogamo et al.22 The carbodiimide-
`mediated introduction of a terminal hydrazido group
`on HA with a variable spacer has recently been
`achieved and has led to the ability to conduct further
`coupling and crosslinking reactions.13,25
`In this study, we have developed methodology for
`introducing side chains into HA by carbodiimide-
`mediated coupling of primary or secondary amines to
`the carboxyl group of the glucuronic acid moiety us-
`ing an active ester intermediate. We demonstrate that
`by “rescuing” the active O-acylisourea by formation of
`a more hydrolysis-resistant and nonrearrangable ac-
`tive ester intermediate, the coupling of primary
`amines to HA is possible. Using this methodology, we
`have generated HA derivatives carrying different
`functional groups for subsequent crosslinking to form
`hydrogels. Characterization of these hydrogels in dif-
`ferent biological assays established that novel biocom-
`
`

`

`154
`
`BULPITT AND AESCHLIMANN
`
`patible and biodegradable materials are formed with
`these HA derivatives.
`
`MATERIALS AND METHODS
`
`Synthesis of HA derivatives
`
`General procedure of carbodiimide/HOBt reactions
`with HA
`
`Sodium hyaluronate (100 mg, 0.25 mmol; MW > 1 × 106;
`Genzyme Pharmaceuticals, Haverhill, UK) was dissolved in
`
`H2O at a concentration of 3 mg/mL. To this solution was
`added a 30-fold molar excess of an amine or hydrazide (pKa
`3–8.5; 7.5 mmol), i.e., ethylenediamine, adipic dihydrazide.
`The pH of the reaction mixture was adjusted to 6.8 with 0.1
`M NaOH/0.1M HCl. 1-Ethyl-3-[3-(dimethylamino)propyl]-
`carbodiimide (EDC) (192 mg, 1 mmol; Aldrich Chemical
`Co.) and 1-hydroxybenzotriazole (HOBt) (135 mg, 1 mmol;
`Fluka Chemical Corp.) was dissolved in dimethylsulfoxide
`(DMSO)/H2O (1:1, 1 mL). After mixing, the pH of the reac-
`tion was maintained at 6.8 by the addition of 0.1M NaOH
`and the reaction was allowed to proceed overnight. The pH
`was subsequently adjusted to 7.0 with 0.1M NaOH and the
`derivatized HA exhaustively dialyzed (Spectra/Por RC 2,
`
`Figure 2. Strategy for coupling of different amines to HA. Generated aldehyde- and amine-functionalized HA derivatives
`are listed.
`
`

`

`IN SITU POLYMERIZABLE HYALURONIC ACID HYDROGEL MATERIALS
`
`155
`
`MW cutoff 12–14,000; Fisher Scientific) against distilled
`H2O. NaCl was added to produce a 5% w/v solution and the
`modified HA was precipitated by addition of 3 vol equiva-
`lents of ethanol. The precipitate was redissolved in H2O at a
`concentration of approximately 5 mg/mL and the purified
`product was freeze-dried and kept at 4°C under N2. The
`yield of product was typically ~ 80%.
`
`Preparation of dimethyl acetal hydrazido-HA (3)
`
`Preparation of N-(2,2-dimethoxyethyl)-4-(methoxycarbonyl)
`butanamide (1). EDC (4.98 g, 0.026 mol) was added to a mix-
`ture of aminoacetaldehyde dimethyl acetal (2.18 mL, 20
`mmol; Aldrich) and methyl monoester of succinic acid (2.64
`g, 20 mmol; Aldrich) in 75 mL of dichloromethane, and the
`reaction mixture was stirred for 24 h at room temperature.
`The solution was extracted successively with 50 mL each of
`ice-cold solutions of 0.75 M sulfuric acid, 1M NaCl, satu-
`rated sodium bicarbonate, and 1M NaCl. The organic phase
`was collected and dried with sodium sulfate. The solvent
`was evaporated under reduced pressure yielding a syrup
`which showed a single spot on charring upon thin-layer
`chromatography (TLC) in solvent A (Rf 0.75) and solvent B
`(Rf 0.24) (see Characterization: Analytical Methods). The ap-
`parent yield of 1 was 65% and the 1H-NMR spectra in deu-
`terated chloroform showed the following peaks: d 5.70 (bs,
`1H, NH), 4.34 (t, 1H, CH-(OCH3)), 3.67 (s, 3H, COOCH3),
`3.43–3.35 (s and t, 8H, CH3OC and CHCH2NH), 2.38–2.26
`(m, 4H, CH2CO).
`Formation of N-(2,2-dimethoxyethyl)-4-(hydrazido)butanamide
`(2) from 1. Anhydrous hydrazine (248 mL, 7.9 mmol; Aldrich)
`was added to a solution of 1 (1.73 g, 7.9 mmol) in 5 mL of
`anhydrous methanol. The mixture was stirred at room tem-
`perature overnight and the solvent subsequently evaporated
`under reduced pressure, yielding a solid residue. The resi-
`due was dissolved in H2O (6 mL) and extracted three times
`with an equal volume of dichloromethane. The aqueous so-
`lution was evaporated to dryness under reduced pressure
`and then further dried overnight in vacuo. The crystalline
`solid (1.04 g, 82% yield) was homogeneous on TLC in sol-
`vent A (Rf 0.10) when visualized by charring. The 1H-NMR
`spectrum indicated the loss of the ester methoxy group
`when compared to 1.
`Sodium hyaluronate was reacted with acyl hydrazide (2)
`using HOBt and EDC as described above, yielding 3 with an
`apparent degree of modification of ~ 65%. The 1H-NMR
`spectra in deuterated water [Fig. 3(B)] showed peaks at d
`3.28 (s, 6H, CH(OCH3)2), 2.50 (bs, 2H, CO ? CH2), 1.9 (bs, 3H,
`HA-NHCO ? CH3), 1.82 (bs, 2H, CH2
`? CO).
`
`1H-NMR of native and modified HA in D2O at
`Figure 3.
`270 Mhz: (A) native HA, (B) N-(2,2-dimethoxyethyl)-4-
`(hydrazido)butanamide-modified HA (3), and (C) lysine
`methyl ester-modified HA (6). Peaks are assigned as indi-
`cated on the structural formula and detailed in Materials
`and Methods. 1H-NMR results for other HA derivatives are
`provided in Materials and Methods.
`
`Preparation of 2-aminoethyl-HA (5).
`
`Sodium hyaluronate was reacted with ethylenediamine
`dihydrochloride (998 mg; Aldrich) using HOBt and EDC as
`described above. The apparent degree of modification was
`~ 25%: 1H-NMR (D2O) d 3.07 (t, 2H, CO ? NHCH2), 2.65 (bs,
`2H, CH2NH2), 1.9 (bs, 3H, HA-NHCO ? CH3).
`
`Preparation of adipic dihydrazide-HA (4)
`
`Preparation of lysine methyl ester-HA (6).
`
`Sodium hyaluronate was reacted with adipic dihydrazide
`(1.31 g; Aldrich) using HOBt and EDC as described above.
`The apparent degree of modification was ~ 65%: 1H-NMR
`(D2O) d 2.20 (m, 2H, NHNHCO ? CH2), 2.10 (m, 2H,
`CH2NHNH2), 1.35–1.55 (m, 4H, CH2CH2).
`
`Sodium hyaluronate was reacted with lysine methyl ester
`(1.748 g; Sigma, St. Louis, MO) using HOBt and EDC as
`described above. The apparent degree of modification was
`~ 25%: 1H-NMR (D2O) d 3.85 (s, 3H, CO2CH3), 3.0 (t, 2H,
`CH2NH2), 2.1 (bs, 3H, HA-NHCO ? CH3), 2.0–1.83 (dm, 2H,
`
`

`

`156
`
`BULPITT AND AESCHLIMANN
`
`NHCHCH2CH2), 1.81–1.70 (m, 2H, CH2CH2), 1.52–1.43 (m,
`2H, CH2CH2CH2) [Fig. 3(C)].
`
`Preparation of polyaldehyde by oxidation of HA
`
`General procedure of carbodiimide/sulfo-NHS
`reactions with HA
`
`To an aqueous solution of sodium hyaluronate (3 mg/mL)
`was added a 30-fold molar excess of an amine (pKa > 8.5; 7.5
`mmol), i.e., 1,4-diaminobutane. The pH of the reaction mix-
`ture was adjusted to 7.5 with 0.1M NaOH/0.1M HCl. EDC
`(192 mg, 1 mmol) and N-hydroxysulfosuccinimide (sulfo-
`NHS) (217 mg, 1 mmol; Fluka) were dissolved in H2O (1
`mL). After mixing, the pH of the reaction was maintained at
`7.5 by the addition of 0.1M NaOH and the reaction was
`allowed to proceed overnight. The HA derivatives were pu-
`rified and stored as described above.
`
`Preparation of aminoacetaldehyde dimethyl
`acetal-HA (7)
`
`Sodium hyaluronate was reacted with aminoacetaldehyde
`dimethyl acetal (781 mg) using sulfo-NHS and EDC as de-
`scribed above. The apparent degree of modification was
`~ 10%: 1H-NMR (D2O) d 3.30 (s, 6H, CH(OCH3)2), 1.9 (bs, 3H,
`HA-NHCO ? CH3).
`
`Preparation of 4-aminobutyl-HA (8)
`
`Sodium hyaluronate was reacted with 1,4-diaminobutane
`dihydrochloride (1.21 g; Aldrich) using sulfo-NHS and EDC
`as described above. The apparent degree of modification
`was ~ 10%: 1H-NMR (D2O) d 2.87 (m, 4H, CO ? NHCH2,
`CH2NH2), 1.9 (bs, 3H, HA-NHCO ? CH3), 1.55–1.35 (dm, 4H,
`CH2CH2).
`
`Preparation of 6-aminohexyl-HA (9)
`
`Sodium hyaluronate was reacted with 1,6-diaminohexane
`dihydrochloride (1.42 g; Aldrich) using sulfo-NHS and EDC
`as described above. The apparent degree of modification
`was ~ 10%: 1H-NMR (D2O) d 2.9 (m, 4H, CO ? NHCH2,
`CH2NH2), 1.9 (bs, 3H, HA-NHCO ? CH3), 1.6–1.1 (m, 6H,
`CH2CH2CH2).
`
`Sodium hyaluronate (300 mg, 0.75 mmol) was dissolved in
`H2O at a concentration of 20 mg/mL. Sodium periodate (160
`mg, 0.75 mmol; Aldrich) was added and the reaction al-
`lowed to proceed for 2 h at room temperature. Low-
`molecular-weight reaction products were removed by ex-
`haustive dialysis (Spectra/Por RC 2) and the polyaldehyde
`polymer solution was saturated with nitrogen by bubbling
`and stored at 4°C.
`
`Characterization of HA derivatives
`
`Analytical methods
`
`Thin-layer chromatography was done with silica gel 60
`F254 (Merck). The solvent systems used were (a) 4:1 (v/v)
`ethyl acetate:acetone; and (b) 3:1 (v/v) toluene:methanol.
`The 1H-NMR spectra were obtained with a Jeol E-270 instru-
`ment at 270 MHz. For NMR spectroscopy, HA was dis-
`solved in D2O/NaOD, pH 14, at a concentration of 1–2 mg/
`mL. Compound x admixed with HA served as a control for
`HA modified with compound x.
`
`Size distribution
`
`Preparations of HA or chondroitin sulfate (type A from
`bovine trachea, MW ~ 45,000; Sigma) were dissolved in PBS
`at a concentration of 1 mg/mL and 1 mL applied on a Su-
`perose 6 FPLC column (HR 10/30; Pharmacia) equilibrated
`in PBS (5.6 mM Na2HPO4, 1.06 mM KH2PO4, pH 7.4, 154
`mM NaCl). The flow rate was 0.25 mL/min and 0.5-mL
`fractions were collected and the absorbance at 205 nm re-
`corded. Elution profiles of different forms of HA under con-
`ditions minimizing HA–HA interactions (50 mM Na2HPO4,
`pH 11, 500 mM NaCl) were identical. N-Deacetylated HA
`was prepared according to Dahl et al.20 Briefly, high-
`molecular-weight (MW > 106) HA (10 mg, 0.025 mmol) and
`hydrazine sulfate (5 mg, 0.038 mmol) were mixed with an-
`hydrous hydrazine (0.6 mL) in a glass tube. The tube was
`sealed and heated to 100°C for time intervals ranging from 0
`to 180 min. After rapid cooling on ice, 0.75 mL of toluene
`was added and the mixture evaporated to dryness under
`reduced pressure. HA preparations were digested with 100
`U/mL testicular hyaluronidase for 2 h.
`
`Deprotection of HA-acetals to form HA-aldehydes
`
`Formation of HA hydrogels
`
`The acetal modified HA was dissolved in H2O at a con-
`centration of 5–10 mg/mL and 1M HCl was added to give a
`final concentration of 0.025M HCl. The solution was then
`allowed to stand at room temperature for 0.5–1.0 h. The
`solution was neutralized by the addition of 1M NaOH,
`yielding the deprotected HA-aldehyde.
`
`General procedure for crosslinking of
`HA derivatives
`
`Functionalized HA was dissolved by agitation at room
`temperature in PBS, pH 7.4–8.5, at a concentration of up to
`20 mg/mL to study the formation of hydrogels at different
`
`

`

`IN SITU POLYMERIZABLE HYALURONIC ACID HYDROGEL MATERIALS
`
`157
`
`HA concentrations, and at a concentration of 13–15 mg/mL
`for subsequent experiments. The degree of modification of
`the HA derivative was derived from the integration of the
`1H-NMR peaks. After complete dissolution, the HA solution
`was transferred into a 1-mL syringe. When reacting HA with
`low-MW crosslinkers, a slight excess of the compound (1.1
`molar equivalent of functional groups) was used in a second
`1-mL syringe. Polyethylene glycol bis(succinimidyl propio-
`nate) ((SPA)2-PEG) (MW ~ 3400) and succinimide ester of
`carboxymethylated four arm polyethylene glycol ((SC)4-
`PEG) (MW ~ 20,000) were purchased from Shearwater Poly-
`mers (Huntsville, AL), 3,38-dithiobis (sulfosuccinimidyl pro-
`pionate) (DTSSP) from Pierce Chemical Co., and glutaralde-
`hyde from Fluka. The solution was freshly prepared in PBS
`at 1/10 of the HA volume. The syringes were connected
`while paying special attention to exclude air; the contents
`were rapidly mixed with 20 passages and allowed to gel.
`When reacting HA molecules with different functionalities,
`0.5–1 equivalent of HA-aldehyde was mixed with 1 equiva-
`lent of HA-hydrazide, depending on the degree of modifi-
`cation of the HA derivatives. At room temperature, gelation
`occurred within 30 s to several minutes, depending on the
`formulation, and the gel properties did not significantly
`change after approximately 5 min.
`
`In vitro and in vivo testing of HA hydrogels
`
`Hyaluronidase digestion
`
`Hyaluronic acid hydrogels were formed as detailed above
`in 1-mL syringes. The syringes containing the crosslinked
`gels were incubated at 37°C for 1 h to ensure the crosslinking
`reaction was complete, after which identical ~ 100-mL cylin-
`drical gels were formed by cutting the syringes with a razor
`blade and extruding the gels. The gels were incubated at
`37°C with different concentrations of bovine testicular hyal-
`uronidase (Sigma), 50–5000 U/mL, in 400 mL of 30 mM citric
`acid, 150 mM Na2HPO4, pH 6.3, containing 150 mM NaCl,
`for the indicated time (0–48 h). Degradation of the gels was
`determined from the release of glucuronic acid into the su-
`pernatant as measured by the carbazole assay.26
`
`Rat subcutaneous implantation
`
`Biomaterials were implanted in 4- to 5-week-old male
`Sprague-Dawley rats. Briefly, a small vertical incision was
`made on either side of the xiphoid cartilage of the sternum,
`and the skin undermined with a blunt instrument to sepa-
`rate the skin from the underlying tissue. Biomaterial discs
`(10 mm in diameter, 3 mm width) were placed into these
`pockets and the skin incisions closed with sutures. For each
`test group, four to six biomaterial specimens were im-
`planted. Animal experiments were performed according to
`NIH Guidelines for the Care and Use of Laboratory Animals
`(NIH Publ. No. 85-23, Rev. 1985).
`Hyaluronic acid hydrogel discs were prepared by
`crosslinking HA derivatives (12–14 mg/mL) in PBS in 3-mL
`syringes as described above. The HA solution was supple-
`
`mented with cell adhesion molecules such as collagen type I
`fibrils (1 mg/mL) or fibronectin (500 mg/mL) prior to
`crosslinking when desired to promote cell infiltration. Col-
`lagen fibrils were prepared by polymerization from dilute
`solution (2–3 mg/mL) of acid-solubilized intact bovine col-
`lagen type I (retaining telopeptides; Organogenesis, Canton,
`MA) in PBS and harvested by centrifugation, following stan-
`dard protocols.27 For induction of chondro-osseous differ-
`entiation, different growth factors including bone morpho-
`genetic protein-2 (BMP-2) (200 mg/mL; Genetics Institute,
`Cambridge, MA), insulin-like growth factor-1 (IGF-1) (500
`ng/mL; Celtrix Pharmaceuticals, Santa Clara, CA), and
`transforming growth factor-b2 (TGF-b2) (50 ng/mL; Celtrix)
`were mixed with the HA solution just prior to crosslinking.
`For inhibition of vascularization, 10 mg/mL suramin
`(Sigma) was added.
`At the indicated time postoperatively, the animals were
`euthanized, and the implants were exposed through a dorsal
`midline skin incision and excised with the attached sur-
`rounding tissue. The tissue was fixed in 10% formalin in
`PBS, dehydrated through a graded ethanol series, and em-
`bedded in paraffin. Five-micrometer sections were cut and
`stained with hematoxylin/eosin and Safranin-O/Fast green.
`
`RESULTS AND DISCUSSION
`
`We focused on the production of hydrogels from
`HA, since HA matches several of the desired proper-
`ties for a biomaterial for delivery of bioactive agents
`such as cells, growth factors, cytokines, and drugs for
`tissue repair. It is biodegradable, provides an excellent
`substratum for cell migration, and, most important,
`has proven its biocompatibility in various forms in
`clinical practice (for discussion, see Balazs and Laur-
`ent.8) The relatively simple repetitive structure of HA
`allows also for specific modification and introduction
`of a large number of functional groups for subsequent
`crosslinking (Fig. 1).
`
`Preparation of HA derivatives
`
`Initial studies
`
`It is well known that sugars can be oxidized using
`periodate,28 and initially, we followed standard pro-
`cedures to generate polyaldehydes from HA by per-
`iodate oxidation. Periodate treatment oxidizes the
`proximal hydroxyl groups (at C2 and C3 carbons of
`glucuronic acid moiety) to aldehydes, thereby opening
`the sugar ring to form a linear chain. While periodate
`oxidation allows for the formation of a large number
`of functional groups, the disadvantage is the loss of
`the native backbone structure. Consequently, the gen-
`erated derivative is presumably not recognized as HA
`by cells. Indeed, hydrogels formed by using periodate-
`oxidized HA as a crosslinker, e.g., in combination with
`the HA-amine derivatives described below, showed
`very limited tissue transformation and poor cellular
`infiltration in the rat ectopic bone formation assay (see
`
`

`

`158
`
`BULPITT AND AESCHLIMANN
`
`below) (Table I). Clearly, periodate oxidation of HA
`was not suitable and our goal had to be to generate an
`activated form of HA that differs minimally from na-
`tive HA to conserve its unique physicochemical prop-
`erties. Also, a minimal change affecting only a rela-
`tively small number of dissaccharide units would pre-
`sumably not alter its property to serve as a cell
`substratum, since short HA oligosaccharides are rec-
`ognized by HA receptors.6 Since deacetylation of the
`N-acetylglucosamine moiety leads to concurrent deg-
`radation of the HA chain19,20 [Fig. 4(A)], our efforts
`focused on the carboxylate group of the glucuronic
`acid moiety (Fig. 1). Initially, attempts were made to
`generate an aldehyde derivative by reduction of the
`carboxyl groups on the glucuronic acid residues using
`9-borabicyclo-3,3-nonane, a method that allows direct
`conversion of the carboxylic acid into an aldehyde.29
`However, this avenue of investigation was unsuccess-
`ful, and we decided to introduce an aldehyde onto HA
`by means of a side chain using carbodiimide-mediated
`coupling.
`
`Introduction of a (functionalized) side chain
`onto HA
`
`Direct carbodiimide-mediated coupling of amines
`to the carboxyl group of HA in an aqueous environ-
`ment, e.g., with 1-ethyl-3-[3-dimethylaminopropyl]
`carbodiimide (EDC), did not yield the predicted prod-
`uct since the O-acyl isourea that is formed as a reactive
`intermediate rearranges rapidly to a stable N-acyl urea
`(Fig. 2). This results in predominant coupling of EDC
`(>99%) as indicated by the 1H-NMR spectrum which
`
`showed the characteristic sharp singlet at d = 2.08 cor-
`responding to the N-methyl protons and the methy-
`lene protons of the N-ethyl methyl group at d = 1.0,
`which is consistent with previous reports.22,23 We
`found that the coupling of primary amines to HA is
`possible via the formation of a more hydrolysis resis-
`tant and non-rearrangable active ester intermediate
`(Fig. 2). We formed active esters of HA with 1-hy-
`droxybenzotriazole (HOBt) or N-hydroxysulfosuccin-
`imide (Sulfo-NHS) using the water-soluble carbodi-
`imide EDC for coupling. Nucleophilic addition to the
`ester formed from HOBt requires the amine to be pre-
`sented in an unprotonated form at acidic pH (about
`5.5–7.0). Only a limited number of amines including
`hydrazides and “activated” amines, e.g., ethylene di-
`amine, have pKa values in a suitable range and are
`consequently relatively unprotonated and reactive
`with the ester-intermediate formed with HOBt (Fig. 2:
`3–6). Simple primary amines, e.g., putrescine, which
`have typically pKa values > 9 do not form significant
`amounts of adduct under acidic coupling conditions.
`The Sulfo-NHS-derived intermediate allows for the
`coupling reaction to be carried out at neutral or
`slightly basic pH (about 7.0–8.5) and consequently
`yields products by reaction with simple primary
`amines (Fig. 2: 7–9).
`
`Preparation of HA-aldehyde derivatives
`
`A heterobifunctional side chain, N-(2,2-dimeth-
`oxyethyl)-4-(hydrazido) butanamide (2), containing a
`protected aldehyde in the form of an acetal and an acyl
`hydrazide for coupling was synthesized using stan-
`
`TABLE I
`Evaluation of Biological Properties of Different HA Hydrogels in Rat Subcutaneous Implantation Model
`
`Crosslinker
`
`Granulomatous
`Inflammation*
`
`Cell Infiltration†
`
`Chondro-osseous
`Differentiation‡
`
`Group
`
`1
`2
`3
`4
`5
`6
`7
`
`8
`
`HA Derivative
`~ 25% AD-HA
`~ 25% AD-HA
`~ 25% AD-HA
`~ 65% AD-HA
`~ 25% AD-HA
`~ 25% AD-HA
`~ 25% AD-HA
`~ 25% AD-HA
`
`+++
`+
`+
`+
`+++
`+
`++
`
`+/++
`
`−/+
`−
`++
`+
`+
`+/++
`+++
`
`++
`
`−
`−
`+++
`+/++
`−
`+/++
`+
`
`++
`
`Glutaraldehyde
`DTSSP
`(SPA)2-PEG
`(SPA)2-PEG
`Periodate-oxidized dextran
`Periodate-oxidized HA
`~ 10% aminoacetaldehyde
`dimethyl acetal-modified HA
`~ 65% N-(2,2-dimethoxyethyl)-
`4-(hydrazido)butanamide-
`modified HA
`~ 25% LME-HA
`9
`(SPA)2-PEG
`+/++
`+++
`++
`HA hydrogels were formed by crosslinking 12 mg/mL of the HA-amine derivatives (adipic dihydrazide-[AD-HA] or lysine
`methylester-[LME-HA] modified HA) with a slight excess of crosslinker (groups 1–4 and 9) or by mixing polysaccharide
`derivatives with different modifications such as to yield the same final polysaccharide concentration (groups 5–8), as de-
`scribed in Materials and Methods. Hydrogels were supplemented with prefibrillized collagen type I and BMP-2 to stimulate
`endochondral bone formation (see Table II).
`*+ = mild inflammation with mononuclear cells; ++ = moderate with occasional foreign-body giant cells; +++ = severe with
`abundant presence of foreign-body giant cells.
`†+ = <10% of implant infiltrated; ++ = >30%; +++ = >80%.
`‡+ = occasional foci of cartilage/bone; ++ = thin layer surrounding implant; +++ = >30% of implant replaced.
`
`

`

`IN SITU POLYMERIZABLE HYALURONIC ACID HYDROGEL MATERIALS
`
`159
`
`Figure 4. Size distribution of derivatized HA. HA preparations were separated on a Superose 6 FPLC column in PBS and
`the absorbance at 205 nm was recorded. (A) HA (MW > 106) was N-deacetylated by hydrazinolyses for the indicated time
`(0–180 min). (B) HA was derivatized with lysine methyl ester (6) or putrescine (8) via a benzotriazole or sulfosuccinimide ester
`intermediate, respectively. (C) HA and HA derivatives were digested with 100 U/mL of testicular hyaluronidase for 2 h (see
`next page). The void volume (v0) and total volume (vT) of the column are indicated by arrows. The elution of chondroitin
`sulfate (CS) with a molecular mass of ~ 45 kDa is indicated in (A).
`
`dard chemistry as detailed in Materials and Methods.
`The side chain was coupled to HA as outlined in Fig-
`ure 2 (3). High-molecular-weight HA (MW > 106) was
`dissolved in water, and a 20-fold molar excess of the
`
`side chain was added to ensure efficient coupling.
`Next, a fourfold molar excess of HOBt was added and
`the pH adjusted to 6.8. Finally, a fourfold molar excess
`of EDC was added to the reaction mixture, which re-
`
`

`

`160
`
`BULPITT AND AESCHLIMANN
`
`sulted in a slowly progressing decrease in pH. The pH
`was maintained at 6.8 for at least 4 h by the addition of
`0.1M NaOH and the reaction was allowed to proceed
`overnight. The progressing slight decrease in pH pre-
`sumably reflects the consumption of the basic primary
`amine over time since the reaction does not result in a
`net change in protons. This is in sharp contrast to the
`rapid pH increase observed when HA is reacted with
`EDC alone, which has been attributed to the net con-
`sumption of a proton,23 and suggests that the reaction
`proceeds through the active ester intermediate.
`A side chain with protected aldehyde functionality
`on one end and a primary amine for coupling on the
`other end, aminoacetaldehyde dimethyl acetal, was in-
`corporated into HA in a similar manner (Fig. 2: 7).
`HOBt was replaced with a fourfold molar excess of
`Sulfo-NHS and the pH was adjusted to 7.5. Upon ad-
`dition of EDC, a similar drop in pH was observed. The
`pH was maintained at 7.5 and the reaction was al-
`lowed to proceed overnight.
`The HA derivatives were purified by exhaustive di-
`alysis against water followed by repeated ethanol pre-
`cipitation to remove unreacted amine and other small
`reaction products. 1H-NMR confirmed that the modi-
`fication was successful, with the characteristic appear-
`ance of the acetal protons at d = 3.30 [Fig. 3(B)]. The
`HA-acetal derivatives were easily activated to the re-
`active aldehydes by mild acid treatment, and when
`mixed with dihydrazides or dihydrazide-modified
`HA (see below) produced hydrogels, further confirm-
`ing that successful modification had been achieved.
`
`Preparation of HA-amine derivatives
`
`Introduction of homobifunctional amines, i.e., eth-
`ylene diamine, lysine methyl ester, histidine methyl
`ester, and adipic, succinic, or suberic dihydrazide,
`which contain at least one amino group with a pKa <
`8.0, onto HA was conducted in a similar manner using
`HOBt and EDC (Fig. 2: 4–6). The degree of modifica-
`tion could be controlled between ~ 10% and 25% for
`amines and ~ 10% and 70% for hydrazides by adjusting
`the molar equivalency ratio of HA:EDC from 1:0.5 to
`1:4. The HA derivatives were purified as described
`above and analyzed by 1H-NMR. The spectra revealed
`that modification was successful. For example, the HA
`modified with lysine methyl ester showed the charac-
`teristic singlet at d = 3.85 corresponding to the methyl
`protons of the methyl ester group and the signal at d =
`3.0 corresponded to the methylene protons adjacent to
`the amino group [Fig. 3(C)]. The remaining methylene
`protons appeared between d = 1.4 and 2.0, and
`showed the expected characteristic splitting patterns.
`Homobifunctional amines having amino groups with
`pKa > 9.0, i.e., 1,4-diaminobutane (putrescine) and 1,6-
`diaminohexane, were coupled to HA using Sulfo-NHS
`and EDC (Fig. 2: 8,9). In this case, the degree of modi-
`fication with a fourfold excess of EDC was ~ 10% as
`assessed by 1H-NMR. The ability of these HA deriva-
`tives (4–6,8,9) to form hydrogels with amino group
`specific crosslinkers such as aldehydes and

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