throbber
CANCER TREATMENT REVIEWS (2004) 30, 193–204
`
`www.elsevierhealth.com/journals/ctrv
`
`LABORATORY–CLINIC INTERFACE
`
`PI3K/Akt signalling pathway and cancerq
`
`Juan Angel Fresno Vara, Enrique Casado, Javier de Castro,
`Paloma Cejas, Cristobal Belda-Iniesta, Manuel Gonzalez-Baron*
`
`Catedra de Oncologıa y Medicina Paliativa, Servicio de Oncologıa Medica,
`Hospital Universitario La Paz, Paseo de la Castellana 261, Madrid 28046, Spain
`
`KEYWORDS
`PI3K;
`Akt;
`Signalling;
`Apoptosis;
`Proliferation;
`Chemotherapy;
`Resistance;
`New cancer drugs
`
`Phosphatidylinositol-3 kinases, PI3Ks, constitute a lipid kinase family
`Summary
`characterized by their ability to phosphorylate inositol ring 30-OH group in inositol
`phospholipids to generate the second messenger phosphatidylinositol-3,4,5-tris-
`phosphate (PI-3,4,5-P3). RPTK activation results in PI(3,4,5)P3 and PI(3,4)P2
`production by PI3K at the inner side of the plasma membrane. Akt interacts with
`these phospholipids, causing its translocation to the inner membrane, where it is
`phosphorylated and activated by PDK1 and PDK2. Activated Akt modulates the
`function of numerous substrates involved in the regulation of cell survival, cell cycle
`progression and cellular growth. In recent years, it has been shown that PI3K/Akt
`signalling pathway components are frequently altered in human cancers. Cancer
`treatment by chemotherapy and c-irradiation kills target cells primarily by the
`induction of apoptosis. However, the development of resistance to therapy is an
`important clinical problem. Failure to activate the apoptotic programme represents
`an important mode of drug resistance in tumor cells. Survival signals induced by
`several receptors are mediated mainly by PI3K/Akt, hence this pathway may
`decisively contribute to the resistant phenotype. Many of the signalling pathways
`involved in cellular transformation have been elucidated and efforts are underway
`to develop treatment strategies that target these specific signalling molecules or
`their downstream effectors. The PI3K/Akt pathway is involved in many of the
`mechanisms targeted by these new drugs, thus a better understanding of this
`crossroad can help to fully exploit the potential benefits of these new agents.
`
`c 2003 Elsevier Ltd. All rights reserved.
`
`Introduction
`
`Carcinogenesis process is the result of a disturbed
`balance between cell division and growth on one
`hand, and programmed cell death (i.e., apoptosis),
`on the other. In the context of this delicate bal-
`
`ance, proteins and signalling pathways regulating
`cell growth, differentiation and development un-
`dergo oncogenic changes far more often than do
`other molecule groups. In recent years, it has been
`shown that PI3K/Akt signalling pathways, involved
`in the above and other processes, are frequently
`disturbed in many human cancers. This pathway
`plays a major role not only in tumor development
`but also in the tumor’s potential response to cancer
`treatment. Many of the new “targeted agents” have
`been specifically designed to act on PI3K/Akt-re-
`lated targets. Therefore, a better understanding of
`
`q We apologize for not citing original work of many colleagues
`because of space constraints.
`* Corresponding author. Tel./Fax: +34-917-277-118.
`E-mail address: mgonzalezb.hulp@salud.madrid.org
`Gonzalez-Baron).
`
`(M.
`
`doi:10.1016/j.ctrv.2003.07.007
`
`0305-7372/$ - see front matter c 2003 Elsevier Ltd. All rights reserved.
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 001
`
`

`
`194
`
`J.A.F. Vara et al.
`
`PI3K/Akt signalling pathway may improve the on-
`cologist’s prognosis ability and accuracy of predic-
`tion as to response to treatment. In the present
`review, we discuss PI3K/Akt pathway components
`and functions as well as its alteration in cancer and
`relationship with the different therapeutic ap-
`proaches. Moreover, we describe the current status
`of the different pharmacological compounds that
`target this important pathway.
`
`PI3K/Akt pathway components and
`functions
`
`PI3K
`
`Phosphatidylinositol-3 kinases, PI3Ks, constitute a
`lipid kinase family characterized by their ability to
`phosphorylate inositol ring 30-OH group in inositol
`phospholipids.1 Class-I PI3Ks are heterodimers
`composed of a catalytic subunit (p110) and an
`adaptor/regulatory subunit (p85). This class is
`further divided into the subclass IA, which is acti-
`vated by receptors with protein tyrosine kinase
`activity (Receptor Protein Tyrosine Kinase, RPTK),
`and the subclass IB, which is activated by receptors
`
`coupled with G proteins. The substrate for class I
`PI3Ks is phosphatidylinositol-4,5-bisphosphate (PI-
`4,5-P2) to generate the second messenger phos-
`phatidylinositol-3,4,5-trisphosphate (PI-3,4,5-P3).
`Three isoforms of catalytic subunit p110, named a,
`b and c, and seven adapting protein generated by
`alternative splicing of three genes (p85a, p85b and
`p55c) are known for PI3K class IA.
`
`Activation mechanism
`
`RPTK activation results in the association of PI3K with
`the receptor through one or two SH2 domains in the
`adaptor unit to phosphotyrosine consensus motifs.
`This leads to allosteric activation of the catalytic
`subunit.2 PI3K activation leads to PI-3,4,5-P3 pro-
`duction in a few seconds (Fig. 1). Polyphosphoinosi-
`tide effects on cells are mediated through the
`specific binding to at least two distinct protein-lipid
`binding domains, namely, the FYVE and pleckstrin-
`homology (PH) domains (PH).3 Proteins containing
`the latter domain represent critical mediators for
`PI3K Class IA-induced signalling. PH domains are
`found in many proteins, including protein serine/
`treonine kinase 30-phosphoinositide-dependent klin-
`ase1 (PDK1) and Akt/PKB, both central for the
`transforming effects of deregulated PI3K activity.
`
`Figure 1 Activation of growth factor receptor protein tyrosine kinases results in autophosphorylation on tyrosine
`residues. PI3K is recruited to the membrane by directly binding to phosphotyrosine consensus residues of growth factor
`receptors or adaptors through one or both SH2 domains in the adaptor subunit. This leads to allosteric activation of the
`catalytic subunit. Activation results in production of the second messenger phosphatidylinositol-3,4,5-trisphosphate
`(PIP3). The lipid product of PI3K, PIP3, recruits a subset of signalling proteins with pleckstrin homology (PH) domains to
`the membrane, including PDK1 and Akt. PTEN, is a PI-3,4,5-P3 phosphatase, which negatively regulates the PI3K/Akt
`pathway. Once activated, Akt mediates the activation and inhibition of several targets, resulting in cellular survival,
`growth and proliferation through various mechanisms.
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 002
`
`

`
`Akt signalling and cancer
`
`PKB/Akt
`
`PKB/Akt was characterized following the isolation
`of two genes termed akt1 and akt2, identified as
`the human homologues for the viral oncogene
`v-akt, which is known to be responsible for a type
`of leukemia in mice.4 Later, two unrelated studies
`revealed that v-akt and its human homologues en-
`coded a protein kinase bearing some resemblance
`to kinase protein C (PKC) and protein kinase A
`(PKA);5–7 therefore, it was called PKB. To date,
`three members of this PKB family have been iso-
`lated, named PKBa (Akt1), PKBb (Akt2), and PKBc
`(Akt3).8 Although they are product of different
`genes, they are closely related to each other, with
`up to 80% of amino acid homology. The three genes
`are expressed differentially, with a broader ex-
`pression for PKBa/Akt1 and PKBb/Akt2 and a more
`restricted expression for PKBc/Akt3. Each isoform
`presents a plekstrin homology (PH) domain of ap-
`proximately 100 amino acids in the N-terminal re-
`gion. There is then the kinase domain, which is very
`similar to those of PKA and PKC.9;10 In this region, a
`treonine residue (T308 in Akt1) is found, whose
`phosphorylation is required for Akt activation.
`Following the kinase domain is a hydrophobic
`C-terminal tail containing a second regulatory
`phosphorylation site (S473 in Akt1). T308 and S473
`phosphorylation occurs in response to growth fac-
`tors and other extracellular stimuli, and is essential
`to maximal Akt activation.11
`
`Activation mechanism
`
`RPTK activation results in PI(3,4,5)P3 and PI(3,4)P2
`production by PI3K at the inner side of the plasma
`membrane. Akt interacts with these phospholipids,
`causing its translocation to the inner membrane,
`where PDK1 is located (Fig. 1). The interaction of
`Akt PH domain with 30-hosphoinositides is thought
`to provoke conformational changes in Akt, result-
`ing in exposure of its two main phosphorylation
`sites. Likewise, PH domains may mediate the Akt
`and PDK1 approach through their heterodimeriza-
`tion. PDK1, which is thought to be constituently
`active, phosphorylates Akt at T308, leading to
`stabilization of the activation loop in an active
`conformation. This model strongly resembles the
`general model for PTKs activation.12 T308 phos-
`phorylation is a requirement for the kinase acti-
`vation, but phosphorylation of the residue located
`at the hydrophobic C-terminal region is also re-
`quired to full activation of the kinase. This Akt S473
`kinase (PDK2) has not been identified as yet; how-
`ever, findings from several recent studies suggest a
`
`
`
`195195
`
`role for protein kinase integrin-linked kinase (ILK)
`in the activation process, but it remains unknown
`whether or not ILK directly phosphorylates Akt at
`S473.13–15 In a later step, active Akt is translocated,
`through an unknown mechanism, to the nucleus
`where many of its substrates are localized.16
`
`Cellular processes regulated by Akt
`
`Akt protein modulates the function of numerous
`substrates involved in the regulation of cell sur-
`vival, cell cycle progression and cellular growth
`(Table 1 and Fig. 1).
`
`survival
`
`Cell-death regulation
`Akt activation induces different cell
`mechanisms.17
`Akt inactivates by phosphorylation the proapop-
`totic factors Bad and procaspase-9, as well as the
`Forkhead family of transcription factors that induce
`the expression of proapoptotic factors such as Fas
`ligand. Moreover, very recently Akt activation
`has been related, in cancer cells, with increased
`resistance to apoptosis induced by TRAIL/APO-2L
`(TNF-Related Apoptosis-Inducing Ligand), a mem-
`ber of the TNF superfamily that has been shown to
`have selective antitumor activity.18–21
`On the other hand, Akt activates by phosphory-
`lation the transcription factor cyclic AMP response
`element-binding protein (CREB), and the IjB kinase
`(IKK), a positive regulator of NF-jB, both of them
`regulating the expression of genes with antiapop-
`totic activity.
`
`Cycle progression and cell growth
`Glycogen synthase kinase-3 (GSK3), phosphodis-
`terase-3B, mammalian
`target
`of
`rapamycin
`(mTOR), insulin receptor substrate-1 (IRS-1), the
`Forkhead family member FKHR, Cyclin-dependent
`kinase inhibitors p21CIP1=WAF1 and p27KIP1, and,
`possibly, Raf1, are all Akt targets involved in pro-
`tein synthesis, glycogen metabolism and cell cycle
`regulation.12
`A key mechanism for Akt modulation of specific
`substrate activity, such as b-catenin, p21, p27,
`Mdm2 or Forkhead transcription factors, involves
`the regulation of their cytoplasmic or nuclear lo-
`calization by phosphorylation. GSK3 inhibition by
`Akt prevents the phosphorylation of the cytoplas-
`mic signalling molecule b-catenin, which impedes
`its degradation; hence it is translocated to the
`nucleus. Once in the nucleus, b-catenin combines
`with different transcription factors, like TCF/LEF-
`1, to induce the expression of several genes, such
`as that of the Cyclin D1, which induces cell cycle
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 003
`
`

`
`196
`
`J.A.F. Vara et al.
`
`Akt-regulated cell processes
`Table 1
`Akt substrates
`
`Effect
`
`Survival
`Bad
`
`CREB
`Forkhead Family (Fkhr, Fkhrl1, Afx)
`
`IKK
`
`Procaspase-9
`Ask1
`Mdm2
`
`Cell growth and proliferation
`GSK3
`p21Cip=Waf1, p27Kip
`mTOR/FRAP
`TSC2
`
`Association of Bad with 14-3-3 proteins; suppression of Bad proapoptotic
`activity
`Increased transcription of CREB-regulated survival genes
`Association with 14-3-3 proteins: Nuclear exclusion and inhibition of
`transcription of proapoptotic genes
`Induction of NF-jB transcriptional activity; activation of transcription of
`survival genes
`Suppression of Caspase-9 induced apoptosis
`Inhibition of stress activated kinases
`Mdm2 nuclear entry; inhibition of p53 regulated processes
`
`Inhibition of GSK3 catalytic activity
`Nuclear exclusion; prevention of its antiproliferative activity
`Modulation of mRNA translation
`Modulation of mTOR activity
`
`progression via regulation of RB hyperphosphory-
`lation and inactivation. In a similar way, decreased
`Cyclin D1 phosphorylation by GSK3 promotes the
`stabilization of this protein.22 Akt phosphorylates
`p21 and inhibits its antiproliferative effects by re-
`taining it within the cytoplasm.23 A similar mech-
`anism has been recently described for p27.24–26
`Following stimulation with growth factors, Mdm2 is
`phosphorylated by Akt and enters the nucleus,
`wherein it induces a decrease in both p53 levels
`and its transactivation. In the absence of the tumor
`suppressor p19/p14ARF, the complex Mdm2-p53
`leaves the nucleus and enters the cytoplasm,
`where p53 becomes degraded through an ubiqui-
`tine/proteasome-mediated process.27
`Finally, p70S6K phosphorylation by PDK1 and
`mTOR phosphorylation by Akt constitute processes
`showing a linkage between both signalling path-
`ways. In fact, it has recently been demonstrated
`that some of the transforming effects of PI3K/Akt
`promoting the cell cycle and growth are mediated
`by mTOR/p70S6K cascade: tuberous sclerosis (TSC)
`is an autosomal dominant disorder characterized
`by the formation of hamartomas in a wide range of
`human tissues. Mutation in either the TSC1 or TSC2
`tumor suppressor gene is responsible for both the
`familial and sporadic forms of this disease. Normal
`cellular functions of hamartin and tuberin, en-
`coded by the TSC1 and TSC2 tumor suppressor
`genes, are closely related to their direct interac-
`tions. Akt stimulates growth by phosphorylating the
`tuberous sclerosis complex 2 (TSC2) tumor sup-
`pressor and inhibiting formation of a TSC1:TSC2
`complex. This complex inhibits the p70 ribosomal
`protein S6 kinase 1 (an activator of translation) and
`activates the eukaryotic initiation factor 4E binding
`
`protein 1 (4E-BP1, an inhibitor of translational
`initiation). These functions of TSC1;TSC2 are
`mediated by inhibition of mTOR.28–32
`
`Characteristic cancer processes involving
`PI3K/Akt
`
`As proposed by Hanahan and Weinberg,33 most of
`genetic/epigenetic changes related to tumor phe-
`notype are representative of a finite succession
`of physiologic disturbances, which, collectively,
`cause the cell to become malignant. In this setting,
`as shown in Table 2, Akt-regulated signalling plays
`an outstanding role in numerous processes which
`are known to be characteristic of cancer.
`
`Alterations of PI3K/Akt pathway in
`human cancer
`
`PI3K gene alterations
`
`The major alteration described to occur in the gene
`PI3K is amplification. The gene PIK3C, which en-
`codes the p110a catalytic subunit of PI3K, is lo-
`cated in the chromosome 3q26, a region that is
`frequently amplified in several human cancers.
`Findings from recent studies have confirmed PIK3C
`amplification in ovarian34 and cervix35 cancer.
`
`Akt gene alterations
`
`No modifications or mutations in the akt gene have
`been found in mammals. Nevertheless, various
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 004
`
`

`
`Akt signalling and cancer
`
`
`
`197197
`
`Table 2
`
`Akt implication in different processes characteristic of cancer
`
`Process
`
`Akt function
`
`Growth signal autonomy
`
`Insensitivity to antiproliferative signals
`
`Inhibition of apoptosis
`
`Unlimited replicative potential
`Angiogenesis
`Invasion and metastasis
`
`Akt overexpression or activation may lead to increased response to
`ambient levels of growth factors
`Induces nuclear entry of Mdm2, which leads to inhibition of p53
`regulated processes. Induces cytoplasmic localization of p21Cip=Waf1and
`p27Kip, promoting proliferation. Stabilizes Cyclin D1
`Inactivates the proapoptotic factors Bad and Procaspase-9. Activates
`IKK, activating the transcription of NF-jB regulated antiapoptotic
`genes. Inactivates Forkhead family transcription factors, inhibiting
`proapoptotic gene expression, such as Fas ligand
`Increases telomerase activity by phosphorylation of hTERT
`Pomotes angiogenesis through eNOS activation
`Contributes to invasiveness by inhibiting anoikis and stimulating MMP
`secretion
`
`studies have found akt amplifications in human
`cancers. The first research work describing akt as
`a potential human oncogene detected akt1 in a
`gastric carcinoma. Akt2 gene amplifications have
`been found in ovarian, pancreas, breast and
`tumors.36;37 Overall,
`such
`stomach malignant
`studies suggest that akt amplification, particularly
`that of akt2, may be a frequent event in different
`human cancers. Although one study has demon-
`strated increased Akt2 protein expression in pan-
`creas cancer with akt2 amplification,38 data about
`Akt levels and its activation in clinical materials
`are still scant.
`
`Further PI3K/Akt pathway alterations
`
`Ligand-dependent activation of protein tyrosine
`kinase receptors, receptors coupled with proteins
`G or integrins results in PI3K activation. Such ac-
`tivation may also occur independently of the re-
`ceptor, as
`is
`the case in cells expressing
`constitutively active Ras.39;40 As surface receptors
`are overexpressed or permanently active in many
`human cancers,12
`their downstream signalling
`pathways are also active. One important example
`is the RPTK ErbB2/HER2/Neu, which is overex-
`pressed due to gene amplification in a large num-
`ber of breast cancers and other types of human
`malignant tumors.41 ErbB2 is an orphan receptor
`that has no specific ligands, which acts as a partner
`for dimerization of other ErbB family members.
`ErbB2-formed heterodimers are potent triggers for
`multiple signalling pathways involved in cell pro-
`liferation, invasion and survival.42 When ErbB2 is
`overexpressed,
`it constitutively associates
`to
`ErbB3. ErbB2–ErbB3 dimers, in turn, are potent
`
`triggers for the PI3K/Akt pathway. In fact, tumor
`cells expressing ErbB2 show constitutive Akt ac-
`tivity.43 Thus, recent findings suggest a major role
`for PI3K/Akt pathway in stimulation of prolifera-
`tion and survival in overexpressing ErbB2 cells.44
`Further examples of PI3K/Akt pathway activation
`are mentioned below in relation to therapeutic
`implications.
`Downstream of PI3K are other molecules regu-
`lating this pathway, such as PI-3,4,5-P3 phosphata-
`ses. PTEN (Phosphatase and tensin homologue
`deleted on chromosome 10; also referred to as
`MMAC1, mutated in multiple advanced cancers 1) is
`a phosphatase with dual activity on lipids and pro-
`teins. It was originally identified as a tumor-sup-
`pressor gene. Its main physiologic lipid substrate is
`PI(3,4,5)P3, i.e., the PI3K product. PTEN dep-
`hosphorylates PI(3,4,5)P3 at 30 inositol position and,
`in this way, acts as a negative regulator for PI3K-
`induced signalling. Studies on PTEN overexpression
`in different cell lines suggest that it acts as a tumor
`suppressor by inhibiting cell growth45 and enhancing
`cellular sensitivity for apoptosis and anoikis, the
`latter being a particular type of apoptosis induced
`in epithelial cells due to alterations in integrin-
`extracellular matrix interaction.46 PTEN is fre-
`quently mutated in advanced stages of several
`human tumors, notably in glioblastoma and endo-
`metrial and prostate cancers. In addition, PTEN
`mutations in germ cell lines result in the rare he-
`reditary syndrome referred to as Cowden’s disease,
`which is associated with a higher risk for develop-
`ment of malignant tumors, notably breast cancer.47
`In summary, PTEN negatively regulates Akt acti-
`vation through PI(3,4,5)P3 dephosphorylation;
`thereby, PTEN activity loss leads to permanent
`PI3K/Akt pathway activation.
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 005
`
`

`
`198
`
`Therapeutic implications and emergent
`drugs
`
`Therapeutic implications
`
`To date, tumor resistance to antineoplastic drugs
`has been attributed to increased drug export from
`the cell or enhanced detoxification mechanisms.
`Despite correlations between drug export and de-
`toxification and resistance, it is increasingly rec-
`ognized that these mechanisms cannot account by
`themselves for tumor cell resistance to antineo-
`plastic agents. Currently, we know that chemo-
`therapy agents kill tumor cells by inducing apoptosis
`and there is increasing evidence that the exploita-
`tion of survival pathways, which may have consid-
`erably contributed to the disease onset, may also be
`important in the development of chemotherapy
`drug resistance.48 Survival signals induced by sev-
`eral receptors are mediated mainly by PI3K/Akt,
`hence this pathway may play a major role in drug
`resistance appearance. In fact, there is convincing
`evidence from recent research suggesting that
`PI3K/Akt pathway activation is related to tumor cell
`resistance to both chemotherapy and radiation.
`It has been recently reported that a correlation
`exists between Her2/Neu overexpression and Akt
`activation in breast cancer, and Akt-induced sig-
`nalling has been suggested to increase stress-
`induced apoptosis resistance in cells overexpress-
`ing Her-2/Neu.49 Regarding Her2/Neu, there is a
`clear-cut relationship between its overexpression
`and increased antineoplastic drug resistance.
`Findings from a recent study suggest a relationship
`between Her2-induced Akt activation and in-
`creased tumor cell resistance to chemotherapy
`drug-induced apoptosis via Mdm2 phosphoryla-
`tion.23 Moreover, it has been suggested that acti-
`vation of Akt1 by Her2/PI3K plays an important role
`in mediating multidrug resistance in human breast
`cancer cells and that Akt may therefore be a novel
`molecular target for therapies that would improve
`the outcome of patients with breast cancer.50
`In ovarian cancer, aberrant Akt expression or
`activation in different cell lines has been described
`to be able to confer paclitaxel resistance.51 It has
`also been reported that PI3K inhibition increases
`paclitaxel efficiency in in vivo and in vitro ovarian
`cancer models.52 Moreover, it has been shown that
`integrin-mediated protection to paclitaxel- and
`vincristine-induced apoptosis
`is dependant on
`PI3K/Akt signalling pathway activation.53
`In a recent study on cell lines from non-small
`cell lung cancer (NSCLC), it has also been observed
`that PI3K/Akt pathway constitutive activation
`
`J.A.F. Vara et al.
`
`promotes cell survival, and its genetic or pharma-
`cological modulation alters the response to dif-
`ferent therapeutic approaches.54 In addition, one
`very recently published article describes that Akt is
`a central mediator in the suppression of anoikis and
`modulation of chemotherapy-induced apoptosis in
`NSCLC cells, and therefore proposes Akt as a
`promising target for small molecule inhibitors to
`shift the apoptotic threshold in cancer cells after
`treatment with standard chemotherapy.55
`In pancreatic cancer cell lines, PI3K/Akt path-
`way has been found to be implicated in chemo-
`therapy drug
`resistance,
`and the potential
`therapeutic benefit of PI3K inhibitors combined
`with gemcitabine for pancreas cancer treatment
`have been described.56
`In bladder cancer PTEN overexpression has been
`shown to be able to induce tumor cell growth
`suppression and enhance sensitivity to doxorubicin.
`Therefore, it has now been recognized that PTEN-
`regulated pathways may be therapeutic targets for
`bladder cancer treatment.57
`On the other hand, results from a recent study
`indicate that Akt overexpression/activation is a
`frequent event in human colon cancer, being less
`common in colon tumors with microsatellite in-
`stability. This study suggests that apoptosis inhi-
`bition during sporadic colon cancer carcinogenic
`process can be attributed, at least partially, to
`Akt.58 Moreover, it has been reported that Akt
`phosphorylation status in human colon carcinoma
`correlates with cell proliferation and apoptosis in-
`hibition, as well as with different clinicopathologic
`parameters like invasion grade, vessel infiltration,
`metastases to lymphatic nodes, and stage.59
`Finally, an interesting research work has shown
`that PI3K/Akt pathway activation, evaluated by
`measuring Akt phosphorylation by means of immu-
`nohistochemistry techniques, may be a marker in
`predicting response to radiation therapy in head and
`neck cancer. Findings from this study suggest that
`EGFR-PI3K signalling may lead to radiation resistance
`and that PI3K may constitute a therapeutic target.60
`
`New drugs
`
`As we have seen above, the PI3K/Akt/PTEN pathway
`has become an attractive target for drug develop-
`ment as such agents might inhibit proliferation, and
`reverse the repression of apoptosis and the resis-
`tance to cytotoxic therapy in cancer cells. Inhibitors
`of proteins that are involved in several PI3K/Akt
`signalling pathways have been under development
`for some time, and some have now entered clinical
`trials. These include inhibitors that target both
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 006
`
`

`
`Akt signalling and cancer
`
`
`
`199199
`
`upstream regulators of PI3K/Akt, such as growth
`factor receptors, and downstream effectors, such
`as the components of the mTOR pathway.
`Used alone or in combination with existing
`therapies, these inhibitors of the PI3K/Akt pathway
`will be developed to exploit activation of the PI3K/
`Akt pathway within cancer cells and/or enhance
`the efficacy of existing chemotherapeutic agents.
`
`Inhibitors targeting pathways upstream of
`PI3K/Akt
`
`Even in tumors in which PI3K/Akt pathway is not
`mutationally activated, this cascade might well be
`stimulated by aberrant activation of upstream
`signalling pathways. The components of the regu-
`latory system for PI3K/AKt that have proved most
`amenable to therapeutic intervention are the
`growth-factor-receptor tyrosine kinases, in partic-
`ular, the EGFR and its close relative ERBB2.
`Several approaches have been used to target the
`ERBB family, but by far the progress which has been
`made the most is in two areas: small-molecule ty-
`rosine kinase inhibitors61;62 and humanized anti-
`bodies against the receptor extracellular domains.63
`In general, antibodies bind to the extracellular do-
`main of the receptors, inhibiting their activation by
`ligand, and promoting receptor internalization and
`downregulation, whereas small molecules compet-
`itively inhibit ATP binding to the receptor, thereby
`hindering autophosphorylation and kinase activa-
`tion. Antibodies with intact Fc binding domains
`might also induce antibody-mediated cellular cyto-
`toxicity against the tumor cells.64
`Preclinical models indicate that EGFR expression
`is required, although the degree of expression
`above an undefined threshold does not predict
`sensitivity to EGFR inhibitors.65–69
`At present, the most advanced of the newer
`therapies in clinical development are anti-receptor
`monoclonal antibodies IMC-C225 (cetuximab, Erbi-
`tux; Imclone), and the reversible small-molecule
`inhibitors of EGFR, ZD1839 (gefitinib,
`Iressa;
`AstraZeneca) and OSI-774 (erlotinib,Tarceva; OSI
`Pharmaceuticals) (Table 3).
`ZD1839 has been through several Phase I trials,
`with the most common toxic side effect being skin
`rash and diarrhoea. Four Phase II trials have also
`been completed. Promising single-agent clinical
`antitumor activity has been reported in advanced
`NSCLC, head and neck cancer and prostate carci-
`noma.70 No effect was seen against metastatic
`renal-cell carcinoma. Phase III randomized trials of
`ZD1839, in combination with gemcitabine and cis-
`platin or with paclitaxel and carboplatin, for the
`
`treatment of NSCLC proved disappointing, given
`the expectations from earlier trials, leading to re-
`evaluation of treatment regimens.
`OSI-774 is similar to ZD1839 in its pharmacolog-
`ical characteristics, with around a tenfold lower
`IC50 against EGFR kinase activity in vitro.71 The
`two drugs had similar toxicities in Phase I studies,
`whereas Phase II trials using OSI-774 as a single
`agent produced promising results against NSCLC,
`ovarian, and head and neck cancer. Other EGFR-
`directed small-molecule tyrosine kinase inhibitors
`in early stage trials include PKI116 (Novartis),
`GW2016 (GlaxoSmithKline), EKB-569 (Genetics In-
`stitute/Wyeth–Ayerst) and CI-1033 (Pfizer).
`The other important class of therapeutic agents
`directed against ERBB-family receptors are hu-
`manized monoclonal antibodies. The most ad-
`vanced of this drug type against EGFR is a chimeric
`antibody – IMC-C225 – which is developed by Im-
`Clone Systems and Bristol-Myers Squibb (Cetux-
`imab, Erbitux). Phase I trials showed that the drug
`was reasonably well tolerated,72 and a Phase II trial
`showed promising results in advanced colorectal
`carcinoma. Unfortunately, widely reported regu-
`latory difficulties have delayed further develop-
`ment of this potentially important drug.
`The other
`important anti-receptor drug is
`trastuzumab (Herceptin), which was developed by
`Genentech. This humanized monoclonal antibody
`against ERBB2 has proved to be effective against
`breast carcinomas in which ERBB2 is highly ex-
`pressed – that is, 20–30% of cases of metastatic
`breast cancer. In Phase III trials, it extended the
`median survival of patients from 20.3 months to
`25.1 months.73 Despite some problems with cardiac
`toxicities, trastuzumab has now been licensed in
`the United States and elsewhere as a single agent
`for treating metastatic breast cancer with ERBB2
`overexpression.
`
`Inhibitors targeting Ras
`The covalent attachment of the farnesyl isoprenoid
`group to the HRas, KRas and NRas proteins is es-
`sential for the biological activity of Ras. Therefore,
`it was an obvious target for the design of new ra-
`tional therapies against the Ras pathway.
`A large number of highly effective farnesyl-
`transferase inhibitors (FTIs) have been identified.
`These were shown to efficiently inhibit the farn-
`esylation of HRas in cells in culture, which led to
`high expectations of being effective against the
`20% of human tumors that have activating muta-
`tions in Ras genes. Unfortunately, this early po-
`tential has not been realized. The mode of action
`of FTIs has become increasingly unclear, and the
`initial spectacular successes that were achieved in
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 007
`
`

`
`200
`
`J.A.F. Vara et al.
`
`Table 3
`
`New drugs targeting PI3K/Akt related molecules
`
`Target
`
`Drug
`
`Class
`
`Development stage
`
`Upstream targets
`EGFR
`
`Antibody
`
`Antibody
`Antibody
`
`Antibody
`
`IMC-C225 cetuximab (Erbitux;
`Imclone)
`ABX-EGF (Abgenix)
`EMD 72000 (Merck KgaA
`Darmstadt)
`RH3 (York Medical
`Bioscience Inc.)
`MDX-447 (Medarex/Merck KgaA) Antibody
`ZD1839 gefitinib (Iressa;
`Kinase inhibitor
`AstraZeneca)
`OSI-774 erlotinib (Tarceva;
`OSI-Pharmaceuticals)
`PKI116 (Novartis)
`CI-1033/PD183805 (Pfizer)
`EKB-569 (Wyeth–Ayerst)
`GW2016/572016
`(GlaxoSmithKline)
`
`Kinase inhibitor
`
`Kinase inhibitor
`Kinase inhibitor
`Kinase inhibitor
`Kinase inhibitor
`
`Phase III
`
`Phase II
`Phase I
`
`Phase II
`
`Phase I
`Phase III
`
`Phase III
`
`Phase II
`Phase I
`Phase I
`Phase I
`
`HER-2/Neu
`
`BCR-ABL/PDGFR/c-Kit
`
`PDK1
`
`Ras
`
`Specific targets
`PI3K
`
`Dowstream targets
`mTOR
`
`Trastuzumab (Herceptin;
`Genentech)
`MDX-210 (Medarex/Novartis)
`2C4 (Genentech)
`17-AAG (Kosan)
`
`Imatinib (STI571/Gleevec;
`Novartis)
`
`Antibody
`
`Registered
`
`Antibody
`Antibody
`HSP90 inhibitor
`
`Phase I
`Phase I
`Phase I
`
`Kinase inhibitor
`
`Registered
`
`UCN-01 (Kyowa Hakko Kogyo)
`
`Staurosporine analogue
`
`Phase I/II
`
`Phase II
`ISIS 2503 (Isis Pharmaceuticals) Antisense oligonucleotide
`R115777 (Johnson and Johnson) Farnesyl transferase inhibitor Phase II/III
`SCH66336 (Schering-Plough)
`Farnesyl transferase inhibitor Phase II
`BMS214662 (Bristol-Myers
`Farnesyl transferase inhibitor Phase I
`Squibb)
`
`Wortmannin
`LY294002
`
`PI3K Inhibitor
`PI3K Inhibitor
`
`CCI-779 (Wyeth)
`
`RAD001 (Novartis)
`
`Rapamycin/sirolimus (Wyeth)
`
`Inhibits mTOR kinase by
`binding to FKBP12
`Inhibits mTOR kinase by
`binding to FKBP12
`
`Inhibits mTOR kinase by
`binding to FKBP12
`
`Preclinical
`Preclinical
`
`Phase II
`
`Phase I as a cancer
`therapeutic Phase II/III
`as an immunosuppressant
`Registered as an
`immunosuppressant
`
`mouse models have not been repeated in human
`patients.
`Despite uncertainty about their mechanism of
`function, FTIs do have marked effects on the
`
`growth and survival of some tumor cell lines in
`vitro and on xenografts in nude mice, although not
`necessarily those expressing activated Ras. The
`effects of FTIs in these pre-clinical systems have
`
`Roxane Labs., Inc.
`Exhibit 1023
`Page 008
`
`

`
`Akt signalling and cancer
`
`
`
`201201
`
`recently been reviewed extensively.74;75 These
`data have prompted the launch of several clinical
`trials using FTIs, even though their real target is
`still controversial.
`A different approach to the therapeutic tar-
`geting of the Ras pathways is to inhibit the ex-
`pression of Ras using short antisense synthetic
`oligonucleotides that are specific for sequences in
`the mRNAs for these proteins.76 Early trials in
`nude m

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