throbber
Signal Transduction Pathway Targets for Anticancer Drug Discovery
`
`Current Pharmaceutical Design, 2000, 6, 361-378
`
`361
`
`Alex A. Adjei*
`
`Mayo Clinic, Division of Medical Oncology, 200 First Street S. W., Rochester, MN 55905,
`USA
`
`Abstract: There are currently over 80 agents officially approved for the treatment
`of cancer world-wide. However, the most common epithelial cancers, which cause
`greater than 75% of cancer deaths, remain incurable. Most drugs have been developed
`empirically by testing large numbers of chemicals on rapidly growing transplantable
`rodent tumors, and more recently, human tumor xenografts. This approach has
`identified prodeminantly DNA-active drugs that are considerably toxic and have
`limited efficacy. Novel molecular targets, which are selective for neoplastic cells, are needed for
`chemotherapeutic agents to improve cure rates of epithelial malignancies, with acceptable toxicity. In
`recent years, agents inhibiting signal transduction pathway molecules have entered clinical trials. These
`include antibodies and small molecules, which inhibit growth factor receptors and their receptor tyrosine
`kinases, inhibitors of cytoplasmic second messengers such as ras, raf and MEK, inhibitors of protein
`trafficking, and inhibitors of protein degradation.
`
`INTRODUCTION
`
`Cell proliferation and differentiation are
`regulated by a number of hormones, growth factors
`and cytokines. These molecules interact with
`cellular receptors and communicate with the nucleus
`of the cell through a network of intracellular
`signaling pathways (Fig 1). In cancer cells, key
`components of these pathways may be altered by
`oncogenes through over-expression or mutation,
`leading to disregulated cell signaling and cell
`proliferation. The components of these abnormal
`signaling pathways, which are specific to neoplastic
`cells, represent potential selective targets for new
`anticancer therapies. These potential targets include
`ligands (typically growth factors), cellular receptors,
`intracellular second messengers and nuclear
`transcription factors. A detailed description of all
`these possible targets is beyond the scope of this
`review. The interested reader is referred to several
`recent, excellent reviews [1-5]. This discussion will
`focus exclusively on
`the
`targets for which
`promising anti-neoplastic agents are in clinical
`trials.
`
`INHIBITION OF GROWTH FACTOR
`RECEPTOR BINDING
`
`The first, obvious point of intervention in a
`signaling cascade is the neutralization of ligands
`
`*Address correspondence to this author at the Division of Medical
`Oncology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA;
`Telephone:
`507-538-0548;
`Fax:
`507-284-1803;
`E-mail:
`adjei.alex@mayo.edu
`
`before they can associate with their receptors. This
`approach has been investigated, initially through the
`use of antibodies, which interact with growth
`factors. Small cell
`lung cancer, and other
`malignancies secrete bombesin-like peptides, such
`as gastrin releasing peptide. A recently published
`phase I trial established that repeated doses of
`monoclonal antibody 2A11, which binds to the
`bombesin-like peptide, GRP with high affinity,
`could be given safely to SCLC patients, and
`sustained plasma levels could be achieved on a 1-
`week schedule of antibody administration [6]. This
`approach is limited by the need to prospectively
`identify patients whose tumors express the receptor,
`and whose plasma contains significant amounts of
`circulating growth factor. Non-specific inhibition of
`several growth factors was explored through the use
`of the polysulfonated naphthylurea, suramin [7].
`This agent neutralized a number of growth factors,
`but presented challenging dosing problems because
`of an extremely long half-life and significant
`toxicities. In spite of evidence of modest activity in
`prostate cancer,
`its development has been
`discontinued, because of toxicity problems.
`
`The second approach to abrogating signaling
`pathways is the prevention of the binding of growth
`factors to their receptors. Several strategies have
`been under development in an attempt to block
`growth factor receptors. The most successful
`approach to date has been the development of
`monoclonal antibodies which bind to receptors and
`by so doing prevent the binding of the endogenous
`ligands.
`
`1381-6128/00 $19.00+.00
`
`© 2000 Bentham Science Publishers B.V.
`
`NOVARTIS EXHIBIT 2014
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 18
`
`

`

`362 Current Pharmaceutical Design, 2000, Vol. 6, No. 4
`
`Alex A. Adjei
`
`(1). Schematic representation of some important signal transduction pathways in cancer cell proliferation. DAG,
`Fig.
`diacylglecerol; IP3, inositide triphosphate; PI3K, phosphoinositide-3-kinase; PLC, phospholipase C; PIP2, phosphoinositide
`diphosphate; PKC, protein kinase C; MEK, mitogen-activated protein kinase kinase; MAP kinase, mitogen-activated protein
`kinase.
`
`The Epidermal Growth Factor Receptor
`(EGFR) Family
`
`The epidermal growth factor (EGF) receptor was
`cloned in 1984 by Ullrich et al. [8]. This receptor
`has two cysteine-rich regions in the extracellular
`domain and a single kinase domain. Three other
`members of this family, HER-2, HER-3 and HER-4
`(referred to also as erbB2, erbB3 and erbB4) are
`known. The designation HER-2 stands for Human
`Epidermal growth factor-like Receptor type 2.
`Members of the EGF receptor family and their
`ligands are overexpressed or expressed as an
`autocrine loop in a number of tumor types,
`including pancreatic, lung, ovarian, renal cell, gastric,
`hepatocellular and breast cancers [10-12].
`
`Anti-EGFR Antibodies
`
`Because over-expression of EGFR has been
`associated with a more aggressive disease and a
`poor prognosis, the blockade of EGFR activation
`has been proposed as a target for anticancer
`therapy. The most promising agent is the human-
`mouse chimeric monoclonal antibody 225 (C225),
`which inhibits activation of the EGFR receptor
`tyrosine kinase. This inhibition of EGFR activation
`
`causes cell cycle arrest in G1. The mechanism of
`growth inhibition has been shown to involve an
`elevation in the levels of p27KIP1 and inhibition of
`cyclin-dependent
`kinase-2
`activity
`[13].
`Preclinically, C225 in combination with several
`chemotherapy
`agents,
`including
`cisplatin,
`doxorubicin and paclitaxel exhibited synergistic
`antitumor activity, with successful eradication of
`well-established
`tumor xenografts
`that were
`resistant to treatment with either C225 or drug alone
`[14]. Phase I clinical trials have established the
`safety of repeated administration of single-agent
`C225 at concentrations that maintain receptor-
`saturating blood levels for up to 3 months [15].
`Phase I
`trials exploring C225
`treatment in
`combination with
`the chemotherapy agents
`mentioned above, are ongoing [16], and single-
`agent phase II trials are in progress. Preliminary
`data indicate that antitumor activity is significantly
`augmented when the antibody
`is utilized
`in
`combination with cytotoxic chemotherapy.
`
`Anti-HER-2 Antibodies
`
`The proto-oncogene HER-2/neu is localized to
`chromosome 17q, and encodes a 185kDA
`
`NOVARTIS EXHIBIT 2014
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 18
`
`

`

`Signal Transduction Pathway Targets
`
`transmembrane glycoprotein receptor with intrinsic
`tyrosine kinase activity. No endogenous ligands for
`the HER-2/neu protein are known. When HER-
`2/neu protein is activated, it can interact with many
`different cellular proteins such as shc, PLC, GAP
`and the ras MAP kinase pathway [17]. The HER-2
`receptor can also form heterodimers with other
`members of the EGFR family. Amplification of the
`HER-2/neu gene or over-expression of the HER-
`2/neu protein has been identified in 10-34% of
`breast cancers. Possible techniques for evaluating
`HER-2/neu status in breast cancer cells include
`gene-based assays such as polymerase chain
`reaction methods and in-situ hybridization utilizing
`both fluorescent (FISH) and non-fluorescent
`approaches. Qualitative protein measurements are
`the
`most
`common
`techniques
`used.
`Immunohistochemistry,
`typically on archival
`tissues
`is utilized. A significant discordance
`between HER-2/neu detection methods has been
`reported [18]. The discordance has been between
`immunohistochemistry methods and between
`immunohistochemistry and gene-based assays [19].
`A number of clinical studies involving over 10,000
`women have examined the relationship of HER-
`2/neu gene and/or protein abnormalities and breast
`cancer outcome. Results of these studies have not
`been uniform. Several studies including the original
`study published by Slamon et al. [20] found that
`HER-2/neu
`over-expression
`independently
`predicted poor overall survival and disease-free
`survival. Some immunohistochemical studies have
`found significant correlation between HER-2/neu
`protein immunoreactivity and disease outcome in
`univariate, but no independent predictive status in
`multivariate analysis [21]. A few studies have
`found no correlation whatsoever with disease
`outcome [22.23]. Compared to the prognostic
`information outlined above, there are fewer studies
`correlating the expression of HER-2/neu protein
`with response to therapy. Several studies have
`found HER-2/neu over-expressing tumors to be
`resistant to tamoxifen therapy [24, 25]. One large
`study in 200 patients, however, failed to show
`resistance
`to
`tamoxifen
`in HER-2/neu over-
`expressing tumors [26]. While HER-2/neu over-
`expression has been associated with enhanced
`response to chemotherapy regimens containing
`doxorubicin in clinical samples, a poor response to
`CMF
`(cyclophosphamide, methotrexate, 5-
`fluorouracil) therapy has been found in the same
`population of patients [27]. In cultured breast
`cancer cell
`lines, HER-2/neu expression
`is
`associated with resistance to paclitaxel [28].
`However, another study
`indicated a 3-fold
`increased response to paclitaxel in the same
`population [29]. The preponderance of evidence
`
`Current Pharmaceutical Design, 2000, Vol. 6, No. 4 363
`
`would suggest a poorer prognosis and poor
`response to some therapeutic agents in patients with
`HER-2/neu expressing tumors. The conflict in the
`data may be explained in part, by the different
`methods used to document HER-2/neu expression.
`
`HER-2,
`
`(rhuMAb
`
`Trastuzumab
`Herceptin(cid:210) )
`is a humanized monoclonal
`Trastuzumab
`antibody that targets the HER-2 receptor with
`demonstrated activity in metastatic breast cancer.
`This is the first monoclonal antibody to be
`approved for the treatment of a solid tumor. In
`cultured cells that express high levels of HER2,
`trastuzumab causes growth arrest in the G0/G1
`phase of the cell cycle [30]. The growth inhibitory
`effects have been explained by a marked induction
`of the cyclin dependent kinase-2 kinase inhibitor,
`p27, as well as the retinoblastoma-related protein
`p130 [30]. These data suggest that treatment of
`HER-2 overexpressing cells is antiproliferative, and
`that cytostasis may result from an inhibition of cell
`cycle progression.
`
`In early preclinical studies, Slamon et al.
`demonstrated interactions between trastuzumab and
`several anticancer agents [31], using in vitro
`clonogenic studies. Synergistic
`interactions at
`clinically relevant drug concentrations were
`observed for trastuzumab in combination with
`cisplatin, thiotepa and etoposide. Additive cytotoxic
`effects were observed with trastuzumab plus
`doxorubicin,
`paclitaxel, methotrexate
`and
`vinblastine. 5-fluorouracil, was found to be less
`than additive with trastuzumab. Studies were
`further
`conducted with
`drug/trastuzumab
`combinations in HER-2/neu-transfected, MCF-7
`human breast cancer xenografts in athymic mice.
`Combinations
`of
`trastuzumab
`and
`cyclophosphamide, doxorubicin
`, paclitaxel,
`methotrexate, etoposide, and vinblastine in vivo
`resulted in a significant reduction in xenograft
`volume compared
`to
`chemotherapy
`alone.
`Combinations of trastuzumab and 5-fluorouracil
`yielded equivalent results to those achieved by 5-
`fluorouracil alone. The 5-FU
`results were
`consistent with the sub-additive effects observed
`with this combination in vitro. The synergistic
`interaction of trastuzumab with alkylating agents,
`platinum analogs and topoisomerase II inhibitors,
`as well as the additive interaction with taxanes,
`anthracyclines and some antimetabolites in HER-
`2/neu over-expressing breast cancer cells guided the
`choice of combination studies in clinical trials.
`Currently, data from six breast cancer trials have
`
`NOVARTIS EXHIBIT 2014
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 18
`
`

`

`364 Current Pharmaceutical Design, 2000, Vol. 6, No. 4
`
`Alex A. Adjei
`
`been published. Three studies have evaluated
`trastuzumab alone for the treatment of metastatic
`breast cancer. Two studies evaluated trastuzumab in
`refractory breast cancer [32]. Response rates were
`11.6 and 15%, respectively [33]. The antibody was
`well tolerated. The third trial evaluated trastuzumab
`as first-line treatment of metastatic breast cancer. A
`preliminary response rate of 24% has been reported
`[34]. The pivotal multinational phase III study
`reported by Slamon et al. evaluated trastuzumab –
`paclitaxel, or doxorubicin/cyclophosphamide in 469
`first-line metastatic breast cancer patients. Overall,
`the median time to progression was improved from
`4.6 to 7.6 months by the addition of trastuzumab,
`with an improvement in overall response from 32%
`to 48%. Toxicity was generally mild, consisting of
`febrile
`episodes
`and mildly
`increased
`myelossuppression. An increased incidence of
`symptomatic cardiac toxicity was noted when
`trastuzumab was added to anthracycline-based
`chemotherapy (19% vs. 3%). The etiology of this
`increased cardiotoxicity is unclear, but continues to
`be investigated [35]. Trastuzumab plus cisplatin was
`evaluated in patients with refractory breast cancer,
`yielding a response rate of 24% [36].
`
`INTRACELLULAR
`PATHWAYS
`
`SIGNALING
`
`The intracellular signaling pathways that are
`activated after growth factors and other cell-
`proliferation-associated
`ligands bind
`to
`their
`receptors are complex and incompletely understood.
`Major components of these effector systems such
`as the protein tyrosine kinases, protein kinase C and
`the ras/MAP kinase pathway have, however, been
`identified.
`
`PROTEIN TYROSINE KINASES
`
`Protein tyrosine kinases (PTKs) catalyze the
`phosphorylation of tyrosine residues on target
`proteins [37]. Two major groups of PTKs have
`been described to date, receptor and non-receptor
`tyrosine kinases. Non-receptor tyrosine kinases are
`cytoplasmic proteins which transduce extracellular
`signals to downstream intermediates in pathways
`that
`regulate
`cell growth,
`activation
`and
`differentiation. Many non-receptor tyrosine kinases
`are linked to transmembrane receptors including
`those for peptide hormones and cytokines. Unlike
`receptor tyrosine kinases, they lack transmembrane
`domains and ligand binding. They are activated by
`ligand binding to their associated receptors or
`events such as cell adhesion, calcium influx or cell
`cycle progression [38]. More than 30 members are
`
`classified in 10 families including src, abl, JAK,
`MKK2, FES [38].
`
`Receptor tyrosine kinases (RTKs) share several
`structural
`features. They
`are glycoproteins
`possessing an extracellular ligand-binding domain,
`which conveys ligand specificity, and a single
`hydrophobic
`transmembrane domain, which
`anchors the receptor to the membrane. Intracellular
`sequences typically contain regulatory regions in
`addition to the catalytic domain. Ligand binding
`induces activation of the intracellular tyrosine kinase
`domain leading to the initiation of signaling events
`specific for the receptor. The RTKs have been
`organized
`into families based on
`sequence
`homology, structural characteristics and distinct
`motifs in the extracellular domain. There are
`currently 19 known families in vertebrates. The
`various subfamilies include receptors for epidermal
`growth factor (EGFR), platelet derived growth
`factor (PDGF), vascular endothelial growth factor
`(VEGF), fibroblast growth factor (FGF) and
`hepatocyte growth factor (HGF). Ligand binding to
`a RTK
`induces
`receptor dimerization with
`conformational changes that result in intermolecular
`phosphorylation at tyrosine residues at multiple
`sites. Receptor heterodimerization can also occur, as
`reported with transforming growth factor alpha
`interaction with receptor heterodimers comprising
`HER-2 and EGFR [39]. In malignant tumors, a
`number of these receptors are over-expressed or
`mutated, leading to abnormal cell proliferation.
`
`PLATELET DERIVED GROWTH FACTOR
`(PDGF)
`
`PDGF is a major mitogen for endothelial cells,
`fibroblasts, smooth muscle cells and glial cells.
`PDGF exists as disulfide-linked homodimers and
`heterodimers of A and B chains, resulting in 3
`isoforms (PDGF-AA, PDGF-BB, PDGF-AB).
`PDGF and its receptors are expressed in a wide
`variety of cultured neoplastic cells including breast,
`prostate and colon cancers. Expression has also
`been documented in tumor biopsies of ovarian
`cancer and gliomas [40].
`
`The tyrphostins are synthetic protein tyrosine
`kinase inhibitors derived from erbstatin, a natural
`product with broad spectrum activity against protein
`tyrosine kinases and protein kinase C. SU101 is a
`tyrphostin derivative, which predominantly inhibits
`the PDGF receptor tyrosine kinases. In a completed
`phase I study, the most common toxicities were
`mild to moderate nausea, vomiting, and fever.
`Neutropenia was uncommon, and occurred only at
`
`NOVARTIS EXHIBIT 2014
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 18
`
`

`

`Signal Transduction Pathway Targets
`
`the highest dose levels [41]. Phase III studies are
`ongoing in recurrent gliomas, and phase II studies
`have been completed in lung, prostate and ovarian
`cancers [42].
`
`VASCULAR ENDOTHELIAL GROWTH
`FACTOR (VEGF)
`
`VEGF has 5 isoforms, which are splice variants
`and exist as disulfide-linked homodimers, with
`some structural similarities to PDGFs. These
`isoforms bind with high affinity to 2 receptors,
`fms-like tyrosine kinase (flt-1) and fetal liver kinase
`(flk-1). The biologic significance of these multiple
`VEGF receptor forms is not understood. VEGF
`stimulates the growth of endothelial cells during the
`process of angiogenesis, but has also been
`identified as a vascular permeability factor. A
`recombinant humanized monoclonal antibody,
`rhuMAb VEGF, has been developed to inhibit the
`effects of VEGF in the treatment of solid tumors.
`Phase II trials have been completed in lung and
`colon cancer. Results are awaited with interest A
`small molecule inhibitor of VEGF tyrosine kinase,
`SU5416 is undergoing phase I testing, and a phase
`I/II study in AIDS-related Kaposi's sarcoma is
`ongoing [43].
`
`A second generation broad spectrum RTK
`inhibitor, SU6668 is currently undergoing phase I
`testing. This agent is a small organic molecule, that
`possesses anti-angiogenic and anti-proliferative
`properties. It inhibits the autophosphorylation of
`three distinct
`tyrosine kinases, Flk-1/KDR;
`PDGFR, and FGFR with IC50 values of 0.2 m M,
`0.2 m M, and 4.1 m M, respectively. EGFR kinase
`activity remains uninhibited (IC50 > 100 m M). In
`vitro kinetic analyses demonstrate that SU6668 is a
`competitive
`inhibitor of ATP binding. This
`mechanism is similar to that of the bioflavonoids
`such as genistein and quercetin, and distinct from
`the
`tyrphostins and
`their derivatives, which
`compete for the substrate binding site of RTKs
`[44].
`
`Current Pharmaceutical Design, 2000, Vol. 6, No. 4 365
`
`GROWTH
`EPIDERMAL
`RECEPTOR (EGFR)
`
`FACTOR
`
`EGFR differs from the other receptor tyrosine
`kinases in that there is a single isoform, from a
`single 26 exon gene located across 110kb on
`chromosome 7p11-13. It serves as the predominant
`receptor for multiple distinct ligands, including
`EGF, TGF-a
`, amphiregulin and HB-EGF (Table
`1). EGFR interacts with most members of the
`EGFR (c-erbB) family of RTKs. As previously
`mentioned, the ligand for c-erbB2 is unknown,
`while erbB3 and erbB4 serve as heregulin and
`neuregulin receptors. The major function of these
`other receptors appears to be as downstream
`effectors of each other. They cross-phosphorylate
`and modulate signaling from each other in specific
`pairs. EGFR interacts with HER-2 and HER-3 but
`not HER-4. HER-4 pairs with HER-2.
`
`It is noteworthy that HER-3 lacks kinase
`activity, but serves as a docking protein to recruit a
`broader spectrum of downstream effectors after
`phosphorylation by EGFR or HER-2
`[45].
`Currently, 3 EGFR tyrosine kinase inhibitors are
`undergoing phase I clinical testing. One of these is
`ZD1839, which is an orally active, and selective
`inhibitor of the EGFR (HER-1) tyrosine kinase. In
`preclinical studies, administration of this agent daily
`for 4 months resulted in significant tumor growth
`delay in rodents bearing human xenografts. Ex vivo
`examination of xenograft tissues revealed a time
`and dose-dependent decrease in c-fos mRNA, a
`marker for EGFR signaling [46]. An ongoing
`phase I trial has enrolled 38 patients. A partial
`response has been noted in NSCLC, with minor
`responses in head and neck and renal cell cancers.
`The most common toxicity is a skin rash [47, 48].
`The second agent in this class, CP-358774 is also a
`selective EGFR tyrosine kinase inhibitor. An
`acneform skin rash, diarrhea and mild hepatic
`transaminase elevations were the most common
`toxicities [48]. The third agent in this class, CI-
`1033 is a non-specific inhibitor of the EGFR family
`(HER-1, HER-2, HER-4) tyrosine kinases [49].
`
`Table 1. Members of the Human Epidermal Growth Factor Gene Family
`
`Gene
`
`HER-1 (c-erbB-1)
`
`HER-2 (c-erbB-2)
`
`HER-3 (c-erbB-3)
`
`HER-4 (c-erbB-4)
`
`Ligand
`
`EGF, TGFa
`
`, Beta cellulin, Amphiregulin, Heparin binding growth factor
`
`? Heregulin
`
`Heregulin, neu differentiation factor 1+2
`
`Heregulin, neu differentiation factor 1+2
`
`NOVARTIS EXHIBIT 2014
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 18
`
`

`

`366 Current Pharmaceutical Design, 2000, Vol. 6, No. 4
`
`Alex A. Adjei
`
`This agent demonstrates an in vitro IC 50 in the low
`nanomolar range with all four RTKs. Results of
`phase I studies with this agent are awaited with
`interest.
`
`PROTEIN KINASE C INHIBITORS
`
`is a family of
`Protein kinase C (PKC)
`serine/threonine directed protein kinases consisting
`of a group of at least 10 related isoenzymes that are
`involved in the transduction of signals for cell
`proliferation and differentiation. PKC has therefore
`been considered a suitable
`target for novel
`antineoplastic drugs. The PKC family is divided
`into Ca2+-responsive
`and Ca2+-unresponsive
`subtypes [50, 51]. The enzyme consists of an N-
`terminal regulatory domain, which binds and
`inhibits the C-terminal catalytic domain.
`
`PKC modulatory agents, which affect the
`catalytic domain, are the staurosporine congeners.
`Staurosporine is the parent compound derived from
`microbial sources two decades ago, and has poor
`selectivity. It is inhibitory towards tyrosine kinases
`and cAMP-dependent protein kinases as well as
`PKC at similar concentrations [52]. Its analogs 7-
`hydroxy-staurosporine (UCN-01) and CGP 41251
`have greater selectivity, and effectively arrest the
`growth of several human-derived tumor cell lines in
`vitro. They also possess antineoplastic activity in
`vivo in human tumors grown as xenografts in nude
`mice. CGP 41251 reverses the multidrug-resistance
`phenotype of cancer cells [53]. Both agents are
`currently under clinical evaluation as potential
`antitumor drugs. These agents are also potent
`modulators of the cyclin dependent kinase system,
`which determines the progression of cells through
`the cell cycle. The nature of this interaction is
`complex. UCN-01 blocks cells in GI phase by
`promoting accumulation of dephosphorylated
`retinoblastoma protein as a consequence of
`inhibition of the activity of certain cyclin-dependent
`kinases, down-regulation of their partner cyclins
`and an increase in the expression of cyclin-
`dependent
`kinase
`inhibitor
`proteins
`[54].
`Preliminary results of early clinical trials suggest
`that UCN-01 and CGP41251 are without
`remarkable toxicity but display high binding to
`human plasma protein [55].
`
`Several studies indicate a role for PKC in the
`regulation of the multi-drug resistance (MDR)
`phenotype, since several PKC inhibitors are able to
`partially reverse MDR and inhibit P-glycoprotein
`(Pgp) phosphorylation. The MDR phenotype is
`also associated with variation in PKC iso-enzyme
`
` over-
`in particular with PKC-
`content,
`expression. Based on these results, it has been
`hypothesized that PKC inhibitors may synergize
`with cytotoxic agents through modulation of MDR.
`However, other potential mechanisms of PKC
`interaction with anticancer drugs exist and have
`been documented, such as the enhancement of
`chemotherapy-induced apoptosis by safingol, a
`specific PKC inhibitor [56]. Safingol is undergoing
`a phase I clinical trial in combination with
`doxorubicin. While no final data are presently
`available, it appears that plasma levels of safingol
`approach those associated with potentiation of
`chemotherapy in animals, and no pharmacokinetic
`interaction between the two drugs exists. Drugs
`targeting PKC are therefore felt to possess potential
`as modulators of cytotoxic agents.
`
`Inhibitors of the regulatory domain of PKC
`include the ether lipids and bryostatins. Bryostatin
`1, a macrocyclic lactone isolated from the marine
`organism Bugula nerutina, is a partial PKC agonist,
`and has shown potent antineoplastic properties in
`in vivo. Bryostatin 1 has both
`vitro and
`antineoplastic and immune-stimulatory properties,
`including the induction of cytokine release and
`expansion
`of
`tumor-specific
`lymphocyte
`populations. Bryostatin
`I has demonstrated
`antitumor activity in phase I trials in patients with
`malignant melanoma,
`lymphoma and ovarian
`carcinoma and is undergoing broad phase II testing,
`both as a single agent, and in combination with
`standard cytotoxic agents [57, 58]. The dose-
`limiting toxicity is myalgia. In a completed phase II
`study, 16 previously treated patients with malignant
`melanoma were treated with bryostatin at 25 m g/m2
`weekly for three courses followed by a rest week.
`The principal toxicities were myalgia, phlebitis,
`fatigue and vomiting. Of 15 patients evaluable for
`tumor response, 14 developed progressive disease.
`One patient developed stable disease for 9 months
`after bryostatin treatment. The authors concluded
`that single-agent bryostatin was ineffective in the
`treatment of metastatic melanoma in patients
`previously
`treated with
`chemotherapy, but
`suggested that it should, however, be investigated
`further in previously untreated patients [59].
`
`antisense
`an
`641A),
`(ISI
`3521
`ISIS
`phosphorothioate oligonucleotide to protein kinase
`C-a
`, has been studied in a phase I trial. The
`antisense approach involves targeting specific
`ribonucleotide (RNA) sequences in order to block
`translation of the RNA message into protein. Drug-
`related toxicities included mild to moderate nausea,
`vomiting, fever, chills, and fatigue, hematologic
`toxicity was
`limited
`to mild
`to moderate
`
`NOVARTIS EXHIBIT 2014
`Breckenridge v. Novartis, IPR 2017-01592
`Page 6 of 18
`
`a
`

`

`Signal Transduction Pathway Targets
`
`Current Pharmaceutical Design, 2000, Vol. 6, No. 4 367
`
`thrombocytopenia. Dose escalation on this study
`was discontinued on attaining peak plasma
`concentrations which approached that associated
`with complement activation in primates. No dose
`limiting toxicity was identified, and clinical activity
`was observed in two patients with non-Hodgkin's
`lymphoma who achieved complete responses [60].
`
`mutations are found in bladder, kidney and thyroid
`carcinomas; and N-ras mutations are found in
`melanoma,
`hepatocellular
`carcinoma
`and
`hematologic malignancies [67]. Association of ras
`with
`the plasma membrane, and consequent
`activation require prenylation, i.e. the attachment of
`a farnesyl group, at the carboxy-terminus.
`
`ras/MITOGEN
`THE
`PROTEIN KINASE PATHWAY
`
`ACTIVATED
`
`ras INHIBITORS
`
`As previously mentioned, effector molecules
`recruited by receptor tyrosine kinases (RTKs)
`include phospholipase C (PLC), phosphoinositide-
`3-kinase (PI3-kinase) and ras. The ras pathway is
`fairly well understood. The effector molecules bind
`to phosphorylated tyrosine residues on the RTKs
`via src homology 2 (SH2) domains or an adapter
`protein, Grb2/SOS in the case of ras. Activated ras
`activates raf, which is a serine/threonine kinase.
`Raf activates mitogen activated protein kinase kinase
`(MEK, MAPKK) which
`in
`turn activates
`mitogen activated protein (MAP) kinase. MAP
`kinase activation results in phosphorylation and
`activation of ribosomal S6 kinase and transcription
`factors such as c-jun, c-myc and c-fos, resulting in
`the switching on of a number of genes associated
`with proliferation [61-63]. Signaling by
`this
`pathway is more complex than outlined above,
`since ras and raf-independent pathways for MAP
`kinase activation exist. Secondly, a subfamily of 54
`kDa MAP kinases which function independently of
`MEK has been identified [64].
`
`The ras protein plays an essential role in signal
`transduction,
`as
`already
`described. Three
`mammalian ras proto-oncogenes encode four
`related and highly conserved proteins, H-ras, N-
`ras, and the splice variants K-ras 4A, and K-ras 4B
`[65]. The p21ras proteins bind guanosine
`triphosphate (GTP) and guanosine diphosphate
`(GDP) with high avidity. When GTP is bound, the
`ras protein is in an “active” state. When GDP is
`bound in the resting state, p21ras is inactive. One
`means of negative regulation of p21ras activity is via
`the GTPase-activating protein (GAP). Single point
`mutations of the ras gene can lead to its constitutive
`activation. These mutated forms have impaired
`GTPase activity. Although they bind GAP, there is
`no “off” sign since GTPase is not activated. This
`results
`in continuous stimulation of cellular
`proliferation, and inhibition of apoptosis [66].
`Oncogenic ras mutations have been identified in
`approximately 30% of human cancers [67]. K-ras
`mutations are frequent in non-small cell lung,
`colorectal and pancreatic carcinomas; H-ras
`
`Because of the high percentage of human tumors
`harboring oncogenic ras mutants, interrupting the
`ras signaling pathway has been a major focus of
`new drug development efforts. The current
`promising approaches taken are i) the inhibition of
`ras
`protein
`expression
`through
`antisense
`oligonucleotides, ii) the prevention of membrane
`localization of ras, and iii) the inhibition of ras
`function through inhibition of downstream ras
`effectors.
`
`Antisense Oligonucleotides
`
`Oligonucleotides that are complementary to
`messenger RNA (mRNA)
`transcripts of
`the
`activated ras oncogene, have been utilized to
`decrease
`ras
`protein
`expression.
`These
`oligonucleotides hybridize
`to complementary
`mRNA sequences and decrease protein expression
`through a variety of mechanisms, including RNase
`H-mediated cleavage of hybridized ras mRNA.
`Roth and co-workers developed a retroviral
`antisense K-ras expression vector, which has been
`shown to eliminate the expression of human ras
`protein in lung cancer cells [68]. This K-ras
`retroviral construct was successfully administered
`intratracheally to nude mice bearing implanted
`human lung cancers. Significant activity was
`observed with 87% of treated mice being tumor-
`free compared to 10% of control mice [69]. A
`persistent problem had been the inability to deliver
`intact antisense RNA agents into tumor cells. In
`recent years, the distribution of phosphorothioate
`oligodeoxynucleotides, presumably by endocytosis
`in rodent tissues after intravenous administration
`has been demonstrated [70]. An investigational
`phosphorothioate antisense oligodeoxynucleotide,
`ISIS 2503 designed to hybridize to the 5’-
`untranslated region of human H-ras mRNA, has
`completed testing in phase I clinical trials. Interim
`results indicate a tolerable toxicity profile, with
`moderate thrombocytopenia and fatigue as the only
`adverse events. An indication of biologic activity
`has been seen, with mild tumor shrinkage in a
`patient with sarcoma [71]. The schedule that has
`
`NOVARTIS EXHIBIT 2014
`Breckenridge v. Novartis, IPR 2017-01592
`Page 7 of 18
`
`

`

`368 Current Pharmaceutical Design, 2000, Vol. 6, No. 4
`
`Alex A. Adjei
`
`Fig. (2). Simplified scheme of signal transduction pathways, with an indication of the molecules to which clinical agents are
`targeted.
`
`1) anti-receptor antibodies such as trastuzumab, receptor tyrosine kinase inhibitors such as monoclonal antibody C225,
`ZD1839, CP258774 and CI-1033
`
`2) farnesyltransferase inhibitors (SCH66336, R115177, L748259, BMS214662); antisense oligonucleotides (ISIS 2503)
`
`3) antisense oligonucleotides (ISIS 5132), HSP90 inhibitor (17AAG)
`
`4) MEK inhibitor (PD 184322)
`
`5) PKC inhibitors (bryostatin, UCN-01, CGP41251) antisense oligonucleotide (ISIS 3521)
`
`6) CCI-779
`
`been selected for furth

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