throbber
INSIGHT REVIEW
`
`NATURE|Vol 438|15 December 2005|doi:10.1038/nature04483
`
`Angiogenesis as a therapeutic target
`
`Napoleone Ferrara1 & Robert S. Kerbel2
`
`Inhibiting angiogenesis is a promising strategy for treatment of cancer and several other disorders,
`including age-related macular degeneration. Major progress towards a treatment has been achieved
`over the past few years, and the first antiangiogenic agents have been recently approved for use in several
`countries. Therapeutic angiogenesis (promoting new vessel growth to treat ischaemic disorders)
`is an exciting frontier of cardiovascular medicine, but further understanding of the mechanisms of
`vascular morphogenesis is needed first.
`
`Early pioneers of angiogenic research observed over a century ago that
`the growth of human tumours is often accompanied by increased vas-
`cularity. They suggested that a key aspect of the cancer process is a dis-
`ease of the vasculature in the whole area affected (reviewed in ref. 1).
`The existence of tumour-derived factors responsible for promoting
`new vessel growth was postulated over 65 years ago2, and a few years
`later it was proposed that tumour growth is crucially dependent on the
`development of a neovascular supply3. In 1971, it was hypothesized
`that inhibition of angiogenesis (antiangiogenesis) would be an effec-
`tive strategy to treat human cancer, and an active search for angiogen-
`esis inducers and inhibitors began4. Extensive research has led to the
`identification and isolation of several regulators of angiogenesis, some
`of which represent therapeutic targets.
`Despite some initial setbacks and negative clinical trial results,
`major progress has been made over the past few years in targeting
`angiogenesis for human therapy. In February 2004, the US Food and
`Drug Administration (FDA) approved bevacizumab, a humanized
`anti-VEGF (vascular endothelial growth factor)-A monoclonal anti-
`body, for the treatment of metastatic colorectal cancer in combination
`with 5-fluorouracil (FU)-based chemotherapy regimens. This fol-
`
`BMC
`
`PDGF–B
`
`c
`
`Pericyte
`
`Capillary bud
`
`Endothelial cells
`
`Other angiogenic
`factors
`such as bFGF
`
`VEGF-A
`
`BMC
`
`BMC
`
`VEGF-A
`
`Other angiogenic
`factors
`
`a
`
`Tumour cells
`
`SDF-1
`
`HGF
`TGF-α
`EGF
`
`PDGF-A
`PDGF-C
`TGF-β
`
`b
`Stromal cells
`
`lowed from a phase III study showing a survival benefit5. In December
`2004, the FDA approved pegaptinib, an aptamer that blocks the 165
`amino-acid isoform of VEGF-A, for the treatment of the wet (neovas-
`cular) form of age-related macular degeneration (AMD)6.
`These achievements have validated the notion that angiogenesis
`is an important target for cancer and other diseases. These advances
`notwithstanding, much progress is needed on a variety of important
`issues; for example, how do we achieve the most effective combina-
`tions of antiangiogenic agents with chemotherapy or other biologi-
`cal agents and how do we select patients that are most likely to
`respond to the treatment? Another issue is that resistance to antian-
`giogenic therapy is emerging7 and thus a better understanding of
`pathways that may mediate tumour angiogenesis in various cir-
`cumstances is necessary. Furthermore, the hope that ‘therapeutic
`angiogenesis’ will provide a treatment for ischaemic disorders still
`remains unfulfilled, in spite of considerable preclinical and clinical
`efforts.
`The main purpose of this review is to summarize recent progress and
`emphasize the issues that need to be resolved before the field of angio-
`genic therapy can make further significant advances.
`
`Figure 1 | A few of the molecular and cellular players in the
`tumour/microvascular microenvironment. a, Tumour cells produce VEGF-
`A and other angiogenic factors such as bFGF, angiopoietins, interleukin-8,
`PlGF and VEGF-C. These stimulate resident endothelial cells to proliferate
`and migrate. b, An additional source of angiogenic factors is the stroma.
`This is a heterogeneous compartment, comprising fibroblastic,
`inflammatory and immune cells. Recent studies indicate that tumour-
`associated fibroblasts produce chemokines such as SDF-1, which may
`recruit bone-marrow-derived angiogenic cells (BMC). The various
`hypotheses on the nature and role of such cells in angiogenesis and tumour
`progression are discussed in the text. VEGF-A or PlGF may also recruit
`BMC. Tumour cells may also release stromal cell-recruitment factors, such
`as PDGF-A, PDGF-C or transforming growth factor (TGF)-ȋ. A well-
`established function of tumour-associated fibroblasts is the production of
`growth/survival factor for tumour cells such as EGFR ligands, hepatocyte
`growth factor and heregulin. c, Endothelial cells produce PDGF-B, which
`promotes recruitment of pericytes in the microvasculature after activation
`of PDGFR-ȋ. HGF, hepatocyte growth factor.
`
`1Genentech, 1 DNA Way, South San Francisco, California 94080, USA; 2Sunnybrook and Women’s College Health Sciences Centre and the University of Toronto, Ontario M5G 2M9, Canada.
`
`© 2005
`
`Nature Publishing Group
`
`© 2005 Nature Publishing Group
`
`967
`
`CELLTRION - EXHIBIT 1046
`
`

`

`INSIGHT REVIEW
`
`NATURE|Vol 438|15 December 2005
`
`Chemotherapy drug targets
`
`Tumour cells
`
`Bone-marrow
`progenitors
`
`Hair follicle
`cells
`
`Epithelial
`mucosal
`cells
`
`Tumour shrinkage/
`response
`
`Myelo-suppression,
`for example,
`neutro-penia
`
`?
`
`Drop in pro-
`angiogenic
`monocytes
`and pericyte
`precursors
`
`a
`
`Antiangiogenic
`effect
`
`Alopecia
`
`Mucositis
`
`Dividing
`endothelial
`cells in
`sprouting
`vessels
`
`Endothelial
`progenitor
`cells (EPCs)
`in bone marrow
`or peripheral
`blood
`
`b
`
`c
`
`Antiangiogenic
`effect
`
`Antiangiogenic
`effect
`
`Figure 2 | Chemotherapy targets. In addition to tumour cells, the intended target for chemotherapy in cancer patients, conventional chemotherapy drugs
`can inhibit the proliferation of, or kill, a number of normal host cell types, including several that, in principle, can contribute to an antiangiogenic effect.
`Targeting of various normal cell populations is generally associated with harmful or undesirable side effects such as myelosuppression, alopecia or
`mucositis. A desirable effect could be antiangiogenesis as a result of targeting. a, Bone-marrow-derived proangiogenic cells that adhere to the walls of new
`blood vessels and further stimulate their growth by paracrine mechanisms. Whether these latter cell types, which probably include monocytes and pericyte
`precursors, are affected directly by chemotherapy or are reduced in numbers by elimination of more primitive bone marrow progenitors which give rise to
`such cells is not yet clearly established. b, Cycling endothelial cells present in sprouting blood vessel capillaries; and c, authentic bone-marrow-derived
`circulating endothelial progenitor cells (EPC) that can incorporate into the lumen of growing vessels and differentiate into endothelial cells. Inhibiting the
`levels or function of VEGF can augment these various antiangiogenic mechanisms of chemotherapy. For example, VEGF is a potent mobilizer of EPC, a pro-
`survival (anti-apoptotic) factor for differentiated, activated endothelial cells, and also may be one of the more important paracrine growth factors secreted
`by proangiogenic vessel adherent bone-marrow-derived monocytes.
`
`The major signalling pathways in tumour angiogenesis
`VEGF/VEGF receptors
`Angiogenesis is a fundamental developmental and adult physiological
`process, requiring the coordinated action of a variety of growth factors
`and cell-adhesion molecules in endothelial and mural cells (reviewed in
`this issue by Coultas, Chawengsaksophak and Rossant, p. 937). So far,
`VEGF-A and its receptors are the best-characterized signalling pathway
`in developmental angiogenesis1,8,9. Loss of a single VEGF-A allele results
`in embryonic lethality1,8,9. This pathway also has an essential role in
`reproductive and bone angiogenesis8. Much research has also estab-
`lished the role of VEGF-A in tumour angiogenesis8,10. VEGF-A binds to
`two receptor tyrosine kinases (RTK), VEGFR-1 (Flt-1) and VEGFR-2
`(KDR, Flk-1) (reviewed in ref. 10). Of the two, it is now generally agreed
`that VEGFR-2 is the major mediator of the mitogenic, angiogenic and
`permeability-enhancing effects of VEGF-A. The significance of
`VEGFR-1 in the regulation of angiogenesis is more complex. Under
`some circumstances, VEGFR-1 may function as a ‘decoy’ receptor that
`sequesters VEGF and prevents its interaction with VEGFR-2 (ref. 10).
`However, there is growing evidence that VEGFR-1 has significant roles
`in haematopoiesis and in the recruitment of monocytes and other bone-
`marrow-derived cells that may home in on the tumour vasculature and
`promote angiogenesis11–13. In addition, VEGFR-1 is involved in the
`induction of matrix metalloproteinases (MMPs)14 and in the paracrine
`release of growth factors from endothelial cells15. Thus the VEGFR-1-
`selective ligands VEGF-B and placental-like growth factor (PlGF) may
`also have a role in these processes. Furthermore, in some cases VEGFR-
`1 is expressed by tumour cells and may mediate a chemotactic signal,
`thus potentially extending the role of this receptor in cancer growth16.
`VEGF-A gene expression is upregulated by hypoxia17. The tran-
`scription factor hypoxia inducible factor (HIF), which operates in con-
`cert with the product of the von Hippel–Lindau (VHL) tumour
`suppressor gene, has a major role in such regulation. Under normoxic
`conditions, the VHL protein targets HIF for ubiquitination and degra-
`dation17.
`In situ hybridization studies demonstrate that VEGF-A messenger
`
`RNA is expressed in many human tumours18. Renal cell carcinomas
`have a particularly high level of VEGF-A expression, consistent with
`the notion that inactivating VHL mutations occur in about 50% of
`such tumours19, thus providing a further explanation for the respon-
`siveness of this tumour type to a VEGF-A blockade20. However,
`VEGF-A upregulation in tumours is not only linked to hypoxia or
`VHL mutations. Indeed, a very broad and diverse spectrum of onco-
`genes is associated with VEGF-A upregulation, including mutant ras,
`erbB-2/Her2, activated EGFR and bcr-abl7,21. Besides VHL, inactiva-
`tion/mutation of various other suppressor genes can also result in
`VEGF upregulation. These genes include those associated with famil-
`ial syndromes characterized by well-vascularized hamartomas22.
`In 1993, it was reported that a murine anti-human VEGF-A mono-
`clonal antibody inhibited the growth of several tumour cell lines in
`nude mice, whereas the antibody had no effect on tumour-cell prolif-
`eration in vitro23. Subsequent studies have shown that many additional
`tumour cell lines, regardless of the tumour’s origin, are inhibited in
`vivo by the same anti-VEGF monoclonal antibody (reviewed in ref.
`24). Tumour-growth inhibition has also been demonstrated using
`independent anti-VEGF approaches including a dominant-negative
`VEGFR-2 mutant25, anti-VEGFR-2 antibodies26, small molecule
`inhibitors of VEGF RTKs27 and soluble VEGF receptors28,29. VEGF-A
`gene inactivation also suppresses angiogenesis in a transgenic model
`of multi-stage tumorigenesis30.
`
`Platelet-derived growth factor (PDGF) and angiopoietins
`Other signalling molecules that have an established role in the devel-
`opment and differentiation of the vessel wall such as PDGF-
`B/PDGFR-ȋ31 and the angiopoietins (Ang), the ligands of the Tie2
`receptor9, may also be therapeutic targets. PDGF-B is required for
`recruitment of pericytes and maturation of the microvasculature31.
`Inhibition of PDGFR-ȋ signalling has been reported to result in a
`tumour microvascular tree that is particularly dependent on VEGF-
`mediated survival signals. Withdrawal of VEGF-A leads to endothe-
`lial apoptosis and vascular regression32. In this context, newly formed
`
`968
`
`© 2005
`
`Nature Publishing Group
`
`© 2005 Nature Publishing Group
`
`

`

`NATURE|Vol 438|15 December 2005
`
`INSIGHT REVIEW
`
`vessels, whether they are tumour-associated or not, are particularly
`vulnerable to VEGF-A blockade, whereas mature vessels, covered by
`extracellular matrix and pericytes, may be resistant to VEGF inhibitors
`and other antiangiogenic agents. Furthermore, recent studies have
`emphasized the significance of tumour-derived PDGF-A (and poten-
`tially PDGF-C) and PDGFR-Ȋ signalling in the recruitment of an
`angiogenic stroma that produces VEGF-A and other angiogenic fac-
`tors33 (Fig. 1). Therefore, combining PDGF and VEGF inhibitors is an
`attractive anti-vascular and anti-tumour strategy.
`Ang-1 is required for further remodelling and maturation of the ini-
`tially immature vasculature. Unlike mouse embryos lacking VEGF-A
`or VEGFR-2, embryos lacking Ang-1 or its receptor Tie2 develop a
`rather normal primary vasculature, but this vasculature fails to undergo
`effective remodelling (reviewed in ref. 9). The generally accepted view
`is that Ang-1 is the major agonist for Tie2, whereas Ang-2 may act as an
`antagonist or a partial agonist34. However, more recent evidence indi-
`cates that, unexpectedly, Ang-2 has a positive role, at least in tumour
`angiogenesis35. Administration of Ang-2 inhibitors to tumour-bearing
`mice has been reported to result in delayed tumour growth, accompa-
`nied by reduced endothelial cell proliferation, consistent with an
`antiangiogenic mechanism. Therefore, inhibitors of Ang-2 may be can-
`didates for clinical development35.
`
`Axon-guidance molecules
`Recently, the role of axon-guidance receptors and ligands in develop-
`mental angiogenesis has received much attention. There are four main
`families: the neuropilins (NRP)/semaphorins, the ephrins, Robo/Slit
`and netrin/Unc5. For recent reviews, see refs 36, 37. Although the sig-
`nificance of these pathways in tumour angiogenesis is far from clear,
`there is emerging evidence that they have a role in some cancer models
`and therefore may be potential therapeutic targets.
`NRP1 and NRP2, previously shown to bind the collapsin/sema-
`phorin family and implicated in axon guidance, are also receptors for
`the heparin-binding isoforms of VEGF-A and seem to potentiate the
`activation of VEGFR-2 by VEGF165 (ref. 38). Therefore, NRPs may
`participate in tumour angiogenesis as positive modulators of VEGF
`signalling in endothelial cells. Furthermore, NRP1 and NRP2 are
`expressed on the cell surface of several tumour cell lines that bind
`VEGF165 and display a chemotactic response to this ligand, suggest-
`ing a pro-tumour activity of NRPs, with or without the involvement of
`VEGF RTK signalling37.
`The ephrins and their tyrosine kinase Eph receptors are a large fam-
`
`ily, initially implicated in neuronal guidance during development and
`subsequently found to have activities in other cell types, including vas-
`cular cells (for a review see ref. 39). The earliest evidence for a role of
`this family in angiogenesis was the report by Pandey et al. that ephrin
`A1 mediates TNF-Ȋ-induced angiogenesis in vivo40. Ephrin B2 and its
`receptor EphB4 are important for distinguishing between developing
`arterial and venous vessels (see p. 937). Recent studies suggest a role for
`Eph/ephrin interactions in malignant tumour progression and angio-
`genesis. Soluble EphB4-expressing human melanoma A375 cells grown
`subcutaneously in nude mice showed reduced tumour growth com-
`pared with control tumours41. Interfering with EphA signalling has
`been also reported to result in some inhibition of angiogenesis in
`tumour models42.
`Slits are secreted proteins that function as chemorepellents in axon
`guidance and neuronal migration through the Roundabout (Robo)
`receptor (reviewed in refs 36, 37). Wang et al. reported the expression
`of Slit2 in several tumour cell types and that Robo1 expression was
`localized to vascular endothelial cells43. Recombinant Slit2 protein
`attracted endothelial cells and promoted tube formation. Neutraliza-
`tion of Robo1 reduced microvessel density and growth of A375 cells
`transplanted in nude mice43.
`
`Negative regulators of angiogenesis
`Angiogenesis is a tightly regulated process and seems to be under the
`control of both positive and negative regulatory factors. Although sev-
`eral potential negative regulators of angiogenesis have been identified,
`relatively little is known about their role in the physiological regulation
`of angiogenesis. Thrombospondin, a large multifunctional glycopro-
`tein secreted by most epithelial cells in the extracellular matrix, inhibits
`angiogenesis associated with tumour growth and metastasis44. Several
`fragments of larger proteins have been described as endogenous
`inhibitors of angiogenesis including endostatin45, tumstatin46 and vaso-
`statin47. The most recently described endogenous inhibitor of angio-
`genesis is vasohibin, which seems to be derived from the endothelium
`and to operate as a feedback regulator48. The precise mechanism of
`action of these proteins remains to be more clearly defined, although
`several hypotheses have been proposed, including that they bind to spe-
`cific integrins in the case of endostatin and tumstatin49.
`
`Role of bone-marrow-derived cells in angiogenesis
`An intensively debated issue in the field is the contribution (as well as
`the precise nature) of bone-marrow-derived endothelial progenitor
`
`Figure 3 | Various strategies to inhibit VEGF signalling.
`These include monoclonal antibodies targeting
`VEGF-A (a) or the VEGF receptors (b, c). d, Chimaeric
`soluble receptors such as the ‘VEGF-trap’ (domain 2 of
`VEGFR-1 and domain 3 of VEGFR-2 fused to a Fc
`fragment of an antibody) are also undergoing clinical
`development. e, Additional extracellular inhibitors are
`aptamers that bind the heparin-binding domain of
`VEGF165 (pegaptanib). A variety of small-molecule
`VEGF RTK inhibitors that inhibit ligand-dependent
`receptor autophosphorylation of VEGFR-1 and
`VEGFR-2 are being tested. Additional strategies to
`inhibit VEGF signalling include antisense and siRNA
`targeting VEGF-A or its receptors.
`
`a
`
`Anti-VEGF
`antibodies
`
`Anti-VEGF-1
`antibodies
`
`b
`
`VEGF
`
`P
`
`P
`
`P
`
`P
`
`VEGFR-1
`
`VEGFR-2
`
`P
`
`P
`
`P
`
`P
`
`Endothelial cell
`
`e
`
`Small-molecule
`VEGFR TK
`inhibitors
`
`d
`
`Soluble
`VEGF
`receptors
`
`e
`
`Aptamers
`
`c
`Anti-VEGFR-2
`antibodies
`
`© 2005
`
`Nature Publishing Group
`
`© 2005 Nature Publishing Group
`
`969
`
`

`

`INSIGHT REVIEW
`
`NATURE|Vol 438|15 December 2005
`
`cells (EPC) to angiogenesis. However, there is little doubt that bone-
`marrow-derived cells participate in angiogenic processes, at least as a
`source of angiogenic factors. In 1997, Asahara et al. reported the iso-
`lation of putative EPC from human peripheral blood, on the basis of
`cell-surface expression of CD34 and other endothelial markers50.
`These cells were reported to differentiate in vitro into endothelial cells
`and seemed to be incorporated at sites of active angiogenesis in vari-
`ous animal models of ischaemia. These findings suggested that incor-
`poration into the lumen of bone-marrow-derived endothelial
`precursor cells contributes to the growing vessels, complementing res-
`ident endothelial cells in sprouting new vessels. Also, ischaemia and
`various cytokines, including VEGF and granulocyte-macrophage
`colony-stimulating factor (GM-CSF), were reported to mobilize EPC
`into sites of neovascularization51. However, the precise contribution of
`these cells in various pathophysiological circumstances was not clearly
`defined.
`Subsequent studies have suggested that the contribution of such
`cells to angiogenesis is dependent on the experimental system
`employed. In the angiogenic-defective, tumour-resistant Id-mutant
`mice, EPC accounted for a large proportion of endothelial cells asso-
`ciated with xenografted tumours52. Rafii and collaborators proposed
`that mobilization of EPC from bone marrow requires angiogenic-fac-
`tor-mediated activation of MMP-9, which leads to the release of the
`soluble KIT ligand. This ligand would in turn promote proliferation
`and motility of EPC within the bone-marrow microenvironment, thus
`creating permissive conditions for their mobilization into the periph-
`eral circulation53. However, in spontaneous tumours occurring in Id-
`deficient mice in the tumour-prone PTEN+/ǁ genetic background, the
`contribution of EPC was less significant54. Also, De Palma et al. sug-
`gested that the percentage of EPC that are truly incorporated into a
`growing vessel wall is very low and that the majority of bone-marrow-
`derived cells homing in on the tumour vasculature are adherent
`perivascular mononuclear cells, which may contain angiogenic fac-
`tors55. Peters et al. recently analysed the tumour endothelial cells in six
`individuals who developed cancers after bone-marrow transplantation
`with donor cells derived from individuals of the opposite sex and
`found that an average of 4.9% of cells of the total endothelial cell pop-
`ulation were derived from the bone marrow56.
`In summary, bone-marrow-derived cells seem to contribute to
`tumour angiogenesis, of which a small and variable proportion are
`probably true EPCs. Bone-marrow-derived circulating pro-angiogenic
`cells, regardless of their precise nature, may be a common target for
`antiangiogenic therapies and may be exploitable as surrogate bio-
`markers for the angiogenic process as well as antiangiogenic thera-
`pies57.
`
`Combination therapies
`It is increasingly likely that cancer therapy, with a few exceptions, will
`need to be combinatorial. It seems logical to target multiple pathways
`simultaneously. Much preclinical evidence indicates that combining
`antiangiogenic agents with conventional cytotoxic agents or radiation
`therapy results in additive or even synergistic anti-tumour effects58. So
`far, it is unclear whether such positive interaction takes place prefer-
`entially with specific types of antiangiogenic or cytotoxic agents. An
`issue that is being debated is the mechanism of such potentiation, as it
`would seem counterintuitive that ‘tumour-starving’ antiangiogenic
`drugs that suppress blood flow in tumours actually increase the effi-
`cacy of chemotherapy. Browder et al.59 and Klement et al.60 proposed
`that chemotherapy, especially when delivered at close regular intervals
`using relatively low doses with no prolonged drug-free break periods
`(‘metronomic therapy’), preferentially damages endothelial cells in
`tumour blood vessels. These cells are presumably dividing, and the
`simultaneous blockade of VEGF-A is thought to blunt a key survival
`signal for endothelial cells, thus selectively amplifying the endothelial
`cell targeting effects of chemotherapy, leading to improved subsequent
`killing of cancer cells.
`A similar process, in principle, may take place when combining
`
`a
`
`b
`
`Figure 4 | Computed tomography chest scans. These scans were taken of a
`NSCLC patient before (a) and after (b) three cycles (nine weeks) of
`treatment with bevacizumab plus carboplatin and taxol (reproduced from
`ref. 71 with permission from American Society of Clinical Oncology). Note
`that the tumour mass in a (arrow) underwent extensive necrosis and
`cavitation in b (arrow). This pattern was seen more frequently in patients
`treated with bevacizumab plus chemotherapy relative to chemotherapy
`alone. Cavitation may be associated with serious bleeding, especially when it
`occurs in proximity to large vessels71.
`
`more conventional maximum-tolerated dose chemotherapy regimens
`with a drug such as bevacizumab61. In addition, bone-marrow-derived
`pro-angiogenic circulating cells, probably including authentic EPC,
`seem to be very sensitive to both conventional cytotoxic and low-dose
`metronomic chemotherapy62. However, levels of such cells can rapidly
`rebound, returning to normal or even increased levels during the
`drug-free break periods after maximum-tolerated dose cytotoxic
`chemotherapy62. Because VEGF-A acts as a mobilizing and probably a
`survival agent for such cells, co-administration of a VEGF-targeting
`agent, especially one with a long half-life in the circulation (for exam-
`ple, anti-VEGF antibodies), would be expected to amplify and sustain
`the suppressive effects of standard (as well as metronomic) chemother-
`apy on bone-marrow-derived circulating pro-angiogenic cells63. Fur-
`thermore, it has been proposed that progressively accelerated
`proliferation and repopulation of cancer cells during intervals of radio-
`therapy or chemotherapy is an important cause of treatment failure64.
`It is tempting to speculate that antiangiogenic treatment during these
`intervals inhibits such repopulation process. Figure 2 illustrates the
`various cellular targets of chemotherapy.
`An alternative hypothesis has been proposed by Jain65. Submaximal
`doses of an antiangiogenic agent such as an anti-VEGFR-2 antibody
`would ‘normalize’ the vasculature that is characteristic of many vessels
`in tumours. This would result in pruning of excessive endothelial and
`perivascular cells, in a decrease in the normally high interstitial pres-
`sures detected in solid tumours and in temporarily improved oxy-
`genation and delivery of chemotherapy to tumour cells65. However,
`according to recent studies, the tumour vasculature can be ‘normal-
`ized’ transiently, and eliciting synergistic effects through this mecha-
`nism requires administration of chemotherapy or radiation therapy
`over a defined time window after the angiogenesis inhibitor66. Con-
`sidering also that in most clinical protocols no such intentional
`sequential administration is performed, it remains to be established
`whether such a mechanism accounts for the long-term beneficial
`effects of combination treatments observed in some trials. By contrast,
`acute administration of angiogenic inhibitors induces vascular
`changes consistent with ‘normalization’ in humans. In this regard, Wil-
`lett et al. reported that a single infusion of bevacizumab to patients
`with rectal carcinoma rapidly decreased tumour perfusion, vascular
`volume, microvascular density and interstitial fluid pressure as well as
`the number of viable, circulating endothelial cells in six colorectal can-
`cer patients67.
`Combinatorial therapies with antiangiogenic agents are not limited
`to those including cytotoxic chemotherapy. Several preclinical and
`clinical trials are exploring the combination of various angiogenesis
`
`970
`
`© 2005
`
`Nature Publishing Group
`
`© 2005 Nature Publishing Group
`
`

`

`NATURE|Vol 438|15 December 2005
`
`INSIGHT REVIEW
`
`inhibitors with other targeted therapies, such as EGFR or Her2
`inhibitors (cetuximab, erlotinib and trastuzumab), PDGFR/ bcr-abl
`inhibitors (imatinib), proteasome inhibitors (bortezomib) and other
`antiangiogenic agents such as inhibitors of integrins (for example
`Ȋvȋ3 and Ȋ5ȋ1).
`
`Clinical trials for antiangiogenesis
`Many angiogenesis inhibitors are currently in clinical trials. It is note-
`worthy that, in parallel to angiogenesis inhibitors, another class of vas-
`cular-targeting or vascular-modulating drugs is being tested, namely
`‘vascular-disrupting agents’. These drugs primarily target existing,
`recently formed vasculature and cause acute vascular occlusion and
`disruption of tumour blood flow68. For an overview of these trials, see
`http://www.cancer.gov/clinicaltrials/developments/anti-angio-table.
`The inhibitors tested include a variety of agents with diverse mecha-
`nisms of action (several of which are not known). At present,
`inhibitors of the VEGF pathway are the most clinically advanced, and
`bevacizumab, a humanized variant of a murine anti-VEGF-A mono-
`clonal antibody that was used in early proof-of-concept studies23, is the
`only FDA-approved antiangiogenic treatment for cancer therapy69.
`Figure 3 illustrates several methods for inhibiting the VEGF pathway.
`Several important clinical studies testing angiogenesis inhibitors
`have been presented at recent oncology meetings, such as the Ameri-
`can Society of Clinical Oncology meeting. Typically, clinical studies
`are presented and discussed at such meetings in advance of peer-
`reviewed publication. Therefore, in the interest of an up-to-date
`overview of the field, a discussion of some of these studies will be
`included here, with the caveat that the data are preliminary and require
`further analysis.
`The clinical trial that resulted in FDA approval of bevacizumab was
`a large, randomized, double-blind, phase III study in which beva-
`cizumab was administered in combination with bolus IFL (irinotecan,
`5FU and leucovorin) chemotherapy as first-line therapy for metasta-
`tic colorectal cancer5. Median survival was increased from 15.6
`months in the bolus-IFL plus placebo arm of the trial to 20.3 months
`in the bolus IFL plus bevacizumab arm. Similar increases were seen in
`progression-free survival, response rate and duration of response. The
`clinical benefit of bevacizumab was seen in all subject subgroups,
`including those defined by performance status, location of primary
`tumour, number of organs involved and duration of metastatic dis-
`ease5. Although bevacizumab was generally well tolerated, some seri-
`ous and unusual toxicities have been noted, albeit at low frequencies.
`Bevacizumab was associated with gastrointestinal perforations and
`wound healing complications in about 2% of patients. In addition, the
`incidence of arterial thromboembolic complications were increased
`about twofold relative to chemotherapy alone, with patients 65 years
`or older with a history of arterial thromboembolic events being at
`higher risk. Although the precise mechanism of this effect is unknown,
`it is conceivable that vascular damage induced by cytotoxic agents can
`be exacerbated by the blockade of VEGF-A.
`Preliminary data of a phase III study indicate that bevacizumab
`confers a survival advantage on patients with previously treated,
`relapsed, metastatic colorectal cancer in combination with FOLFOX4
`chemotherapy (5-fluorouracil, leucovorin and oxaliplatin), relative to
`chemotherapy alone (B. Giantonio, P. J. Catalono, N. J. Meropol, E. P.
`Mitchell, M. A. Schwartz et al., unpublished data).
`The role of bevacizumab in other tumour types and settings is
`currently under investigation, and phase III clinical trials of this drug
`in non-small-cell-lung cancer (NSCLC), renal cell cancer and metasta-
`tic breast cancer are ongoing. An early phase III trial of advanced,
`heavily pretreated, metastatic breast cancer showed that adding beva-
`cizumab to capecitabine chemotherapy did not improve progression-
`free survival, despite a doubling of the response rate (that is, tumour
`shrinkage of 50% or more) in the bevacizumab-treated arm of the
`trial70. Thus, the responses seemed to be very short in duration. How-
`ever, an interim analysis of a phase III study of women with previously
`untreated metastatic breast cancer treated with bevacizumab in com-
`
`bination with weekly paclitaxel chemotherapy showed that the study
`met its primary efficacy endpoint of improving progression-free sur-
`vival, compared with paclitaxel alone (K. Miller, unpublished data).
`Furthermore, administration of bevacizumab in combination with
`paclitaxel and carboplatin to patients with NSCLC resulted in
`increased response rate and time to progression relative to chemother-
`apy alone in a randomized phase II trial71. The most significant adverse
`event was serious haemoptysis. This was primarily associated with
`centrally located tumours with squamous histology, cavitation and
`central necrosis and proximity of disease to large vessels71. Figure 4
`illustrates the extensive tumour necrosis and cavitation that may result
`from the combination treatment71. More recently, preliminary results
`from a large, randomized phase III clinical trial for patients with pre-
`viously untreated advanced non-squamous NSCLC show that patients
`who received bevacizumab in combination with paclitaxel and carbo-
`platin lived longer than patients who received chemotherapy alone (A.
`B. Sandler, R. Gray, J. Brahmer, A. Dowlati, J. H. Schiller et al., unpub-
`lished data). Serious bleeding was infrequent but occurred more com-
`monly in the bevacizumab arm of the trial.
`Besides bevacizumab, several other VEGF inhibitors are being clin-
`ically pursued. A variety of small-molecule RTK inhibitors targeting
`the VEGF receptors have been developed. The most advanced are
`SU11248 and Bay 43-9006. SU11248 inhibits VEGFRs, PDGFR, c-kit
`and Flt-3 (ref. 72) and has been reported to have considerable efficacy
`in imatinib-resistant gastrointestinal stromal tumour (R. G. Maki, J. A.
`Fletcher, M. C. Heinrich, J. A. Morgan, S. George et al., unpublished
`data). Bay 43-9006 was initially identified as a raf kinase inhibitor and
`subsequently shown to inhibit several RTKs including VEGFRs. An
`interim analysis of phase III data indicates that Bay 43-9006
`monotherapy results in a significant increase in progression-free sur-
`vival in patients with advanced renal cell carcinoma (B. Escudier, C.
`Szczylik, T. Eisen, W. M. Stadler, B. Schwartz et al., unpublished data).
`Follow-up of such phase III data is ongoing to determine whether an
`overall survival benefit occurs. An earlier randomize

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