throbber
--…… REVIEW
`
`Promoting the formation of new collateral vessels in ischemic tissues using angiogenic growth factors (therapeutic
`angiogenesis) is a an exciting frontier of cardiovascular medicine. Conversely, inhibition of the action of key
`regulators of angiogenesis, such as VEGF, constitutes a promising approach for the treatment of solid tumors and
`intraocular neovascular. syndromes. These concepts are being tested now in clinical trials.
`
`Clinical applications of angiogenic growth factors
`and their inhibitors
`
`NAPOLEON£ FERRARA1 &
`KARI ALITAL02
`
`In embryos, blood vessels form through
`two distinct processes, vasculogenesis
`in(cid:173)
`and angiogenesis. Vasculogenesis
`valves the de novo differentiation of en-
`in
`from mesodermal precursors, whereas
`dothelial cells
`angiogenesis new vessels are generated from pre-existing ones1.
`Vasculogenesis takes place only during embryonic develop(cid:173)
`men t and leads to the formation of a primary vascular plexus.
`Later these rather uniformly sized endothelial channels are re(cid:173)
`modeled into a mature system consisting of a tree-like hierar(cid:173)
`chy of large and small vessels. New capillaries then form
`I through angiogenesis, either by sprouting or by splitting (in(cid:173)
`I tus susc빡ion) from their vessels of origin. In adults, angiogen(cid:173)
`esis is essential for the female reproductive cycle, and for repair,
`remodeling and regeneration of tissues, for exa~ple during
`wound healing2. Neovascularization is also important in patho(cid:173)
`logical processes such as tumor growth and metastasis2.
`The known endothelial cell specific growth factors and theu
`receptors can be classified into vascular endothelial growth fac(cid:173)
`tor (VEGF) and angiopoietin (Ang) families3 (Fig. 1).Among the
`various angiogenic factors, VEGF is probably the most essen(cid:173)
`tial for the development and differentiation of the vascular
`system4. Loss of a single VEGF allele results in embryonic
`leth ality5·6 (Fig. 2) . Even selective inactivation of the heparin(cid:173)
`binding isoforms of VEGF, leaving one functional isoform
`(VEGF120), is insufficient for the proper development of the
`cardiovascular system and results in myocardial ischemia and
`perinatal or early postnatal lethality7. Also, other angiogenic
`factors, such as FGFs may work more indirectly, some of them
`through the VEGFs and their receptors8, so that a thorough
`knowledge of the signal transduction pathways of VEGFs and
`angiopoietins is essential for their use in therapeutic settings.
`
`Therapeutic angiogenesis and inhibition of arterial restenosis
`An exciting frontier of cardiovascular medicine is therapeutic
`angiogenesis. Promoting the formation of new collateral ves(cid:173)
`sels on the ischemic myocardium, leg muscles and other tissues
`would have an important effect on the treatment of disorders
`for which pharmacological intervention has been ineffective in
`controlled trials and for which therapy is now limited to surgi(cid:173)
`cal revascularization or endovascular interventional therapy9 .
`Several angiogenic molecules have been tested in animal
`models, including bFGF, aFGF, FGF-5, VEGF isoforms, VEGF(cid:173)
`C, HGF/SF and Ang-l/Ang-2. The factors tested most exten(cid:173)
`sively are VEGF and bFGF. In some cases, the recombinant
`protein was tested. In others, gene transfer using naked DNA
`or adenoviral vectors was used. A single intra-arterial adminis(cid:173)
`tration of 500-1000 µg of rhVEGF165 augmented perfusion and
`in a rabbit model of
`development of collateral vessels
`hindlimb ischemia in which the femoral artery was surgically
`removed10. Similar results were obtained in the same model
`
`NATURE MEI기 CINE • VOLUME 5 • NUMBER 12 • DECEMBER 1999
`
`with intramuscular or intra-arterial ad(cid:173)
`ministration of aFGF, bFGF, HGF/SF and
`VEGF-C (refs. 11-14). VEGF administra-
`tion after removal of the femoral artery
`not only resulted in increased vascularization but also led to
`recovery of the normal endothelial reactivity to various media(cid:173)
`tors15. Arterial gene transfer with cDNA encoding VEGF iso(cid:173)
`forms also led to revascularization to an extent comparable to
`that achieved with the recombinant protein 16. Moreover, ad(cid:173)
`ministration of a VEGF165 adenovirus vector shortly after com(cid:173)
`mon iliac artery ligation in the rat was capable of stimulating
`an angiogenic response that protects against subsequent oc(cid:173)
`clusion of the femoral artery, indicating that gene transfer of
`VEGF might be useful in the prophylaxis of advancing arterial
`occlusive disease1 7. As little as 2 µg rhVEGF delivered over 4
`weeks periadventitially, distal to the occlusion, resulted in a
`significant increase in coronary blood flow and functional im(cid:173)
`provement in a pig model of chronic myocardial ischemia18.
`Very similar results were obtained using bFGF (ref. 19).
`Unexpectedly, even a single intracoronary administration of
`VEGF (or bFGF) was efficacious in this model to an extent
`comparable to that of 4-week infusion, despite the fact that
`only a small fraction of protein localizes to the ischemic area20.
`Given such results, it is conceivable that young and otherwise
`healthy animals are very responsive to exogenous growth fac(cid:173)
`tors in the context of ischemia. At least some of this respon(cid:173)
`siveness may be due to the upregulation of VEGF receptors in
`the endothelia of ischemic tissues21 . Adenovirus-mediated
`gene transfer of VEGF121 (ref. 22) or FGF-5 (ref. 23) also resulted
`in collateral vessel growth and functional improvement in
`porcine models of myocardial ischemia.
`These encouraging animal studies led to clinical trials using
`recombinant VEGF165, aFGF, bFGF or gene therapy with plas(cid:173)
`mid or with adenoviral vectors. There is considerable debate
`whether gene therapy or administration of recombinant pro(cid:173)
`tein would be preferable. Delivery of angiogenic proteins by
`gene therapy might not only minimize their systemic side ef(cid:173)
`fects, such as hypotension (VEGF) or nephrotoxicity (bFGF),
`but also provide a slow release of the encoded factor for 1-2
`lasting angiogenic response.
`to a more
`leading
`weeks,
`However, slow release of the recombinant protein, using mi(cid:173)
`crospheres or heparin-alginate formulations, might achieve
`the same results, without the potential risks associated with
`the use of viral vectors.
`Arterial gene transfer of naked plasmid DNA encoding
`VEGF1 65 in a patient with severe limb ischemia produced angio(cid:173)
`graphic and histologic evidence of angiogenesis in the knee,
`mid-tibial and ankle levels 4 weeks after the transfer24. In a sub(cid:173)
`sequent study, the VEGF165 plasmid cDNA was injected intra(cid:173)
`muscularly25. Gene transfer was done in ten limbs of nine
`patients with nonhealing ischemic ulcers and/or rest pain due
`
`1359
`
`Mylan Exhibit 1045
`Mylan v. Regeneron, IPR2021-00880
`Page 1
`
`

`

`wall was not very efficient35, but secreted proteins such as VEGF
`could be used for therapeutic gene transfer trials using infu(cid:173)
`sion-perfusion catheters36 or histamine-induced increase of en(cid:173)
`dothelial permeability37. Because VEGF and VEGF-C share one
`receptor (VEGFR-2) but differ in the other receptor, VEGF-C
`and VEGF1 65 might have overlapping but distinct effects in the
`vessel wall. However, VEGF-C gene transfer inhibits intimal
`thickening early, and the protective effect is at least equal to
`that seen with VEGF165 gene transfer38.
`
`inhibition of vascular endothelial growth factor
`
`Therapeuti[
`Tumors
`The growth of tumor xenografts in transparent chambers in
`mice is preceded by an increase in vascular density, indicating
`that the rapid growth of tumors depends on the development
`of a neovascular supply39. In 1971, inhibition of angiogen esis
`was proposed as a valid strategy for the treatment of solid tu(cid:173)
`mors and the search for the mediator(s) of tumor angiogen esi s
`was begun얘
`Although inhibition of bFGF (ref. 41) or angiopoietin/Tie2
`(refs. 42,43) may inhibit tumor growth, so far VEGF and its re(cid:173)
`ceptors constitute the most extensively investigated system in
`tumor angiogenesis and are now a main target of anti-cancer
`strategies. VEGF mRNA is substantially upregulated in most
`human tumors4. Although tumor cells represent the main
`source of VEGF, tumor-associated stroma is also an important
`site of VEGF production44. There is a correlation between VEGF
`expression and microvessel density in primary breast cancer
`sections45. A similar correlation has been described in several
`other malignancies,
`including
`gastric
`carcinoma46.
`Furthermore, there are increases in plasma levels of VEGF in
`tumor patients compared with tumor-free individuals, and
`high VEGF levels before chemotherapy are associated with a
`poor outcome'‘7
`Direct evidence for involvement of VEGF in tumorigenesis
`was first demonstrated using monoclonal antibodies against
`
`따 따
`
`다 /
`
`·%
`
`VE-
`Cadhenn
`
`따
`
`따
`
`νk
`
`따 順
`
`때
`
`Pll 14L
`
`Mig 「ation, permeability, DNA synthesis, survival
`
`∼、 ι- 「~r
`
`ang1ogenes1s
`
`Lymphang1ogenes1s
`
`REVIEW······
`
`to peripheral arterial disease. Improvement in the ankle(cid:173)
`brachial index and distal flow in eight limbs were reported25.
`Additional small trials by the same group have also shown that
`local injection of the VEGF1 65 plasmid DNA resulted in clinical
`improvement in patients affected by myocardial ischemia26 or
`Burger's disease (thromboangiitis obliterans)27. However, none
`of these studies were placebo-controlled. Clinical trials using
`VEGF-C naked DNA or adenovirus mediated gene transfer of
`VEGF1 21 in myocardial ischemia patients are now in phase I.
`Femoral angiograms from a patient with limb ischemia, before
`and 3 months after transfection of a VEGF1 65 plasmid/liposome
`expression vector, show increased vascular density after the
`treatment (Fig. 3). However, the trial is ongoing and some cau(cid:173)
`tion should be used in interpreting such data, until more pa(cid:173)
`tients and the effect of placebo are more extensively evaluated.
`Clinical trials using recombinant VEGF1 65 and bFGF are also
`ongoing. In a phase I study in patients with coronary ischemia
`in which rhVEGF1 65 was administered by intracoronary infu(cid:173)
`sion, the molecule was safely tolerated at all doses tested28.
`There was evidence of improvement in perfµsion in seven of
`fifteen subjects and improved collateralization in five of seven
`who underwent follow-up coronary angiography. However, a
`subsequent placebo-controlled phase II study,
`in which
`rhVEGF was delivered as a single intracoronary infusion, fol(cid:173)
`lowed by three intravenous infusions, has not demonstrated
`clinical benefit29. The treatment was not better than placebo in
`treadmill time and pain relief, at least at 60 days29. Brief expo(cid:173)
`sures to rh VEGF1 65, such as those achieved in this trial, may be
`insufficient to trigger and maintain a therapeutically meaning(cid:173)
`ful angiogenic response, especially in the context of extensive
`atherosclerotic disease. Also, systemic administration of
`rhVEGF1 65 or other factor may fail to generate an appropriate
`angiogenic concentration gradient from ischemic to non-is(cid:173)
`chemic areas, a requisite aspect of angiogenesis in a variety of
`physiological and path이ogical circumstances1. Moreover, the
`placebo effect is probably greater than initially suspected, and
`even patients with very compromised myocar-
`dial function may show a substantial improve(cid:173)
`ment with placebo. A phase II study with bFGF
`for coronary ischemia is now ongoing.
`Local gene transfer into the vascular wall of(cid:173)
`fers a promising alternative for the treatment
`of the complication of restenosis after percuta(cid:173)
`neous transcoronary angioplasty (PTCA) and
`coronary stenting. Restenosis occurs in many
`treated patients in 6 months, leading to ob-
`struction in 20-35% of the patients30. The
`pathogenesis of restenosis depends on en-
`dothelial damage, which also predisposes ar-
`teries to other pathological conditions, such as
`spasms or thrombosis. Prophylaxis of resteno-
`sis could therefore be based on strategies for
`endothelial protection or enhancement of en-
`dothelial repair and endothelial growth factors
`or vascular gene transfer could be used for
`this31. Re-endothelization in balloon-injured
`rat carotid artery was accelerated by a single
`dose of recombinant VEGF injected into the
`bloodstream or locally32·33. Vessel status was
`also improved by injection of VEGF plasmid
`into adventitial surface of rabbit carotid arter(cid:173)
`ies34. Intravascular gene transfer in the arterial
`
`g 잉R 淑
`
`Fig. 1 VEGFs, thei 「 「eceptors and some of their endothelial effects in cells and tissues.
`Ligand binding induces recepto 「 dime「ization and subsequent auto/transphosphorylation,
`activates various signal transduction pathways and leads to differential cellular responses.
`sVEGFR-1, soluble VEGFR-1; HSPG, heparan sulphate proteoglycan; NP-1, neuropilin-1 ; avbi,
`integrin avb3 (reported to make a molecular complex with activated VEGFR-2; ref. 95). VE(cid:173)
`cadherin is also able to form a complex with VEGFR-2, a requirement for VEGF-dependen~
`anti-apoptotic signals involving the Pl3-kinase/Akt pathway96. Pl l 14L, point mutation 01
`VEGFR-3 affecting patients in a family with lymphoedema97
`
`1360
`
`NATURE MEI기〔INE • VOLUME 5 • NUMBER 12 • DECEM BER 1999
`
`Mylan Exhibit 1045
`Mylan v. Regeneron, IPR2021-00880
`Page 2
`
`

`

`,.:---
`
`Fig. 2 Yolk sac of El 0.5 VEGF+i+ and VEGF +/- mouse embryos5. There is
`an apparent absence of vasculature in the yolk sac of the heterozygous,
`wh ich die around Ell. This is probably the only example among verte(cid:173)
`brates of lethality after inactivation of a single allele of a gene that is not
`maternally imprinted
`
`VEGF in human xenografts in nude mice48. These initial studies
`: showed that several tumor cell lines can be substantially
`I growth-inhibited by this treatment샌 These findings were ex(cid:173)
`tended to a broad variety of tumor cell lines, including carcino(cid:173)
`mas, sarcomas and gliomas4. Intravital videomicroscopy
`techniques have augmented our understanding of VEGF in tu(cid:173)
`imaging of the vasculature
`morigenesis49·50. Non-invasive
`dem onstrated a nearly complete suppression of tumor-associ(cid:173)
`ated angiogenesis in animals treated with monoclonal antibod(cid:173)
`ies against VEGF compared with controls, providing a direct
`verification that inhibition of angiogenesis is the mechanism
`of tumor suppression after anti-VEGF treatment49. Intravital
`microscopy techniques have also been used to investigate the
`effects of VEGF on the permeability and other properties of
`tumor vessels50. Treatment with antibodies against VEGF re(cid:173)
`sulted in time-dependent reductions in vascular permeability,
`in the diameter and tortuosity and eventually to a regression of
`tumor blood vessels; thus, VEGF is also an essential survival fac(cid:173)
`tor for tumor endothelial cells50. Further evidence that VEGF
`action is required for tumor angiogenesis has been provided by
`the finding that retrovirus-mediated expression of a dominant
`negative VEGFR-2 mutant, which inhibits signal transduction
`through wild-type VEGFR-2 receptor, suppresses the growth of
`glioblastoma multiforme as well as other tumor cell lines in
`vivo51. Furthermore, high local expression of the soluble extra(cid:173)
`cellular domain of VEGFR-1 or VEGFR-2, achieved by adminis(cid:173)
`tration of the recombinant proteins, adenoviral-mediated gene
`transfer or by stable transfection of tumor cells, may signifi(cid:173)
`cantly inhibit tumor growth, metastasis and mortality rate in
`nude mice52·53.
`Several strategies have been used to generate VEGF inhibitors
`suitable for clinical trials. One approach involves the ‘human(cid:173)
`ization’ of mouse monoclonal antibodies. A chief advantage of
`‘humanized' antibodies is a high degree of specificity, com(cid:173)
`bin ed with a long half-life and little or no immunogenicity. A
`1humanized’ high-affinity monoclonal antibody against VEGF
`(rhuMAb VEGF) with the same affinity and biological proper-
`ties as the original murine antibody has been described54.
`Toxicological studies in primates have shown that the effects of
`rhuMAb VEGF are limited to inhibition of angiogenesis in the
`female reproductive tract and in the epiphyseal growth plate in
`
`NATURE MEI기〔 INE • VOLUME 5 • NUMBER 12 • DECEMBER 1999
`
`·····REVIEW
`
`sexually immature animals that have not completed statural
`growth55. rhuMAb VEGF is now in phase II clinical trials for the
`treatment of non-small cell lung carcinoma and colorectal car(cid:173)
`cinoma in conjunction with standard chemotherapy and for
`breast and renal cell carcinoma as a single agent. In addition,
`small molecules that inhibit VEGFR-2 signal transduction are
`in cancer patients56.
`trials
`undergoing phase II clinical
`Furthermore, monoclonal antibodies against VEGFR-2 are en(cid:173)
`tering clinical trials.
`
`Retinal ischemia and other conditions
`Diabetes mellitus, occlusion of the central retinal vein or pre(cid:173)
`maturity with subsequent exposure to oxygen can all be associ(cid:173)
`intraocular neovascularization57. A common
`ated with
`denominator among these conditions is retinal ischemia57. The
`new blood vessels may lead to vitreous hemorrhage, retinal de(cid:173)
`tachment, neovascular glaucoma, and eventual blindness.
`Diabetic retinopathy is the leading cause of blindness in the
`working population. The hypothesis that ischemia-induced
`VEGF may be pathogenic in these conditions was initially
`tested by measuring VEGF levels in the eye fluids of patients. In
`a large series with 165 patients, a strong correlation was found
`between concentrations of VEGF in both aqueous and vitreous
`and active proliferative retinopathy associated with diabetes,
`occlusion of central retinal vein or prematurity58. Direct evi(cid:173)
`dence for the role of VEGF as a mediator of intraocular neovas(cid:173)
`cularization has been generated in several animal models,
`including a primate model of iris neovascularization and a
`mouse model of retinopathy of prematurity. In the former, in(cid:173)
`traocular administration of monoclonal antibodies against
`VEGF substantially inhibits the neovascularization that follows
`the occlusion of central retinal veins59. Likewise, soluble
`VEGFR-1 or VEGFR-2 extracellular domains fused to the im(cid:173)
`munoglobulin y Fc domain suppress retinal angiogenesis in the
`that growth
`also evidence
`is
`mouse model60. There
`hormone/insulin-like growth factor-1 is involved in ischemia(cid:173)
`induced retinal neovascularization61.
`Neovascularization is a principal cause of visual loss also in
`the wet form of age-related macular degeneration (AMD), the
`overall leading cause of blindness62. Several studies have docu(cid:173)
`mented the immunohistochemical localization of VEGF in sur(cid:173)
`gically resected choroidal neovascular membranes from AMD
`patients63. These findings suggest involvement of VEGF in the
`progression of AMD-related choroidal neovascularization.
`Anti-VEGF strategies for AMD are now being explored in clini(cid:173)
`cal trials. One approach consists in the intravitreal administra(cid:173)
`tion of a recombinant humanized anti-VEGF Fab antibody
`fragment. Another strategy involves the injection of 2’-fluo(cid:173)
`ropyrimidine RNA 이igonucleotide ligands (aptamers)64.
`VEGF inhibition may also have therapeutic value for the
`treatment of ischemic-reperfusion related brain edema and in(cid:173)
`jury. VEGF antagonism has shown beneficial effects in a mouse
`model of cortical ischemia65; reducing acutely the volume of
`edematous tissue and resulting in a significant sparing of corti(cid:173)
`cal tissue.
`VEGF is important in angiogenesis in the female reproduc(cid:173)
`tive tract. VEGF inhibition results in suppression of corpus lu(cid:173)
`teum angiogenesis in rodents66 and primates55. VEGF inhibitors
`might be used to treat conditions characterized by ovarianhy(cid:173)
`perplasia and hypervascularity, such as the polycystic ovary
`syndrome66. VEGF-dependent angiogenesis may also be impor(cid:173)
`tant pathogenically in endometriosis. Furthermore, VEGF is a
`
`1361
`
`Mylan Exhibit 1045
`Mylan v. Regeneron, IPR2021-00880
`Page 3
`
`

`

`REVIEW
`
`...... ..
`
`mediator of the ovarian growth and increased vascular perme(cid:173)
`ability of ovarian hyperstimulation syndrome, a potentially
`fatal condition characterized by massive ovarian enlargement
`that may follow medical induction of ovulation with go(cid:173)
`nadotropins67.
`
`Perspectives
`VEGF165 binds to neuropilin-1, which functions as a ligand
`binding subunit of putative transmembrane receptors mediat(cid:173)
`ing specific signals for different semaphorins, the molecules
`mediating the collapse of axonal growth cones68. Neuropilin is
`expressed in endothelial cells and enhances the mitogenic ef(cid:173)
`fects of VEGFR-2 upon VEGF165 stimulation. Thus, there may be
`an as-yet ill-defined cross-regulation of cellular signals between
`these two families of factors. These findings lead to the intrigu(cid:173)
`ing conclusion that the processes of axon guidance and devel(cid:173)
`opment of a network of capillary tubes share at least some
`common molecular mechanisms. In addition, the angiopoietin
`receptor/Tie and ephrin families of endothelial tyrosine kinases
`have important functions in the formation and maintenance
`of the vascular system69.71. Endothelial cell-specific members of
`the TGF-~ receptor and Notch families have also been de(cid:173)
`scribed72·73. Given this complexity of vascular endothelial sig(cid:173)
`naling, therapies using VEGF alone or any other single
`angiogenic factor may produce incompletely functioning or
`unstable endothelial channels with defective arteriovenous
`and pericellular differentiation, characteristic of many tu(cid:173)
`mors74. Combinations of growth factors may be preferable in
`future therapies directed to neovascularization of tissues, with
`an adequate investment of the formed vessels with perien(cid:173)
`dothelial matrix and pericyte/smooth muscle cells. In fact, a
`more heterogenous set of genes coordinating angiogenic func(cid:173)
`tions may be provided by active ongoing research of hypoxia(cid:173)
`regulated gene expression in mammalian cells75. Also, some
`virus-encoded proteins, such as the VEGFR-2 activating HIV
`Tat protein76, Kaposi sarcoma herpesvirus-associated G-protein(cid:173)
`coupled receptor77 or Orf virus encoded VEGF-£78-80 may offer
`new insights into the mechanism of regulation of angiogenesis.
`Although recent research has focused on the combination of
`VEGF and Ang-1 as being especially promising, it is not known
`now which growth factor combinations will prove to be the
`most effective therapeutically. VEGF and bFGF have a very
`synergistic effect in the induction of angiogenesis, both in vitro
`and in vivo4. The interaction between VEGF and HGF/SF is also
`being actively investigated. Although transgenic expression of
`Ang-1 in the skin epidermis under the keratin (K)14 promoter
`has been associated with neovascularization81’ other studies,
`using defined amounts of the recombinant protein in a model
`of adult neovascularization, have failed to demonstrate strong
`angiogenic responses to Ang-1, unless it is used in combina(cid:173)
`tion with VEGF (refs. 71,82). This discrepancy may be ex(cid:173)
`plained by the fact that the expression of the K14 promoter is
`initiated already at midgestation, and thus the results may re(cid:173)
`flect persistence of the fetal neovascularization. It is possible,
`however, that Ang-1 may provide a co-factor for combination
`therapies. A further unresolved issue is the correct dosage of
`growth factor(s) . This seems particularly important for a mole(cid:173)
`cule like VEGF, which has several isoforms and such a tight
`dose-response effect that a 50% reduction in expression re(cid:173)
`sults in lethality during embryonic life5·6. Conversely, continu(cid:173)
`ous
`local overexpression of VEGF may
`result
`in a
`hemangioma-like vasculature and thus can be deleterious83.
`
`Also, it is unknown whether an angiogenic treatment m ay be
`sufficient to induce functional blood vessels for prolonged pe(cid:173)
`riods or will need to be re-administered periodically in order to
`maintain such vessels.
`A K14-driven VEGF-C transgene induced lymphangiogenesis
`but no angiogenesis in mouse skin84, and recombinant VEGF-C
`also stimulated lymphatic vessel hyperplasia in mature chick
`chorioallantoic membrane85. Thus, besides angiogenesis, it may
`also become possible to direct therapeutic lymphangiogenesis
`in patients, such as after evacuation of axillary lymph nodes in
`breast carcinoma surgery.
`Despite the potential redundancy of tumor angiogenesis fac(cid:173)
`tors, inhibition of VEGF alone seems sufficient to achieve con(cid:173)
`siderable tumor growth suppression in a wide variety of
`models. However, it remains to be established whether tumors
`are able to activate, after prolonged therapy, alternative angio(cid:173)
`genic pathways that might confer resistance to the treatment.
`These issues should be addressed in the current clinical trials
`with various VEGF inhibitors. A challenge now in anti-VEGF
`(and anti-angiogenic) therapy is devising appropriate and reli(cid:173)
`able markers to monitor tumor progression. There is consider(cid:173)
`able debate whether blood vessel count
`in biopsy
`specimens45·46 may provide a reliable indicator of response to
`the treatment. There are also efforts to identify surrogate end(cid:173)
`points, applying non-invasive approaches, such as magnetic
`resonance imaging86 .
`VEGF is not only a mitogen but also a potential survival fac(cid:173)
`tor for endothelial cells4. Such a 'maintenance’ function seems
`to be developmentally regulated, as it is very dependent on the
`age of the animal87. VEGF inactivation during early postnatal
`life, achieved by Cre-loxP-mediated inducible gene targeting of
`by administration of a soluble VEGFR-1 chimeric protein, re(cid:173)
`sults in regression of the vasculature, kidney failure and lethal(cid:173)
`ity87. However, in adult animals a similar treatment h as no
`effects on the existing vasculature. Therefore, a process of mat(cid:173)
`uration occurs in endothelial cells such that VEGF eventually is
`not essential for survival. This switch seems to take place in the
`mouse around the fourth postnatal week. Absence of pericyte
`
`Fig. 3 Angiography of the lower extremity of a patient with limb 15·
`chemia before (PRE) and 3 months afte「 (3 MO) the transfection of a
`VEG Fl 65 plasmid/liposome expression vector, showing strongly increaseu
`vascula 「 density after the treatment. Courtesy H. Manninen, P. Matsi, "·
`Makinen, M. Hilpelainen, M. Laitinen, E. Alhava and S. Yla-Herttuala, A. I.
`Virtanen Institute and Kuopio University Hospital (Kuopio, Finland).
`
`1362
`
`NATURE MEDI디 NE • VOLUME 5 • NUMBER 12 • DECE MB ER 1999
`
`Mylan Exhibit 1045
`Mylan v. Regeneron, IPR2021-00880
`Page 4
`
`

`

`… ·REVIEW
`
`~:erage in im
`dependence on VEGF (ref. 88). However, other evidence sug(cid:173)
`gests that the molecular/intracelullar nature of this switch may
`be more complex and mostly still to be determined87. In juve(cid:173)
`nile animals, VEGF is essential for endochondral bone forma(cid:173)
`tion and longitudinal growth89'55. In the fully developed
`anim al, VEGF may be required mainly for active angiogenic
`processes such as corpus luteum development or wound heal(cid:173)
`ing. Neverthless, VEGF may be important for endothelial
`I homeostasis in the adult in certain circumstances; for example
`during disease states. Indeed, prolonged VEGF inhibition failed
`to induce glomerular damage in normal primates55 or ro(cid:173)
`dents87·90, despite the strong constitutive expression of the
`VEGF mRNA in podocytes and other cell types in the adult kid(cid:173)
`ney4. However, administration of VEGF inhibitors to rats with
`mesangiopr이iterative nephritis results in impaired glomerular
`endothelial regeneration and increased endothelial cell death90.
`Some CD34+ hematopoietic progenitor cells mobilized by
`GM-CSF from human peripheral blood, bone marrow, fetal
`liver or umbilical cord blood were shown to express VEGFR-2
`on their surface91, and VEGFR-2 is expressed on human
`hematopoietic stem cells92. Endothelial progenitor cells expand
`and differentiate into endothelial cells after addition of bFGF
`and VEGF to the cultures, and they can thus be considered to
`provide endothelial progenitor cells91 93. The endothelial prog(cid:173)
`enitor cells from bone marrow may be mobilized using the stro(cid:173)
`mal-derived factor 1 chemokine, the GM-CSF cytokine or tissue
`hypoxia94. As these cells may be capable of participating in ac(cid:173)
`tive angiogenesis after entry into the circulatory system94, they
`provide an interesting possibility for the delivery of cellular or
`gene therapy to sites of neovascularization.
`Finally, the first placebo-controlled clinical study with
`rhVEGF may have brought a more realistic assessment of the
`potential of therapeutic angiogenesis and raised a number of
`questions. For example, how can one explain the discrepancy
`between the considerable efficacy observed even with very
`small amounts of growth factors in animal models of coronary
`or limb ischemia and the rather disappointing clinical results?
`An essential difference may lie in the fact that young and oth(cid:173)
`erwise healthy animals are able to mount an effective endoge(cid:173)
`nous angiogenic response that can be maximized by an
`additional stimulus provided by a recombinant protein or gene
`therapy. In contrast, patients with extensive atherosclerotic
`disease may have poor responses. It is possible, however, that a
`more persistent exposure to an individual growth factor or to a
`combination of growth factors may be effective. Clinical trials
`now ongoing should answer at least some of these questions
`over the next 2-3 years.
`
`1. Risau, W. Mechanisms of angiogenesis. Nature 386, 671 674 (1997).
`2. Folkman, J. Angiogenesis in cancer, vascular, rheumatoid and other diseases.
`Nature Med. 1, 27-31 (1995).
`3. Korpelainen, E.I. & Alitalo, K. Signaling angiogenesis and lymphang1ogenes1s
`Curr. Opin. Cell Biol. 10, 159 164 (1998).
`4. Ferrara, N . & Davis-Smyth, T. The biology of vascular endothelial growth factor.
`Endocr. Rev. 18, 4 25 (1997).
`5. Ferrara, N . et al. Heter。zygous embryonic lethality induced by targeted inact1va(cid:173)
`tion of the VEGF gene. Nature 380, 439-442 (1996) .
`6. Carmeliet, P. et al. Abnormal blood vessel development and lethality in embryos
`lacking a single VEGF allele. Nature 380, 435-439 (1996).
`7. Carmeliet, P. et al. Impaired myocardial angiogenesis and ischemic cardiomy(cid:173)
`opathy in mice lacking the vascula 「 endothelila growth factor isofo 「ms VEGF164
`and VEG Fl 88. Nature Med. 5, 495-502 (1999).
`8. Seghezzi, G. et al. Fibroblast growth factor-2 (FGF-2) induces vascular endothe(cid:173)
`lial growth factor (VEGF) expression in the endothelial cells of forming capillar(cid:173)
`ies: an autocrine mechanism contributing to angiogenesis. }. Cell Biol. 171,
`1659-1673 (1998)
`
`9. Rivard, A. & Isner」 .M . Angiogenesis and vascul 。genesi s in t「eatment of card10-
`vascular disease. Mo/. Med. 4, 429-440 (1998) .
`10. Takeshita, S. et al. Therapeutic angiogenesis. A single intra-arterial bolus of vas(cid:173)
`cul a 「 endothelial growth fact。r augments revascularizati 。 n in a rabbit ischemic
`hind limb model.}. Clin. Invest. 93, 662-670 (1994)
`11. Pu, L.Q. et al. Enhanced revascularization of the ischemic limb by ang1 。genic
`therapy. Circulation 88, 208-215 (1993) .
`12. Asahara, T. et al. Synergistic effect of vascular endothelial growth factor and
`basic fibroblast growth factor on angiogenesis in vivo. Circulation 92, 11365-11371
`(1995).
`1 3. Van Belle, E. et al. Potentiated angiogenic effect of scatter factor hepatocyte
`growth factor via induction of vascular endothelial growth factor: the case for
`paracrine amplification of angiogenesis. Circulation 97, 381-390 (1998).
`14 Witzenbichler, B. et al. Vascular endothelial 9 「owth factor-C (VEGF-C/VEGF-2)
`promotes angiogenesis in the setting 。f tissue ischemia. Am. }. Patho/‘ 153,
`381-394 (1998) .
`15. Bauters, C. et al. Physi이ogical assessment of augmented vascularity induced by
`VEGF in ischemic rabbit hindlimb. Am. j ‘ Physiol‘ 267, Hl 263-Hl 271 (1994).
`16. Takeshita, S. et al. Gene transfer of naked DNA encoding for

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