throbber
Current Treatment Options in Oncology (2014) 15:365–379
`DOI 10.1007/s11864-014-0294-4
`
`Upper Gastrointestinal Cancers (L Rajdev, Section Editor)
`
`Targeting the mTOR Signaling
`Pathway in Neuroendocrine
`Tumors
`
`Jennifer Chan, MD, MPH
`Matthew Kulke, MD, MMSc*
`
`Address
`*Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline
`Avenue, Boston, MA 02215, USA
`Email: matthew_kulke@dfci.harvard.edu
`Email: jang@partners.org
`
`Published online: 5 August 2014
`* The Author(s) 2014. This article is published with open access at Springerlink.com
`
`Keywords Neuroendocrine tumor I Carcinoid I Pancreatic neuroendocrine tumor I mTOR inhibitor I Everolimus
`
`Opinion statement
`
`Neuroendocrine tumors (NETs) are a heterogeneous group of malignancies character-
`ized by variable but most often indolent biologic behavior. Well-differentiated NETs
`can be broadly classified as either carcinoid or pancreatic NET. Although they have sim-
`ilar characteristics on routine histologic evaluation, the 2 tumor subtypes have differ-
`ent biology and respond differently to treatment, with most therapeutic agents
`demonstrating higher response rates in pancreatic NETs compared with carcinoid. Until
`recently, systemic treatment options for patients with advanced NETs were limited.
`However, improvements in our understanding of signaling pathways involved in the
`pathogenesis, growth, and spread of NETs have translated into an expansion of treat-
`ment options. Aberrant signaling through the mechanistic pathway of rapamycin
`(mTOR) pathway has been implicated in neuroendocrine tumorigenesis. Additionally,
`altered expression of mTOR pathway components has been observed in NETs and has
`been associated with clinical outcomes. Targeting the mTOR pathway has emerged as
`an effective treatment strategy in the management of advanced NETs. In a randomized,
`placebo-controlled study of patients with advanced pancreatic NET, treatment with the
`mTOR inhibitor everolimus was associated with improved progression-free survival
`(PFS). Largely based upon these data, everolimus has been approved in the United
`States and Europe for the treatment of patients with advanced pancreatic NET. The ac-
`tivity of everolimus remains under investigation in patients with carcinoid tumors. In a
`randomized study of patients with advanced carcinoid tumors associated with carcinoid
`syndrome, the addition of everolimus to octreotide was associated with improved PFS
`compared with octreotide. However, the results did not meet the prespecified level of
`statistical significance based on central review of radiographic imaging. Results from a
`randomized study examining the efficacy of everolimus in patients with nonfunctional
`gastrointestinal and lung NETs are awaited. In addition, further investigation is needed
`to determine whether primary tumor site or other clinical and molecular factors can im-
`
`Ex. 1076-0001
`
`

`

`366
`
`Upper Gastrointestinal Cancers (L Rajdev, Section Editor)
`
`pact response to mTOR inhibition. Although everolimus can slow tumor progression,
`significant tumor reduction is rarely obtained. Targeting multiple signaling pathways
`is a treatment strategy that may provide better tumor control and overcome resistance
`mechanisms involved with targeting a single pathway. Results of ongoing and future
`studies will provide important information regarding the added benefit of combining
`mTOR inhibitors with other targeted agents, such as VEGF pathway inhibitors, and cy-
`totoxic chemotherapy in the treatment of advanced NETs.
`
`Introduction
`Neuroendocrine tumors (NET) are a rare and het-
`erogeneous group of neoplasms that arise from
`neuroendocrine cells located throughout the body.
`These tumors are characterized by their ability to
`secrete peptides resulting in distinctive hormonal
`syndromes. NETs consist of a spectrum of disease
`ranging from well-differentiated, low-grade tumors
`to poorly differentiated, high-grade carcinomas
`[1•, 2]. In general, poorly differentiated, high-
`grade carcinomas represent aggressive cancers that
`have a different natural history and response to
`treatment compared with well-differentiated, low-
`grade NETs.
`A number of different complex classification sys-
`tems exist for grading NET pathology [1•]. In the
`2010 World Health Organization (WHO) classifica-
`tion, neuroendocrine neoplasms of the digestive sys-
`tem are categorized as low-grade (G1), intermediate-
`grade (G2), and high-grade (G3) based upon mitotic
`count and proliferative index (Ki-67) [3]. High grade
`carcinomas are those with a mitotic count of 920 per
`10 high powered fields (HPF) or a Ki-67 proliferation
`index of 920 %. High grade carcinomas have a more
`aggressive biology and are generally treated with plat-
`inum-based chemotherapy regimens used to treat
`small cell lung cancer. In contrast, well-differentiated,
`low- and intermediate-grade NETs have a more indo-
`lent biology and lower measures of cell proliferation.
`W e l l - d i f f e r e n t i a t e d N E T s c a n b e b r o a d l y
`subclassified as either carcinoid or pancreatic NETs.
`Carcinoid tumors may arise from multiple different
`organs and historically have been classified according
`to site of embryonic origin, namely foregut (gastric,
`bronchial), midgut (small intestine, appendix, proxi-
`mal large bowel), and hindgut (distal colon, rectum,
`genitourinary) [4]. While carcinoid and pancreatic
`NETs may have similar histologic characteristics, these
`
`2 tumor subtypes have different biology and respond
`differently to therapy, with most agents demonstrating
`higher response rates in pancreatic NET patients com-
`pared with carcinoid.
`When NETs are diagnosed at an early stage, surgical
`resection is often curative. Unfortunately, curative sur-
`gery is rarely an option for patients with metastatic dis-
`ease. Recent studies have demonstrated that in
`addition to improving symptoms related to hormone
`hypersecretion, somatostatin analogs slow disease pro-
`gression in patients with small bowel carcinoid tumors
`and gastrointestinal neuroendocrine tumors, including
`pancreatic NET [5, 6]. Treatment approaches with
`targeted therapy, including the use of agents inhibiting
`the vascular endothelial growth factor (VEGF) and
`mTOR signaling pathways and other pathways involved
`in neuroendocrine tumorigenesis, also provide new ther-
`apeutic options for patients with NET [7, 8••].
`Notably, there are a subset of patients with NETs that
`appear histologically well- or moderately differentiated
`but are associated with Ki-67 proliferation indices
`920 % that fall into the high-grade range. The most ap-
`propriate therapy for this subgroup of patients has not
`been well established. A recent series of patients with
`high-grade gastrointestinal neuroendocrine carcinomas
`demonstrated that response rates to platinum-based che-
`motherapy were lower in patients with a Ki-67 G55 %
`[9]. Because sensitivity to platinum-based chemotherapy
`appears to be associated with higher Ki-67 proliferation
`rates, other cytotoxic agents, such as temozolomide, or
`targeted agents, such as mTOR inhibitors or angiogenesis
`inhibitors, may play a role in the management of the
`management of these patients.
`The aim of this review is to provide an overview of
`the role of the mTOR pathway in the pathogenesis of
`neuroendocrine tumors and to review the role of
`mTOR inhibitors in the treatment of this disease.
`
`Ex. 1076-0002
`
`

`

`Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors
`
`Chan and Kulke
`
`367
`
`The mTOR pathway
`
`The mechanistic target (originally referred to as “mammalian target”) of
`rapamycin (mTOR) is an intracellular serine/threonine kinase that regulates
`key cell functions involved in cell survival, proliferation, and metabolism.
`mTOR interacts with several proteins to form 2 multiprotein complexes re-
`ferred to as mTOR complex 1 (mTORC1) and 2 (mTORC2) [10]. By inte-
`grating signals from growth factors and nutrients, mTOR regulates various
`anabolic and catabolic cellular processes [11, 12•, 13].
`mTORC1, which is the better characterized of the 2 complexes, is activat-
`ed by extracellular growth factors and nutrients (Fig. 1). When active,
`mTORC1 phosphorylates the translational regulators eukaryotic initiation
`factor 4E (eIF4E) binding protein 1 (4E-BP1) and S6 kinase 1 (S6K1). These
`events lead to cell proliferation by promoting translation of specific mRNAs
`encoding proteins regulating cell-cycle progression, angiogenesis, energy
`metabolism, and metastasis [14]. mTORC1 also promotes lipid biosynthesis
`and suppresses autophagy through phosphorylation of other key cellular
`effectors [12•].
`Compared with mTORC1, less is known about mTORC2. It also responds
`to growth factor signals, and when active, mTORC2 regulates cell survival,
`cytoskeletal remodeling, and cell migration [15, 16]. It also serves to regulate
`
`Fig. 1. The mTOR signaling pathway. Simplified representation of key components of the mTOR signaling network. The mTOR
`pathway plays an important role in mediating growth factor signals that stimulate cell growth and proliferation and regulate
`angiogenesis and cell metabolism. Arrows represent activation; bars represent inhibition. Adapted from Yao et al, 2013 [66].
`
`Ex. 1076-0003
`
`

`

`368
`
`Upper Gastrointestinal Cancers (L Rajdev, Section Editor)
`
`the PI3K/AKT pathway via phosphorylation and activation of Akt [17].
`Whereas mTORC1 is sensitive to inhibition by rapamycin, mTORC2 is
`considered insensitive to rapamycin [12•].
`In addition to regulation by energy and nutrient status, the mTOR path-
`way responds to growth factors through signaling involving the phos-
`phatidylinositol 3-kinase (PI3K) pathway (Fig. 1) [13]. Binding of insulin or
`insulin-like growth factors to their receptors leads to phosphorylation of
`insulin receptor substrate (IRS). PI3K is subsequently recruited to the cell
`membrane, leading to phosphorylation of phosphatidylinositiol-4,5-
`bisphospate (PIP2) to phosphatidylinositiol-3,4,5-bisphospate (PIP3), and
`ultimately activation of Akt. The phosphatase PTEN is an inhibitory regulator
`of the PI3K-Akt-mTOR pathway that antagonizes the action of PI3K by de-
`phosphorylating PIP3 to PIP2, causing suppression of PI3K-dependent cell
`signaling.
`mTOR is linked to the PI3K-Akt pathway by the tuberous sclerosis pro-
`teins TSC1 and TSC2, which act as a heterodimer that negatively regulates
`mTOR signaling. In response to insulin and other growth factors, TSC2 is
`phosphorylated and inactivated by Akt, which then leads to mTOR activation
`[18–20].
`The mTOR pathway and pathogenesis of NET
`
`Several observations support the importance of the mTOR pathway in the
`pathogenesis of NET. First, although most NETs arise sporadically, NETs can
`arise within the context of several familial cancer syndromes that are due to
`mutations in genes encoding proteins that lie upstream from mTOR. Neu-
`rofibromatosis type 1 (NF-1) and tuberous sclerosis (TS) are autosomal
`dominant tumor susceptibility syndromes caused by inactivating mutations
`in the tumor suppressor genes NF1 and TSC1 and TSC2, respectively [21].
`NF1 encodes the protein neurofibromin, which regulates TSC1 and TSC2
`[22]. Loss of NF1 in neurofibromatosis leads to constitutive activation of
`mTOR and is associated with NETs involving the ampulla of Vater, duode-
`num, and mediastinum. Loss of function of TSC1 and TSC2 leads to mTOR
`activation in patients with tuberous sclerosis, which has been associated with
`pancreatic NETs [23].
`Second, whole exome sequencing analysis of sporadic pancreatic NETs
`has identified somatic mutations in genes involved in the mTOR pathway,
`including PTEN, TS2, and PIK3CA, in 15 % of cases [24•]. Additionally,
`chromosomal changes, including loss of 16p, the region containing TSC2,
`and loss of 10q, which contains PTEN, have been reported in pancreatic NET
`[25, 26].
`Altered expression of mTOR pathway components also has been observed
`in NETs and has been associated with clinical outcomes in several studies. In
`an analysis of gene expression profiles of 72 primary pancreatic NETs, TSC2
`and PTEN were found to be downregulated in most of the primary tumors
`[27•]. In this study, 85 % of primary tumors showed altered protein levels of
`TSC2, PTEN, or both. Low levels of expression of TSC2 and PTEN were as-
`sociated with shorter disease-free and overall survival. Moreover, 8/25
`(32 %) patients with low levels of TSC2 and PTEN developed liver metas-
`
`Ex. 1076-0004
`
`

`

`Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors
`
`Chan and Kulke
`
`369
`
`tases and progression of disease compared with none of 20 patients with
`normal levels of both TSC2 and PTEN. Studies have also demonstrated that
`expression of mTOR and its downstream targets are associated with clinical
`outcome [28, 29•]. In an analysis of tumor from 195 patients with NETs
`arising in various sites, primarily small intestine, expression of mTOR or its
`activated downstream target p-EIF4EBP1 was associated with a higher pro-
`liferative index. Furthermore, high expression of mTOR or its activated
`downstream products were associated with shorter survival [29•].
`Interestingly, there appears to be differential expression of mTOR depend-
`ing on the primary tumor site. Expression levels of mTOR and activation of
`its downstream targets have been found to be higher in foregut tumors
`compared with midgut tumors [28]. Additionally, although low expression
`of PTEN, TSC1, and TSC2 have been found in pancreatic NETs, TSC1 and
`TSC2 expression appear preserved in small intestinal NET [29•]. This suggests
`that there may be potential differences in the mechanisms of mTOR activa-
`tion in different subgroups of NETs.
`
`Treatment
`Targeting the mTOR pathway
`
`& The mTOR inhibitor rapamycin and its analogs bind FK506 binding
`protein, and this complex binds to mTORC1, inhibiting downstream
`signaling [30]. Everolimus and temsirolimus are rapamycin deriva-
`tives that have been evaluated in the treatment NET (Tables 1 and 2).
`
`Everolimus
`Pancreatic NET
`
`& The activity of everolimus in pancreatic NET was explored in the
`RADIANT-1 trial, an international multicenter phase II trial of 160
`
`Table 1. Clinical trials of mTOR inhibitors in carcinoid tumors
`
`Agent
`
`No. patients
`
`Tumor
`response
`rate (%)
`
`Median
`TTP or
`PFS
`
`Reference
`
`Phase II studies
`Everolimus a
`Temsirolimus a
`Phase III studies
`RADIANT-
`Everolimus + octreotide LAR vs.
`2
`Placebo + octreotide LAR
`RADIANT-
`Everolimus vs.
`4
`Placebo
`PFS progression-free survival, TTP time to progression
`a Data from the subset of patients with carcinoid tumors in these phase II studies of unselected patients with NET are presented
`
`Yao et al. 2008 [37]
`Duran et al. 2006 [41]
`
`Pavel et al. 2011[38••]
`
`30
`21
`
`216
`214
`Ongoing
`
`17
`5
`
`2
`2
`
`63 wk
`6.0 mo
`
`16.4 mo
`11.3 mo
`
`Ex. 1076-0005
`
`

`

`370
`
`Upper Gastrointestinal Cancers (L Rajdev, Section Editor)
`
`Table 2. Clinical trials of mTOR inhibitors in Pancreatic NET tumors
`
`Agent
`
`No. patients
`
`Phase II studies
`
`RADIANT-1
`
`Everolimusa
`Everolimus
`Everolimus + octreotide
`
`Temsirolimusa
`
`30
`115
`45
`
`15
`
`Tumor
`response
`rate (%)
`
`27
`9
`4
`
`7
`
`Median
`TTP or
`PFS
`
`50 wk
`9.7 mo
`16.7 mo
`
`Reference
`
`Yao et al. 2008 [37]
`Yao et al. 2010 [31]
`
`10.6 mo
`
`Duran et al. 2006 [41]
`
`Phase III studies
`RADIANT-3
`
`CALGB 80701
`
`207
`203
`Ongoing
`
`Everolimus vs.
`Placebo
`Everolimus + octreotide vs.
`Everolimus + bevacizumab
`+ octreotide
`PFS progression-free survival, TTP time to progression
`a Data from the subset of patients with pancreatic NET in this phase II study of unselected patients with NET are presented
`
`5
`2
`
`11 mo
`4.6 mo
`
`Yao et al. 2011[8••]
`
`patients, 45 of whom also received concurrent treatment with octreotide
`at the discretion of investigators [31]. Upstream regulation of the IGF
`pathway is thought to be a potential resistance mechanism for everolimus
`[32, 33]. Because octreotide has been shown to reduce serum IGF-1 levels
`in patients with advanced solid tumors, the use of everolimus plus a so-
`matostatin analog to target both upstream and downstream components
`of the mTOR pathway has been postulated to potentially have greater
`efficacy than single agent therapy. Among patients receiving octreotide
`plus everolimus, median PFS was longer compared with those receiving
`everolimus alone (17 vs 9.7 months). However, whether the addition
`octreotide to everolimus contributed to higher PFS is uncertain since the
`study was not randomized or designed to make this comparison.
`& Everolimus monotherapy subsequently was compared with best
`supportive care alone in the placebo-controlled RADIANT-3 trial,
`which included 410 patients with advanced pancreatic NET [8••].
`Approximately 40 % of patients also received somatostatin analog
`therapy. Everolimus was associated with a significant prolongation in
`median PFS (11.0 vs 4.6 months, hazard ratio [HR] for progression
`0.35, 95 % confidence interval [CI] 0.27–0.45). Confirmed objective
`partial radiographic responses were observed in 5 % of patients re-
`ceiving everolimus compared with 2 % of those receiving placebo.
`The rate of tumor stabilization was high, 73 % among patients re-
`ceiving everolimus vs 51 % in the placebo group.
`& Drug-related adverse among patients with pancreatic NET receiving
`everolimus included stomatitis, rash, diarrhea, and fatigue [8••]. The
`most common grade 3 or 4 drug-related adverse events were sto-
`
`Ex. 1076-0006
`
`

`

`Carcinoid tumors
`
`Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors
`
`Chan and Kulke
`
`371
`
`matitis (7 %), anemia (6 %), and hyperglycemia (5 %). Though rare,
`everolimus has been associated with serious, adverse events, in-
`cluding pneumonitis.
`& Everolimus causes hyperglycemia, particularly in those with pre-
`existing hyperglycemia. In the RADIANT-3 trial, the frequency of
`severe (grade 3 or 4) hyperglycemia was higher in those with pre-
`existing diabetes mellitus or baseline hyperglycemia (15 % vs 3 % in
`those without diabetes or baseline hyperglycemia) [34]. Because of
`this effect, everolimus may be of particular value in patients with
`hypoglycemia related to insulinoma [35, 36]. In 1 report, 4 patients
`with malignant insulinoma and refractory hypoglycemia experienced
`normalization of glucose levels while receiving everolimus [35]. Two
`of these patients had an objective radiographic antitumor response,
`which may have led to improvements in insulin secretion. Clinical
`improvement in the 2 patients with stable disease suggests a possible
`direct effect of everolimus on insulin production and/or release or an
`effect on peripheral insulin sensitivity.
`
`& Everolimus has been evaluated in combination with octreotide in a
`phase II study of patients with advanced NETs. Partial responses were
`observed in 5 of 30 (17 %) patients with carcinoid tumors, with a
`median PFS of 63 weeks in this group of patients [37].
`& The activity of everolimus was further investigated in patients with
`functional NET in the randomized, placebo-controlled RADIANT-2 trial
`[38••]. In this study, 429 patients with advanced NETs associated with
`carcinoid syndrome and radiographic disease progression in the pre-
`ceding 12 months were randomly assigned to octreotide LAR with either
`everolimus or placebo. Half of patients had a primary small bowel tu-
`mor; lung primary tumors were the second most common tumor type.
`The median PFS as assessed by central radiology review was 16.4 months
`for patients receiving everolimus and octreotide LAR compared with
`11.3 months for patients receiving placebo and octreotide LAR (HR 0.77,
`95 % CI 0.59–1.00; P=0.026). These results did not meet the prespecified
`level of statistical significance. However, based on local investigator ra-
`diology assessment, combined therapy was associated with a median PFS
`duration of 12.0 months compared with 8.6 months with placebo (HR
`0.78, 95 % CI 0.62–0.98; P=0.018). Additionally, imbalances in prog-
`nostic variables favoring the control group, including disease site and
`performance status, could have affected the primary outcome results. A
`subsequent analysis found a significant PFS benefit for everolimus after
`adjusting for randomization imbalances (HR for progression 0.62, 95 %
`CI 0.51–0.87, P=0.003) [39].
`& The best overall radiographic response was a partial response in 2 %
`of both groups; stable disease was the best response in 84 % of the
`patients treated with everolimus and 81 % of patients receiving
`
`Ex. 1076-0007
`
`

`

`372
`
`Upper Gastrointestinal Cancers (L Rajdev, Section Editor)
`
`placebo [38••]. Patients treated with everolimus had a higher rate of
`biochemical responses. Serum chromogranin A levels decreased in 46 %
`of patients receiving everolimus compared with 36 % of patients re-
`ceiving placebo, and 24 hour urinary 5-hydroxyindoleacetic acid (5-
`HIAA) excretion decreased in 61 % of patients receiving everolimus
`compared with 54 % of patients receiving placebo. Data on control of
`symptoms related to carcinoid syndrome were not reported.
`& Additional studies are also needed to determine whether primary tumor
`site of origin may impact response to everolimus. Patients with advanced
`colorectal NET have a particularly poor prognosis. In a post-hoc analysis
`of the RADIANT-2 study, patients with colorectal NETs receiving evero-
`limus plus octreotide LAR had significantly longer PFS (29.9 mo; n=19)
`compared with those receiving placebo plus octreotide LAR (6.6 mo; n=
`20) [40]. Furthermore, some degree of tumor shrinkage was more fre-
`quently noted in patients receiving everolimus plus octreotide LAR
`compared with those receiving placebo plus octreotide LAR (67 % vs
`37 %).
`& The RADIANT-4 trial, a phase III study in which patients with ad-
`vanced, nonfunctional lung or gastrointestinal NETs were random-
`ized to receive everolimus or placebo, recently completed accrual
`(clinical trials.gov, NCT01524783). The results of this study will
`provide important information regarding the activity of everolimus
`in the treatment of nonpancreatic NET.
`
`& The single-agent activity of temsirolimus was evaluated in a multicenter
`phase II study of 37 patients with advanced, progressive neuroendocrine
`tumors [41]. Although the intent-to-treat response rate for the cohort was
`low (6 %), 54 % of patients experienced stable disease while on treatment
`with a median time to progression (TTP) of 6 months. Higher baseline
`tumor levels of phosphorylated mTOR predicted for better outcomes. Fur-
`thermore, temsirolimus appeared more active in patients with pancreatic
`NET compared with carcinoid; median TTP in patients with pancreatic NET
`was 10.6 months compared with 6 months in the carcinoid subgroup.
`However, the small size of this study limits definite conclusions regarding
`the impact of primary tumor site on efficacy of temsirolimus.
`
`Temsirolimus
`
`Future directions with mTOR inhibitor therapy
`
`& Rapamycin and its derivatives are generally cytostatic rather than
`cytotoxic. One of the factors contributing to their limited clinical
`
`Ex. 1076-0008
`
`

`

`Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors
`
`Chan and Kulke
`
`373
`
`success is the existence of multiple feedback loops regulating cell
`survival (Fig. 1). Under normal circumstances, mTORC-1 activation
`of S6K1 promotes degradation of insulin receptor substrate (IRS),
`leading to attenuation of PI3K signaling. Inhibition of mTORC1 can
`lead to increased PI3K signaling by relieving this negative feedback
`[33, 42]. In addition, mTORC1-mediated signaling can inhibit
`mTORC2 through phosphorylation of rictor, one of the components
`of mTORC2. By blocking this feedback loop, rapamycin can con-
`tribute to mTORC2-mediated AKT activation. Furthermore, studies
`have also demonstrated that inhibition of the PI3K/AKT/mTOR
`pathway can result in activation of other receptor tyrosine kinases,
`resulting in downstream signaling promoting cell growth [43].
`& Targeting multiple signaling pathways may provide better tumor
`control and overcome resistance mechanisms. Combining an mTOR
`inhibitor with somatostatin analogs, inhibitors of the VEGF pathway
`and cytotoxic chemotherapy have been evaluated as treatment
`strategies for NETs.
`
`Combining mTOR inhibitor and somatostatin analog
`
`& Because octreotide has been shown to decrease IGF-1 levels and
`PI3K/Akt signaling in vitro, it has been postulated that combining
`an mTOR inhibitor with a somatostatin analog might result in
`enhanced antitumor activity [44]. Everolimus has been evaluated
`in combination with octreotide in several studies, including pa-
`tients with pancreatic NET in stratum 2 of the RADIANT-1 trial
`and patients with carcinoid tumors in the phase III RADIANT-2
`trial. In the RADIANT-1 trial, patients receiving octreotide and
`everolimus had longer PFS compared with patients receiving
`everolimus monotherapy [31]. However, the study was not ran-
`domized or designed to make this comparison. In the RADIANT-2
`trial, although combined therapy with everolimus and octreotide
`was associated with a significantly longer PFS duration compared
`with everolimus and placebo based on local investigator radiology
`review, the improvement in PFS was not statistically significant
`according to central radiology review [38••]. Further investigation
`is needed to determine whether there are specific subsets of pa-
`tients with advanced NETs who benefit most from the addition of
`everolimus to octreotide.
`& Pasireotide is a novel somatostatin analog that binds to a broader
`range of somatostatin receptor subtypes (sst) than octreotide. Com-
`pared with octreotide, pasireotide has a greater binding affinity to
`sst1, sst3, and sst5 and comparable affinity with sst2 [45]. Increased
`receptor binding may lead to additional antiproliferative activity and
`
`Ex. 1076-0009
`
`

`

`374
`
`Upper Gastrointestinal Cancers (L Rajdev, Section Editor)
`
`growth inhibition in NET [44]. A phase I study has established the
`feasibility of combining pasireotide and everolimus [46]. Hypergly-
`cemia was a commonly observed toxicity. A partial radiographic tu-
`mor response was noted in 1/21 patients (5 %), and 17/21 (81 %)
`experienced at least some tumor regression as the best response to
`therapy. The COOPERATE-2 study, a multi-center randomized phase
`II study, recently completed accrual and has examined the combi-
`nation of everolimus alone or in combination with pasireotide LAR
`in patients with advanced, progressive pancreatic NET (clinical
`trials.gov, NCT01374451). The results of this study will provide ad-
`ditional information regarding the added efficacy of combining an
`mTOR inhibitor with a somatostatin analog.
`
`Combining mTOR inhibitor and VEGF pathway inhibitor
`
`& A key role for angiogenesis and VEGF pathway signaling in NET is sug-
`gested by clinical observations that neuroendocrine tumors are vascular
`tumors. Expression of VEGF has been demonstrated in carcinoid and
`pancreatic NETs [47, 48]. Increased expression of VEGF receptor-2
`(VEGFR-2) has been demonstrated in tissue from gastrointestinal carci-
`noid tumors and a carcinoid cell line [49, 50]. Additionally, pancreatic
`neuroendocrine tumors also show widespread expression of VEGFR-2
`and -3 in addition to platelet-derived growth factor receptors (PDGFRs) α
`and β, stem-cell factor receptor (c-kit) [51–53].
`& The tyrosine kinase inhibitor sunitinib has shown activity against a
`range of signaling pathways and growth factors/receptors, including
`VEGFR-1, -2 and -3, PDGFR-α and -β, KIT, RET, FMS-like tyrosine
`kinase-3 (FLT3), and colony-stimulating factor receptor (CSF-1R). In
`a randomized phase III study examining the activity of sunitinib in
`patients with progressive pancreatic NET, sunitinib was associated
`with a median progression-free survival (PFS) of 11.4 months, as
`compared with 5.5 months for placebo (PG.001) [7]. Two other
`small molecule tyrosine kinase inhibitors (TKIs), sorafenib, and
`pazopanib, have also been evaluated in phase II studies [54, 55].
`Although response rates to TKIs in carcinoid tumors have been low,
`all studies report a high rate of disease stabilization and potentially
`encouraging progression-free survival durations.
`& Bevacizumab, a monoclonal antibody against VEGF, has been eval-
`uated in a randomized phase II study of patients with advanced or
`metastatic carcinoid tumors on a stable dose of octreotide. Patients
`were randomly assigned to receive 18 weeks of bevacizumab or
`pegylated IFN-α 2b [56]. During the first 18 weeks of therapy, 18 %
`of the bevacizumab-treated patients experienced radiographic partial
`responses, and 77 % had stable disease. Furthermore, after 18 weeks,
`
`Ex. 1076-0010
`
`

`

`Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors
`
`Chan and Kulke
`
`375
`
`95 % of patients treated with octreotide plus bevacizumab remained
`progression-free compared with only 68 % of those receiving
`octreotide plus IFN-α.
`& A number of recently completed and ongoing studies have evaluated
`the combination of an mTOR inhibitor with inhibitors of the VEGF
`pathway. Combining everolimus with tyrosine kinase inhibitors of
`VEGFR and other growth factor receptors may be limited by toxicity.
`In a phase I study of everolimus in combination with sorafenib,
`dose-limiting toxicity precluding escalation to full doses of each
`agent was observed [57]. However, the combination of the everoli-
`mus and bevacizumab was shown to be well tolerated and associated
`with antitumor activity (overall response rate 26 %) in an initial
`phase II study enrolling patients with low or intermediate grade
`neuroendocrine tumors [58]. Furthermore, encouraging early results
`have been noted in a phase II trial of temsirolimus plus bevacizumab
`in 55 patients with progressive NET. In a preliminary report, a con-
`firmed partial response was documented in 20 patients (37 %), and
`44 (80 %) remained progression-free at 6 months [59]. Results of
`CALGB 80701, a phase II trial of patients with advanced pancreatic
`NETs randomized to receive either everolimus and octreotide or
`everolimus plus bevacizumab and octreotide, will provide additional
`information about the benefits of combined mTOR and VEGF
`pathway inhibition (clinical trials.gov, NCT01229943).
`
`Combining mTOR inhibitor and cytotoxic chemotherapy
`
`& Cytotoxic chemotherapy has minimal activity in patients with ad-
`vanced carcinoid tumors. In contrast, pancreatic NETs may respond
`well to treatment with streptozocin and other alkylating agents [60,
`61]. Recent prospective and retrospective studies have suggested that
`temozolomide-based regimens may be comparable in efficacy and
`more tolerable than streptozocin-based regimens. In retrospective
`series, temozolomide-based therapy has been associated with overall
`response rates of 8 %–70 % in patients with advanced pancreatic
`NET [62–64].
`& The combination of temozolomide and everolimus has been evalu-
`ated in a phase I/II study of patients with advanced pancreatic NET
`[65]. Treatment was associated with known side effects of each drug
`without evidence of synergistic toxicity. Encouraging evidence of
`antitumor activity with this combination was observed. Among 40
`evaluable patients, 16 (40 %) experienced a partial response. The
`median PFS duration was 15.4 months, which is superior to the re-
`ported PFS observed with everolimus alone in the randomized,
`placebo-controlled RADIANT-3 study. However, these results need to
`
`Ex. 1076-0011
`
`

`

`376
`
`Upper Gastrointestinal Cancers (L Rajdev, Section Editor)
`
`Conclusions
`
`be interpreted with caution since this was a single-arm study. Fur-
`thermore, disease progression prior to study enrollment was not a re-
`quirement in this study, as it was in the RADIANT-3 study. Future studies
`evaluating the relative efficacy of combining chemotherapy with an mTOR
`inhibitor compared with treatment with either agent alone are warranted.
`
`Recent improvements in our understanding of the molecular biology of NETs
`have led to an expansion of treatment options for patients with advanced
`disease. Studies indicate that the mTOR pathway plays an important role in
`the pathogenesis of NET. The mTOR inhibitor everolimus has been shown to
`significantly delay disease progression in patients with pancreatic NET. Fur-
`ther studies evaluating everolimus in advanced carcinoid are anticipated.
`Results of ongoing and future studies will provide important information
`regarding the added benefit of combining an mTOR inhibitor with cytotoxic
`chemotherapy and other targeted agents, such as VEGF pathway inhibitors,
`in the treatment of advanced NETs.
`
`Compliance with Ethics Guidelines
`
`Conflict of Interest
`Jennifer Chan reports grants from Novartis, Merck, and Bayer. Matthew Kulke reports grants from Novartis and
`has served as a consultant to Pfizer, Ipsen, Lexicon, and Novartis.
`
`Human and Animal Rights and Informed Consent
`This article does not contain any studies with human or animal subjects performed by any of the authors.
`
`Open Access This article is distributed under the

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