throbber
Rational Approaches to Design of Therapeutics
`Targeting Molecular Markers
`
`Richard J. Klasa, Alan F. List, and Bruce D. Cheson
`
`This paper introduces novel therapeutic strategies
`focusing on a molecular marker relevant to a
`particular hematologic malignancy. Four different
`approaches targeting specific molecules in unique
`pathways will be presented. The common theme
`will be rational target selection in a strategy that
`has reached the early phase of human clinical trial
`in one malignancy, but with a much broader poten-
`tial applicability to the technology.
`In Section I Dr. Richard Klasa presents preclini-
`cal data on the use of antisense oligonucleotides
`directed at the bcl-2 gene message to specifically
`downregulate Bcl-2 protein expression in non-
`
`Hodgkin’s lymphomas and render the cells more
`susceptible to the induction of apoptosis.
`In Section II Dr. Alan List reviews the targeting
`of vascular endothelial growth factor (VEGF) and its
`receptor in anti-angiogenesis strategies for acute
`myeloid leukemia (AML) and myelodysplastic
`syndromes (MDS).
`In Section III Dr. Bruce Cheson describes
`recent progress in inhibiting cell cycle progression
`by selectively disrupting cyclin D1 with structurally
`unique compounds such as flavopiridol in mantle
`cell lymphoma as well as describing a new class of
`agents that affect proteasome degradation pathways.
`
`I. ANTISENSE OLIGONUCLEOTIDES DIRECTED AT THE
`BCL-2 GENE MESSAGE IN NON-HODGKIN’S LYMPHOMA
`
`Richard J. Klasa, MD*
`
`Hematologic malignancies in general and non-Hodgkin’s
`lymphomas (NHLs) in particular are frequently associ-
`ated with gain of function mutations, many character-
`ized by balanced chromosomal translocations. Genome-
`wide surveys of gene expression are identifying both
`known and new transcripts that are overexpressed in dif-
`ferent histological subtypes of lymphoma.1 As we de-
`velop molecular classifications of these diseases it is
`assumed that a few key genes will account for the par-
`ticular survival advantage conferred on malignant lym-
`phocytes as compared to their normal counterparts. These
`genes and their protein products would provide rational
`targets for the development of therapeutic strategies to
`reverse this upregulation associated with the malignant
`phenotype.
`
`* Division of Medical Oncology, British Columbia Cancer
`Agency, 600 West 10th Avenue, Vancouver BC V52 4E6
`Canada
`
`Dr. Klasa is on the international advisory boards of Schering
`AG and Hoffmann-LaRoche, and is on the speakers’ bureau
`and advisory board for Berlex Canada.
`
`Non-Hodgkin’s Lymphoma and Bcl-2
`Over the past quarter century cytogenetic analysis has
`identified a number of reciprocal translocations that fre-
`quently occur in histologically identifiable subtypes of
`NHL.2 The transposition of the bcl-2 gene to the immu-
`noglobulin heavy chain promoter region in the t(14;18)
`translocation is associated with > 90% of follicular lym-
`phomas (FL) at diagnosis and 10% of diffuse large B-
`cell lymphomas (DLBCL), making it the most frequent
`event identified in NHL. Additionally, 50% of DLBCL
`overexpress the BCL-2 protein through other mecha-
`nisms, such as gene duplication, and are associated with
`a poorer prognosis after anthracycline-based combina-
`tion chemotherapy.3 BCL-2 is also overexpressed in
`mantle cell lymphoma (MCL), chronic lymphocytic leu-
`kemia (CLL), multiple myeloma (MM) and acute myel-
`ogenous leukemia (AML).4 This same widespread pat-
`tern of distribution is also seen in a variety of solid tu-
`mors including melanoma, small cell lung carcinoma,
`and colon carcinoma as well as prostate and breast car-
`cinoma, especially once the last two are hormone inde-
`pendent. The obvious conclusion is that BCL-2
`overexpression, by whatever means, confers a funda-
`mental advantage to malignant cells and that disruption
`of this overexpression might have therapeutic potential.
`Bcl-2 is an anti-apoptotic member of a large family
`of genes involved in the regulation of programmed cell
`death.5,6 Pro-apoptotic (BAX and BCL-Xs) and anti-
`
`Hematology 2001
`
`443
`
`DR. REDDY’S LABS., INC. EX. 1004 PAGE 1
`
`

`

`apoptotic (BCL-2, BCL-XL) molecules reside within the
`inner mitochondrial membrane and can homo- and
`heterodimerize upon appropriate stimulus. These inter-
`actions control the release of substances such as cyto-
`chrome C from the mitochondria into the cytosol through
`the opening or closing of specific pores in the membrane,
`with permeability determined by the relative abundance
`of the different molecules. Cytochrome C is central to
`the activation of caspases that initiate the apoptotic pro-
`cess. Thus, an overabundance of BCL-2 can prevent or
`retard activation of the apoptotic machinery and allow
`survival under conditions that might otherwise be lethal
`to a cell (Table 1).
`
`Antisense Oligonucleotide
`Reverse complementary or “antisense” oligonucleotides
`(ASOs) are short sequences of single stranded deoxyri-
`bonucleotides complementary to the coding regions of
`a gene that are designed to hybridize by Watson-Crick
`base pairing to messenger-RNA (m-RNA) sequences and
`thus facilitate their degradation.7,8 Naturally occurring
`antisense sequences have been identified as regulators
`of gene expression in a number of systems, supporting
`their potential for therapeutic development.9,10 The for-
`mation of a heteroduplex of m-RNA with the DNA of
`the ASO engages RNaseH, an enzyme that proceeds to
`specifically cleave off the m-RNA moiety, destroying
`the message and putatively leaving the therapeutic ASO
`molecule able to hybridize to another message se-
`quence.11 This results in a reduction in the target m-RNA
`pool, which subsequently leads to reduction in the spe-
`cific protein encoded (Figure 1; see color page 551).
`The presence of the ASO may also prevent the m-RNA
`from appropriately docking with the ribosomal machin-
`ery that would allow translation into a functional pro-
`tein. The end result is loss of expression of that protein
`in the cell.
`ASOs of 16-24 bases in length provide target speci-
`ficity while shorter or longer sequences can result in ran-
`dom hybridization within the transcript repertoire. Se-
`lecting the target areas within a messenger RNA must
`ultimately take into account its tertiary structure, which
`will determine the accessibility of an area for hybridiza-
`
`Table 1. Properties of BCL-2.
`
`• Oncogenic protein
`• Anti-apoptotic
`• Mitochondrial, endoplasmic reticulum, nuclear membrane
`localizations
`• Homotypic and heterotypic dimerization within family
`• Membrane channel/pore function
`• Cytochrome C release from mitochondria via BCL-2 family
`channels regulates cell fate under stress
`
`tion. These target areas are defined in oligonucleotide
`arrays where the entire antisense sequence to an m-RNA
`is displayed in overlapping segments on a slide. The in-
`tensity of hybridization of the labeled message deter-
`mines the candidate therapeutic ASOs.12 Screening of
`oligonucleotide libraries has also identified RNA sites
`that are most accessible to hybridization and correlated
`these sites with protein downregulation and biological
`function.13,14 More empirically, the first 6 codons of the
`open reading frame downstream of the AUG start site
`have repeatedly been found to be accessible to hybrid-
`ization and have been chosen for initial development of
`ASOs against a number of genes.
`As organisms have developed a sophisticated sys-
`tem for dealing with rogue strands of DNA both inside
`and outside the cell, the development of therapeutic
`molecules required chemical modifications to confer
`nuclease resistance and a favorable pharmacokinetic
`profile.15,16 Substitutions in the phosphodiester linkage
`of the bases in the ASO backbone has yielded a number
`of molecules now in clinical development with phospho-
`rothioates being the most widely studied first genera-
`tion molecules (Figure 2). The sulfur substitution yields
`an ASO that is nuclease resistant and capable of enter-
`ing the cell. It demonstrates good hybridization kinetics
`and has little in the way of non-sequence-dependent ef-
`fects or toxicities at concentrations required to down-
`regulate the target message. Additionally, although in
`tissue culture a delivery system such as cationic lipid is
`required for efficient intracellular penetration of these
`highly charged molecules, in vivo ASOs have been
`shown to be active in free form, possibly due to interac-
`tion with blood lipoproteins.17,18
`The correlation of a biologic effect with the spe-
`cific downregulation of target message and protein in
`vivo has been a major focus of the development of ASOs.
`However, ASOs can be very potent immune stimulators,
`by virtue of unmethylated CpG motifs presented in the
`context of certain flanking sequences, and therapeutic
`
`Figure 2. Phosphodiester substitutions in first generation
`antisense oligonucleotides.
`
`444
`
`American Society of Hematology
`
`DR. REDDY’S LABS., INC. EX. 1004 PAGE 2
`
`

`

`Antisense (G3139)
`Reverse Polarity (G3622)
`Mismatch (G4126)
`
`(AS)
`(RP)
`(MM)
`
`5′ tct ccc agc gtg agc cat
`5′ tac cga gtg cga ccc tct
`5′ tct ccc agc atg tgc cat
`
`this as background, we set out to test the in vivo combi-
`nation of ASOs targeting bcl-2 with a cytotoxic agent
`commonly used in the treatment of lymphoma.
`
`Figure 3. Oligodeoxynucleotides targeting Bcl-2.
`
`activity could in part be attributed to nonspecific sys-
`temic immune effects rather then to a specific ASO/
`mRNA interaction.19-22 Additionally, structures such as
`guanosine quartets can demonstrate sequence specific
`but non-antisense biological activity in vitro.23,24 Experi-
`mental designs therefore strive, by the use of appropri-
`ate control oligonucleotides (sense, missense, one and
`two base mismatch mis-sense) and various strains of
`immunodeficient animals, to isolate effects that can be
`attributed to the specific downregulation of target mes-
`sage and protein (Figure 3).
`
`ASOs to bcl-2 and Chemotherapy
`Escalating doses of G3139, cyclophosphamide and the
`combination of both agents were evaluated in severe
`combined immunodeficiency (SCID) mice bearing a sys-
`temic human DoHH2 lymphoma xenograft.35 Experi-
`ments confirmed that G3139 was able to downregulate
`BCL-2 expression in vitro and that treatment with G3139
`alone resulted in prolongation of median survival and
`cure of some animals. (Figure 5, left panel). This effect
`was dose and schedule dependent with no long-term sur-
`vivors seen when a dose of 5mg/kg was given daily for
`14 consecutive days as opposed to > 40% when the dose
`was increased to 12.5 mg/kg on the same daily schedule
`or either 5 or 12.5 mg/kg were administered for 14 treat-
`
`Figure 4. Targeting Bcl-2 may promote apoptosis following
`chemotherapy and irradiation.
`
`ASOs directed at bcl-2
`An 18-mer phosphorothiolated oligonucleotide, G3139,
`directed against the first six codons of the open reading
`frame of the bcl-2 gene message has been developed by
`Genta Pharmaceuticals (Figure 3). Studies of G3139 uti-
`lizing the BCL-2 overexpressing lymphoma cell lines
`DoHH-2 and SU-DHL-4 in vitro have shown down-
`regulation of message and protein expression.25 Tumor
`xenograft models in SCID mice have demonstrated thera-
`peutic activity that is specific when compared to control
`animals as well as animals treated with reverse polarity
`sense, 2-base mismatch mis-sense and non-sense oligo-
`nucleotides.25,26 Extensive pharmakokinetic as well as tox-
`icity studies have been performed identifying a dose range
`with a good therapeutic index.15,16 These findings supported
`the development of clinical trials using G3139 alone as
`treatment for BCL-2 overexpressing follicular lymphomas.
`Further therapeutic potential is sug-
`gested by in vitro experiments confirming
`that bcl-2 plays a major role in the response
`of malignant cells to various stresses which
`produce cellular damage, including chemo-
`therapy (Figure 4).4,27,28 Malignant cell lines
`transfected with a bcl-2 gene with result-
`ant overexpression of the protein product
`demonstrated increased resistance to vari-
`ous chemotherapeutic agents.29-32 Addition-
`ally, cell lines overexpressing BCL-2 were
`rendered more sensitive to killing by che-
`motherapeutic agents when either antisense
`oligonucleotides directed at the bcl-2 mes-
`sage were introduced into the culture or the
`cells were transfected with a vector bear-
`ing the antisense sequence.33,34 This has
`been correlated with a demonstration of
`downregulation of bcl-2 expression. With
`
`Figure 5. Survival of cohorts of 6 mice treated with oligonucleotides alone (left
`panel) or with cyclophosphamide (CY) (right panel).
`All six surviving animals treated with cyclophosphamide and G3139 sacrificed at 90
`days with no molecular evidence of disease detected.
`Saline=control animals ; AS=antisense oligonucleotide G3139 directed at bcl-2 ;
`SN=reverse sequence sense control ; MM= 2-base mismatch control.
`
`Hematology 2001
`
`445
`
`DR. REDDY’S LABS., INC. EX. 1004 PAGE 3
`
`

`

`ments on alternate days (28-day schedule). Similarly,
`cyclophosphamide treatment alone resulted in no long-
`term survivors at lower doses but was able to cure ani-
`mals at high doses. The addition of G3139 to low dose
`cyclophosphamide resulted in the cure of the majority
`of animals (Figure 5, right panel). The interaction be-
`tween the two agents does show dose-response correla-
`tions; for the two low doses of cyclophosphamide tested,
`increasing the dose of G3139 from 2.5 to 5 mg/kg re-
`sulted in longer median survivals and overall increase in
`long-term survivors. A rather striking result was achieved
`when a completely ineffective dose of cyclophospha-
`mide (15 mg/kg, median survival 36 days and no long-
`term survivors) was combined with a modestly effective
`dose of G3139 (2.5 mg/kg, 61 day median survival and
`16% long-term survivors) to produce a 72-day median
`survival and 50% long-term survivors. Mice sacrificed
`at 90 days showed no histological evidence of any dis-
`ease in all tissues analyzed, including immunoperoxidase
`staining for BCL-2, or molecular detection of bcl-2, by
`PCR. This suggests that chemotherapy at very modest
`doses could be made much more effective without in-
`creasing toxicity when combined with an antisense oli-
`gonucleotide. Such an increase in the efficacy of cur-
`rently available agents could significantly alter the prog-
`nosis of a large number of moderately chemotherapy
`sensitive human tumors, resulting in longer median sur-
`vivals and increasing the potential for cure.
`The model has direct relevance to the clinical situa-
`tion faced in NHL, where patients typically present with
`a chemotherapy-sensitive tumor at diagnosis that re-
`gresses only to recur within months to years post-treat-
`ment. The DoHH2 cell line was derived from a follicu-
`lar lymphoma carrying a t(14;18), which results in bcl-2
`gene overexpression. The aggressive nature of the dis-
`ease in this model is, however, more suggestive of a trans-
`formation to a higher-grade histology, a common event
`in follicular lymphoma. Indeed, a recent re-exploration
`of the molecular and cytogenetic features of the cell line,
`using more sensitive detection techniques, has revealed
`a second translocation involving the c-myc oncogene
`with a resultant derivative chromosome 8 carrying
`t(8;14;18).36 We have recently described this phenom-
`enon of double translocation of both bcl-2 and c-myc in
`a subset of patients with small non-cleaved cell (Burkitt-
`like) lymphoma, which represents a very aggressive form
`of the disease.37
`
`Clinical Studies
`G3139 has been studied as a single agent in a phase 1
`trial in heavily pretreated (median of 4 prior regimens)
`patients with relapsed NHL.38,39 Twenty-one patients with
`follicular (9), small lymphocytic (8), diffuse large B-
`cell (3) or mantle cell (1) lymphomas that expressed
`
`BCL-2 were treated at 8 dose levels ranging from 4.6 to
`195.8 mg/m2/day by continuous subcutaneous infusion
`for 14 consecutive days. No significant toxicity was seen
`up to doses of 110 mg/m2/day. One complete and 2 mi-
`nor responses as well as 9 disease stabilizations were
`seen in this heavily pretreated group. BCL-2 protein was
`decreased in 7 out of 16 samples examined, including 2
`from accessible tumor sites and 5 samples of peripheral
`blood or marrow mononuclear cells.
`One study combining a chemotherapeutic agent with
`G3139 has been reported in metastatic melanoma,40 and
`studies are ongoing in a number of other solid tumors
`(melanoma, prostate carcinoma) and hematological ma-
`lignancies (myeloma, chronic lymphocytic leukemia, and
`acute myeloid leukemia). A phase 1 study at our institu-
`tion in relapsed follicular lymphomas combining esca-
`lating doses of both cyclophosphamide and G3139 has
`not identified any unexpected toxicity. The last patient
`enrolled has received cyclophosphamide 750 mg/m2 with
`2.3 mg/kg/day of G3139 by continuous intravenous in-
`fusion for 14 consecutive days (Table 2).
`
`Conclusions
`The identification of overexpression or aberrant expres-
`sion of genes that result in a gain of function, through
`genome wide surveys of cells and tissues in varying
`states, will provide unprecedented insight into the biol-
`ogy of hematological malignancies. Specific down-
`regulation of such overexpression with antisense oligo-
`nucleotides allows disruption of single gene function at
`the messenger RNA and protein level and the study of
`downstream events in the involved molecular pathways
`both in vitro and in vivo. Genes that are critical to the
`differential growth and survival advantage enjoyed by
`malignant cells are being identified and are logical thera-
`peutic targets. The development of ASOs directed at the
`bcl-2 gene provides a model by which a systemic therapy
`for a metastatic disease has been taken from the labora-
`tory through preclinical studies to early phase clinical
`trials, building on knowledge of this particular gene’s
`role in cellular apoptosis. Combining multiple antisense
`
`Table 2. Phase I study of G3139 and cyclophosphamide in
`relapsed follicular lymphoma.
`
`G3139 mg/kg/day Cyclophosphamide
`mg/m2/IV
`CIVI
`(day 1 to 14)
`(day 8)
`0.6
`250 done
`1.2
`250 done
`2.3
`250 done
`2.3
`500 done
`2.3
`750 ongoing
`3.1
`1000
`5.0
`1000
`
`Level
`1
`2
`3
`4
`5
`6
`7
`
`446
`
`American Society of Hematology
`
`DR. REDDY’S LABS., INC. EX. 1004 PAGE 4
`
`

`

`strategies with other therapeutic modalities has the po-
`tential to increase the specificity of the treatments avail-
`able to our patients, thus improving their efficacy and
`reducing toxicity.
`
`II. TARGETING ANGIOGENESIS IN HEMATOLOGIC
`MALIGNANCIES
`
`Alan F. List, MD*
`
`The seminal observations that the growth and metastatic
`potential of solid tumors is dependent upon the forma-
`tion of new blood vessels triggered an enormous expan-
`sion in angiogenic research that has yielded novel thera-
`peutics targeting an array of angiogenic molecules. In-
`vestigations of the relevance of angiogenesis in hemato-
`logic malignancies are still at an early stage, but accu-
`mulating evidence indicates that the angiogenic profile
`of many hematologic malignancies is distinct from that
`of solid tumors. As progeny of a common endothelial
`and hematopoietic stem cell, hematologic malignancies
`may elaborate and respond to angiogenic factors in a
`paracrine or autocrine fashion, contributing to tumor cell
`survival and expansion, adhesion, bone resorption and
`immune suppression.
`
`Angiogenesis
`Blood vessel development is characterized by two dis-
`tinct biologic processes, vasculogenesis, and angiogen-
`esis. Vasculogenesis, which is largely restricted to em-
`bryonic development, involves de novo endothelial cell
`differentiation from mesodermal precursors as the pre-
`requisite for coordinated blood vessel generation.1 An-
`giogenesis, the process of new blood vessel formation
`from preexisting vessels, is responsible for the genera-
`tion of neovasculature in adult life, and occurs physi-
`ologically during wound healing and within female re-
`productive organs during the menstrual cycle, as well as
`in pathologic conditions such as proliferative retinopa-
`thy, arthritic synovium, and human malignancies.2 An-
`giogenesis is a multistep process that includes both acti-
`vation and resolution phases. The activation phase is
`responsible for the sequential events of basement mem-
`brane degradation, endothelial cell proliferation and
`migration, and capillary lumen formation. The resolu-
`tion phase is responsible for the maturation and stabili-
`zation of the newly formed microvasculature through
`the recruitment of pericytes, promotion of basement
`membrane reconstitution, and subsequent extinction of
`the endothelial cell mitogenic response. A large number
`of pro-angiogenic molecules and endogenous angiogen-
`
`* Arizona Cancer Center, 1515 N Campbell, Room 3945, PO
`Box 245004, Tucson AZ 85774
`
`esis inhibitors that coordinate the angiogenic response
`have been delineated (Table 3).3 Vascular endothelial
`growth factor (VEGF), first identified in 1989 and later
`isolated from the HL-60 myeloid leukemia cell line, is a
`critical regulator of vascular development that is respon-
`sible for activation of endothelial cell proliferation dur-
`ing vasculogenesis and the direction of capillary sprout-
`ing during angiogenesis.4 Indeed, gene inactivation stud-
`ies indicate that VEGF is essential to the neoplastic an-
`giogenic response.5
`
`VEGF and Receptor Tyrosine Kinases
`The VEGF-A molecule is a disulfide-linked homodimer
`represented by five different isoforms generated by al-
`ternate exon splicing of gene message.6 The correspond-
`ing VEGF monomers range in size from 121 to 206
`amino acids, with smaller molecules (i.e., 121, 145, and
`165 amino acids) representing the secreted and diffusable
`isoforms, whereas the larger proteins (189, 206 amino
`acids) are sequestered by heparin sulfate residues present
`on cell surfaces or within the extracellular matrix.7,8 Al-
`though all isoforms are biologically active, the VEGF165
`isoform predominates and is recognized as a more po-
`tent and bioavailable endothelial cell mitogen.9,10 Recent
`investigations indicate that the VEGF family is composed
`of five members in addition to the prototype, VEGF-A,
`including VEGF-B, VEGF-C, VEGF-D, VEGF-E and
`PIGF (placental growth factor).6 Within the arterial wall,
`VEGF is produced by smooth muscle cells in response
`to oxidative stress and other stimuli.11 Autocrine pro-
`duction of VEGF and corresponding receptor upregu-
`lation is also demonstrable in endothelial cells in response
`to hypoxia, nitric oxide, VEGF deprivation and other
`cellular stresses.12,13
`Trophic response to the VEGF family members is
`directed by selective interaction with structurally ho-
`mologous type III receptor tyrosine kinases (RTKs), in-
`cluding VEGFR-1, originally termed fms-like tyrosine
`kinase (FLT-1),14 VEGFR-2 or kinase insert domain-con-
`taining receptor (KDR)/fetal liver kinase-1 (flk-1),15 and
`the recently characterized VEGFR-3 or FLT-4 receptor
`(Figure 6).16 Each of these receptors contains seven ex-
`tracellular immunoglobulin homology domains that cre-
`ate the ligand-binding site, a single short transmembrane-
`spanning sequence, and a cytoplasmic tail that contains
`the tyrosine kinase domain akin to that of the c-kit and
`PDGF-receptors.17,18 The external ligand-binding com-
`ponent of VEGFR-3 differs from the other VEGF re-
`ceptors in that the fifth immunoglobulin domain is
`cleaved during receptor processing to yield covalent,
`disulfide-linked subunits.19 In adults, VEGFR-1 and
`VEGFR-2 expression is limited to the vascular endothe-
`lium, monocytes (VEGFR-1), and primitive hematopoi-
`etic precursors (VEGFR-2), whereas VEGFR-3 is re-
`
`Hematology 2001
`
`447
`
`DR. REDDY’S LABS., INC. EX. 1004 PAGE 5
`
`

`

`Table 3. Endogenous stimulators and inhibitors of angiogenesis.
`
`Native Inhibitors
`Interferon (IFN)-α,γ
`
`Prolactin (16 Kd fragment),
`IFN-α,γ
`
`Tissue inhibitor of
`metalloprotease
`(TIMP-1, TIMP-2)
`Plasminogen activator-
`inhibitor-1 (PAI-1)
`Zinc
`p53 Rb
`
`Angiogenic Molecules
`VEGF (A-D), PIGF
`Angiogenin
`Angiotropin
`Epidermal growth factor (EGF)
`Fibroblast growth factor
`(acidic and basic; FGF)
`Hepatocyte growth factor/scatter
`factor (HGF, SF)
`Platelet-derived growth factor (PDGF)
`Tumor necrosis factor-α (TNFα)
`Interleukin-8 (IL-8)
`Insulin-like growth factor-1 (IGF-1)
`Cathepsin
`Proteases and
`Protease Inhibitors Gelatinase-A, -B (MMP2,9)
`Stromelysin
`Urokinase-type plasminogen
`activator (uPA)
`Copper
`c-myc
`ras
`c-src
`v-raf
`c-jun
`Signal Transduction Thymidine phosphorylase
`Enzymes
`RAS - arnesyl transferase
`Geranylgeranyl transferase
`Interleukin-1
`Interleukin-6
`Interleukin-8


`3 integrin
`v
`Angionpoietin-1 (Ang-1)
`Antionstatin II (AT1 receptor)
`Endothelin (ETB receptor)
`Erythropoietin
`Hypoxia
`iNOS
`Platelet-activating factor (PAF)
`Prostaglandin E, COX-2
`Thrombopoietin
`
`regulate extracellular matrix (ECM)
`degradation and endothelial cell migra-
`tion via protease elaboration.31 VEGF-
`E is an alternate ligand for VEGFR-2
`and has biologic activities similar to
`VEGF-A.32 Neuropilin-1 (NRP-1), a
`cell surface glycoprotein, acts as a co-
`receptor for VEGF-A165 to enhance its
`binding to VEGFR-2 on endothelial
`cells. Unlike the VEGF receptors, NRP-
`1 lacks an intracellular domain and
`therefore is not a direct mediator of
`cytokine signaling.33
`VEGF-A exerts its biologic effects
`via interaction with either VEGFR-1
`or VEGFR-2. VEGF receptors dimer-
`ize in response to ligand engagement,
`activating the tyrosine kinase located on
`the intracellular cytoplasmic tail. The
`resultant auto-phosphorylation of these
`receptors on specific tyrosine residues
`creates docking sites for cytoplasmic
`signaling molecules.34 Activation of
`VEGF receptors in endothelial cells trig-
`gers recruitment of adaptor and signal-
`ing proteins that contain Src homology
`domain-2 (Sh-2) including Shc and
`Sck.35,36 Recruitment and phosphoryla-
`tion of these adaptor molecules permit
`their binding to the p85 subunit of phos-
`phatidyl inositol-3 (PI-3) kinase and to
`phospholipase-Cg, activating the PI-3
`kinase and ERK (stress activated pro-
`tein kinase-1, 2) kinase signaling cas-
`cades.37-39 PI-3 kinase phosphorylates
`Akt (protein kinase-B), a serine/threo-
`nine kinase involved in antiapoptotic
`signaling.40 Akt activation permits its
`translocation to the plasma membrane,
`where it phosphorylates and inactivates a variety of pro-
`apoptotic molecules, including BAD, caspase-9, mito-
`chondrial Raf, and the forkhead transcription factor-1
`(FKHR-1).41 Similarly, through PI-3 kinase activation,
`VEGF-A activates focal adhesion kinase in endothelial
`cells, a step that is critical to the recruitment and activa-
`tion of cell adhesion molecules containing β
`
`1, β2, and β
`3
`integrins.42,43 Indeed VEGFR-2 interaction with its ligand
`triggers receptor association with PI-3 kinase, vascular
`endothelial (VE)-cadherin and α

`3, components that
`V
`are essential to the creation of an active multimeric sig-
`naling complex. Thus, VEGFR-2 activation assures
`proper juxtaposition of the receptor with cytoskeletal
`proteins.44,45
`
`Growth factors
`
`Trace Elements
`Oncogenes
`
`Cytokines
`
`Endogenous
`
`Interleukin-10
`Interleukin-12
`
`Angiopoietin-2
`Angiotensin
`Angiotensin II (AT2 receptor)
`Caveolin-1, caveolin-2
`Endostatin
`Interferon-α
`Isoflavones
`Platelet factor-4
`
`Thrombospondin
`Troponin-1
`
`Abbreviations: VEGF, vascular endothelial growth factor; PIGF, placental growth factor;
`iNOS, inducible nitric oxide synthetase; MMP, matrix metalloproteases
`
`stricted to lymphatic endothelium.20-24 The critical role
`for these receptors and that of VEGF has been demon-
`strated in gene inactivation studies. VEGF-A and the
`VEGFR-2 receptor are essential to embryonic vascu-
`logenesis and definitive hematopoiesis.25-27 In vitro in-
`vestigations indicate that VEGFR-2 signaling is respon-
`sible for proliferation and differentiation of endothelial
`cells in response to VEGF-A stimulation, whereas
`VEGFR-1 has been implicated as a decoy receptor and
`potential activator of endothelial permeability.28 Ligand
`binding to VEGFR-3 is restricted to VEGF-C and VEGF-
`D, which together direct lymphangiogenesis.17,29,30 The
`actions of the remaining VEGF family members are less
`well-defined; however, receptor recognition for VEGF-
`B appears restricted to VEGFR-1 where it is believed to
`
`448
`
`American Society of Hematology
`
`DR. REDDY’S LABS., INC. EX. 1004 PAGE 6
`
`

`

`transcriptional activator of other genes in-
`cluding erythropoietin, inducible nitric ox-
`ide synthetase (iNOS), insulin growth fac-
`tor (IGF-2), glycolytic enzymes, and tumor
`growth factor-β (TGF-β).53
`
`Hemangioblast and Hematopoiesis
`During embryogenesis, pluripotential he-
`matopoietic stem cells originate from cell
`clusters in the ventral floor of the pre-um-
`bilical dorsal aorta.54 These common he-
`matopoietic/endothelial (HE) precursors
`share an antigenic phenotype characterized
`by expression of the progenitor cell antigen,
`CD34, and VEGFR-2.27,55 Murine gene
`knockout studies indicate that VEGFR-2 ex-
`pression is required for vasculogenesis and
`for establishment of definitive hematopoie-
`sis. Although HE progenitors can be gener-
`ated in vitro from VEGFR-2 deficient em-
`bryonal stem cells, VEGFR-2 signaling is
`essential for progenitor migration and endot-
`helial commitment. Post-natally, heman-
`gioblasts, or HE progenitors, persist and con-
`tribute to long-term hematopoiesis and to the
`generation of circulating endothelial cells.
`Confirmation of leukemia specific gene ex-
`pression (i.e., bcr/abl in chronic myeloid leu-
`kemia) and the identification of donor-spe-
`cific DNA alleles (i.e., in allograft recipi-
`ents) within vascular endothelium provides definitive
`proof that bone marrow-derived hemangioblast progeny
`contribute to the maintenance of endothelium in adults.56-
`58 These primitive hemangioblast progenitors represent
`less than 2% of circulating CD34+ cells and display a
`surface phenotype characterized by CD34+, AC133+,
`VEGFR-2+, platelet endothelial cell adhesion molecule
`(PECAM), and the stromal derived factor-1 (SDF-1) re-
`ceptor, CXCR-4.59 Under appropriate culture conditions,
`hematopoietic commitment of these precursors is demon-
`strable by acquisition of the leukocyte common antigen,
`CD45, and β1 integrins, whereas angioblasts display VE-
`cadherin.
`VEGF receptorexpression extends beyond the primi-
`tive progenitor compartment during adult hematopoie-
`sis. VEGFR-1 is expressed by monocytes and commit-
`ted CD34+ progenitors, and is responsible for activation
`of migration and maturation skewing, respectively.22,23,60
`With the exception of primitive hematopoietic progeni-
`tors, VEGFR-2 expression appears restricted to mega-
`karyocytes.20 Within the adult bone marrow, cellular
`expression of angiogenic molecules outside of stromal
`elements is restricted to erythroblastic islands. Both
`VEGF-A and PIGF are secreted by erythroblasts within
`
`Figure 6. Interaction of vascular endothelial growth factor (VEGF) family
`members with their cognate receptors on endothelial cells.
`The VEGF receptors harbor seven extracellular immunoglobulin (Ig)-homology
`domains. The fifth Ig domain of VEGFR-3 is cleaved after biosynthesis to yield a
`stable disulfide bond linking the Ig subunits. VEGFR-1 and VEGFR-2 initiate signals
`essential to the angiogenic response, whereas VEGFR-3 regulates lymph angiogen-
`esis. NRP-1 (neuropilin-1) binds to the carboxy-terminal sequence of VEGF165 to
`enhance binding of the angiogenic molecule to the VEFGR-2 receptor.
`Abbreviations: TK, tyrosine kinase; MMP, matrix metalloproteases; PIGF, placental
`growth factor.
`
`VEGF Regulation
`Transcriptional regulation of VEGF is influenced by a
`number of signals that converge upon the oxygen sensi-
`tive transcription factor, hypoxia-inducing-factor (HIF)-
`1a. HIF is a heterodimeric protein composed of α and β
`subunits.46 The β subunit is expressed constitutively,
`whereas the a subunit is a member of a family of DNA-
`binding proteins that contain trans-activation domains
`within the carboxy-terminus. Under normal oxygen con-
`ditions, HIF-1a is degraded rapidly by the ubiquitin-
`proteasome pathway.47 Hypoxia stabilizes the HIF-1a
`protein by inhibiting its proteasome degradation. The
`von Hippel-Lindau (VHL) tumor suppressor gene is one
`component of the ubiquitin ligase complex that
`ubiquinates and promotes the degradation of HIF-1a
`under normoxic conditions. Inactivation of VHL, as oc-
`curs in the von Hippel-Lindau syndrome, stabilizes HIF-
`1a and sustains tumor production of VEGF.48 Inactiva-
`tion of a negative regulator of HIF-1a such as VHL is
`only one of many signals which converge upon HIF-1a
`to activate VEGF expression. Alternate signals include
`sustained RAS activation, β-integrin activation, stress-
`induced ERK signaling, and inactivation of p53.49-52 In
`addition to its regulatory effects on VEGF, HIF-1a is a
`
`Hematology 2001
`
`449
`
`DR. REDDY’S LABS., INC. EX. 1004 PAGE 7
`
`

`

`erythroid islands,61 suggesting that

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