Current status of novel agents in advanced gastroesophageal adenocarcinoma
Review Article

Current status of novel agents in advanced gastroesophageal adenocarcinoma

Nishi Kothari, Khaldoun Almhanna

Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA

Correspondence to: Khaldoun Almhanna, MD, MPH. Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA. Email: khaldoun.almhanna@moffitt.org.

Abstract: Gastroesophageal (GE) adenocarcinomas are highly lethal malignancies and despite multiple chemotherapy options, 5-year survival rates remain dismal. Chemotherapy is the mainstay of treatment but patients are often limited by toxicity and poor performance status. Because of molecular heterogeneity, it is essential to classify tumors based on the underlying oncogenic pathways and develop targeted therapies that act on individual tumors. Trastuzumab, a human epidermal growth factor receptor type 2 (HER2) monoclonal antibody, was the first such agent shown to improve response rate, progression free survival (PFS), and overall survival (OS) when added to cisplatin based chemotherapy in patients with HER2 over-expressing GE junction (GEJ) and gastric adenocarcinomas. However, HER2 over expressing GE tumors are in the minority and the need for additional targeted agents is urgent. Though many agents are in development, incorporating targeted therapy in the treatment of GE cancers comes with a unique set of challenges. In this review, we outline oncogenic pathways relevant to GE adenocarcinomas, including HER2, epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), and c-Met, and discuss recent trials with agents targeting these pathways.

Keywords: Gastric and esophageal adenocarcinoma; targeted therapy


Submitted Nov 07, 2014. Accepted for publication Nov 11, 2014.

doi: 10.3978/j.issn.2078-6891.2014.098


Introduction

Gastroesophageal (GE) adenocarcinomas are commonly diagnosed at an advanced stage and are extremely lethal, with median survival of less than 1 year for metastatic disease (1,2). Over the last 50 years, survival has improved only modestly despite considerable improvements in diagnosis, surgical techniques, and multidisciplinary approaches to care.

Chemotherapy remains the cornerstone of treatment for GE patients with locally advanced and metastatic disease. Many chemotherapy agents have activity including platinums, irinotecan, fluorouracil, taxanes and anthracyclines. Treatment with a combination of three agents has been shown to lead to modest improvements in survival compared to two agents, but at the expense of significant toxicity (3).

The pathogenesis of GE cancers involves multiple genetic and epigenetic alterations, chromosomal aberrations, gene mutations, and altered molecular pathways. During recent years, the molecular heterogeneity underlying carcinogenesis and metastasis has begun to be elucidated. Some of these molecular abnormalities and signaling pathways are amenable to pharmacological interventions (Figure 1). Targeted therapies been evaluated in the pre-clinical setting and are now rapidly moving to clinical trials (Table 1). The vascular endothelial growth factor (VEGF) receptor, epidermal growth factor receptor (EGFR), human epidermal growth factor receptor type 2 (HER2), insulin-like growth factor receptor (IGF-R), phosphatadylinositol 3-kinase (PI3k)/protein kinase B (Akt)/mammalian target of rapamyin (mTor) pathway, c-Met, fibroblast growth factor receptor (FGFR), poly [adenosine diphosphate (ADP)]-ribose polymerase (PARP) inhibitors, and immunotherapies have been investigated as therapeutics. We will discuss molecular targets and the novel drugs currently approved and in development for patients with GE.

Figure 1 Targeted therapy in gastric cancer and sites of action. VEGFR, vascular endothelial growth factor receptor; EGFR, epidermal growth factor receptor; PI3K, phosphatadylinositol 3-kinase; PTEN, phosphatase and tensin homolog; PDK-1, phosphoinositide-dependent kinase 1; Akt, protein kinase B; mTOR, mammalian target of rapamyin; HGF, hepatocyte growth factor; MEK, mitogen-activated protein/extracellular signal-regulated kinase kinase; ERK, extracellular-regulated kinase.
Table 1
Table 1 Targeted agents and clinical trials for gastric and gastroesophageal cancers
Full table

HER2 inhibition

The HER2 receptor is a member of the EGFR/HER family involved with signal transduction, leading to cell growth and differentiation. The HER2 gene is a proto-oncogene, located at the long arm of human chromosome 17 (4), which encodes for a 185 KD transmembrane glycoprotein receptor with intracellular tyrosine kinase activity (5).

HER2 over-expression and amplification in GE ranges from 7-34% of patients, depending on the population studied. The primary tumor site appears to have higher concordance of HER2 amplification by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) than regional lymph node or distant metastases (6-8). By consensus, HER2 is considered to be negative if IHC is 0 or 1+. HER2 is positive if IHC 3+. IHC of 2+ is considered equivocal and merits confirmatory testing with FISH (9).

Preclinical studies have shown that anti-HER2 therapies have significant activity for both in vitro and in vivo gastric cancer models (10,11). The most common approaches to targeting HER2 are through inhibition by monoclonal antibodies (trastuzumab and pertuzumab) or tyrosine kinase inhibitors (TKIs) (lapatinib). Both types of blockade have been examined in clinical trials of patients with GE cancers.

Trastuzumab, pertuzumab, and trastuzumab emtansine (TDM-1)

Trastuzumab is a humanized monoclonal antibody that has been approved by the US Food and Drug Administration (FDA) since 1998 for the treatment of breast cancer. Trastuzumab targets the extracellular binding domain of the HER2 receptor and has been combined with cytotoxic chemotherapy in patients with gastric and GE junction (GEJ) tumors in several trials. The trastuzumab for gastric cancer (ToGA) study was an internatinoal, open-label phase III trial that randomized patients with treatment naive metastatic or locally advanced unresectable gastric or GEJ adenocarcinoma with over-expressed HER2 to chemotherapy with trastuzumab versus chemotherapy alone. HER2 overexpression was defined as staining 3+ by IHC or by FISH positivity (12).

Patients received cisplatin plus fluoropyrimidine every 3 weeks for six cycles, with or without intravenous trastuzumab at 6 mg/kg after a one time loading dose of 8 mg/kg.

A 2.7-month improvement in median overall survival (OS) for patients who received trastuzumab was demonstrated (median OS 13.8 months compared with 11.1 months). Response rate, time to progression, and duration of response were significantly higher in the trastuzumab plus chemotherapy group as well. Of note, the median survival in the chemotherapy only arm was higher than expected in this study, potentially related to the high proportion of Asian patients in the study (55%). The combination was generally well tolerated with only a slightly increased risk of asymptomatic left ventricular dysfunction and transfusion reaction. This study led to the first FDA approval for targeted therapy for gastric and GEJ adenocarcinoma in 2010 (13).

Based on these encouraging results, several other studies with trastuzumab are being conducted. The HELOISE trial (a study of herceptin in combination with cisplatin/capecitabine chemotherapy in patients with HER2-positive metastatic gastric or GEJ cancer) is currently recruiting patients to evaluate the optimal dose of trastuzumab in advanced gastric and GEJ tumors (14). In the non-metastatic setting, NCT01130337 is a phase II study which treats patients with trastuzumab, capecitabine, and oxaliplatin for three cycles prior to surgery. If an R0 or R1 resection is achieved, patients receive an additional three cycles of treatment. Trastuzumab will be continued for a total of 1-year (15). Similarly, the TOXAG study (a study of the combination of oxaliplatin, capecitabine, and herceptin and chemoradiotherapy in the adjuvant setting in operated patients with HER2+ gastric or GEJ cancer) is ongoing (16). The HER-FLOT study (Herceptin in combination with FLOT as perioperative treatment for patients with HER2-positive locally advanced esophagogastric adenocarcinoma) gives trastuzumab with FLOT (5FU, leucovorin, docetaxol, and oxaliplatin) for four cycles prior to surgical resection. Patients then receive an additional four cycles of chemotherapy with trastuzumab and nine additional cycles of trastuzumab alone (17). For locally advanced esophageal or GEJ adenocarcinoma, RTOG 1010 is a phase III trial which randomizes patients to weekly paclitaxel, carboplatin, and radiation with or without trastuzumab prior to surgery (18). The results of these studies could change the treatment paradigm for HER2 overexpressing GE cancers.

As resistance to HER2 therapy has begun to arise, there has been interest in the second generation HER2 targeting agent pertuzumab, which binds to a distinct site on the HER2 (and potentially HER3) receptor and leads to the disruption of dimerization and blockade of downstream signaling. Based on pre-clinical work in GEJ, as well as the efficacy of the combination of trastuzumab and pertuzumab in breast cancer (19), the JACOB phase III study (a study of perjeta in combination with herceptin and chemotherapy in patients with HER2-positive metastatic GEJ or gastric cancer) randomizes patients with metastatic or locally advanced unresectable disease to first line cisplatin, fluoropyrimidine, and trastuzumab with or without pertuzumab (20).

TDM-1 is an antibody-drug conjugate which utilizes HER2 overexpression to deliver a cytotoxic agent directly to cancer cells is being evaluated in GEJ patients expressing HER2; a second line phase II/III trial of TDM-1 in advanced gastric cancer is currently recruiting; the study has three arms; TDM-1 at 3.6 mg/kg every 3 weeks, TDM-1 at 2.4 mg/kg every week, or physician’s choice of single agent pa clitaxel or docetaxel (14).

Lapatinib

Lapatinib is an oral small molecule dual TKI of EGFR and HER2. It has been approved for the treatment of HER2-positive advanced breast cancer previously treated with trastuzumab and in conjunction with hormonal therapy for triple positive metastatic breast cancer (21-23).

Lapatinib has been evaluated in combination with standard chemotherapy in patients with gastric and GEJ adenocarcinomas. In the phase III LOGIC study (lapatinib optimization study in HER2-positive gastric cancer), patients with HER2 over-expressed advanced gastric and GEJ adenocarcinomas were randomized to chemotherapy (capecitabine and oxaliplatin) plus lapatinib versus placebo (24). This study did not meet its primary endpoint of improvement in OS, though certain subgroups (the Asian population and patients under age 60 years) were shown to have a benefit.

The second line phase III TyTAN trial (a phase III Asian study of tykerb in combination with paclitaxel as second-line therapy in gastric cancer) compared weekly paclitaxel with or without lapatanib in second line patients with HER2-positive advanced disease. Again, there was no OS or progression free survival (PFS) benefit for the lapatinib group, though there was a statistically significant increased response rate (25). At present, lapatinib is not ready for widespread implementation in GEJ but ongoing studies might better define its role in combination with other targeted agents.

Of the monoclonal antibodies, only trastuzumab is approved for locally advanced unresectable and metastatic GEJ and gastric cancers. However, with the results of adjuvant trastuzumab trials as well as the pertuzumab and TDM-1 studies, the role for monoclonal antibodies in GE cancers will likely expand significantly.


EGFR inhibition

The EGFR is a trans-membrane glycoprotein receptor for the EGF family of extracellular protein ligands (26) and is overexpressed in several gastrointestinal (GI) malignancies. Ligand binding to the extracellular domain leads to EGFR activation and phosphorylation of the intracellular tyrosine kinase, which then directs activation of Ras/Raf/mitogen activated protein kinase (MAPK) or the Akt/mTOR pathway (27). EGFR overexpression occurs in 30-50% of GE. It is associated with older age, more aggressive histology, and advanced stage (28-30).

The most common approaches to inhibit the EGFR are by inhibition of the EGFR via monoclonal antibodies (i.e., cetuximab and panitumumab) or TKIs (i.e., gefitinib, erlotinib). Both methods have been studied in patients with GE.

Cetuximab

Cetuximab is an immunoglobulin G 1 (IgG1) type chimeric monoclonal antibody that binds to the extracellular domain of the human EGFR and competitively inhibits the binding of EGF and other ligands, as well as ligand-induced tyrosine kinase auto-phosphorylation. This antibody-receptor interaction prevents receptor dimerization and thereby blocks ligand-induced EGFR tyrosine kinase activation. Cetuximab also induces EGFR internalization, down-regulation, and degradation (31). It is currently approved for the treatment of advanced KRAS wild type colorectal cancer as well as squamous cell head and neck cancers (32,33).

Based on promising phase II data, the phase III trial EXPAND (erbitux in combination with xeloda and cisplatin in advanced GE) randomized 904 patients to cisplatin with capecitabine with or without cetuximab. However, no PFS or OS benefit for the cetuximab group was found (34). RTOG 0436 was a phase III trial which randomized patients with locally advanced esophageal cancer to weekly concurrent cisplatin (50 mg/m2), paclitaxel (25 mg/m2) for 6 weeks and daily radiation 50.4 Gy/1.8 Gy fractions ± weekly cetuximab (400 mg/m2 day 1 then weekly 250 mg/m2) for 6 weeks (35). No OS benefit to cetuximab was found.

Unlike in colorectal cancer, KRAS mutations have not been shown to be a negative predictive biomarker for response to cetuximab in GE (36). Though other biomarkers including EGFR expression, copy number, and phosporylation have been evaluated, the sample sizes and retrospective nature of these analyses have precluded meaningful conclusions (37-40).

Panitumumab

Panitumumab is the first fully human IgG2 monoclonal antibody targeting EGFR. In gastric cancer, the REAL-3 study [a randomised open-labelled multicentre trial of the efficacy of epirubicin, oxaliplatin, and capecitabine (EOX) with or without panitumumab in previously untreated advanced oesophago-gastric cancer] did not show any benefit at preplanned interim analysis and was stopped early (41). However, these negative results may have been partly due to decreased doses of chemotherapy in the combination arm (42). In the single arm phase II ACOSOG Z4051 trial, patients with potentially resectable disease were given neoadjuvant docetaxel, cisplatin, and panitumumab as well as radiation (43). Some disease activity was found but at the expense of significant toxicity.

Gefitinib

Gefitinib is an oral EGFR TKI with promising activity against several types of malignancy in early phase trials. Based on phase II data (44), a phase III trial (NCT01243398) randomized patients with advanced GE to gefitinib versus placebo after progression on chemotherapy. The study is complete and the pending results will help better delineate the activity of gefitinib in GE (45).

Erlotinib

Erlotinib is another oral EGFR TKI, which has been approved in the US for the treatment of lung and pancreatic cancer. In a phase II analysis, erlotinib was found to be active in patients with GEJ cancer with a response rate of 9%, but with no responses in gastric cancer (46).


Vascular endothelial growth factor receptor (VEGFR) inhibition

Angiogenesis is an important aspect of tumorigenesis and is critical for tumor growth and survival. The VEGF plays a pivotal role in the control of angiogenesis, tumor growth, and metastasis in many human cancers (47) including GE, which makes it an attractive target for treatment. VEGF-A is an essential mediator of physiologic and pathologic angiogenesis (48), and its activities are mediated by two tyrosine kinase receptors, VEGFR-1 and VEGFR-2. Serum VEGF concentration has been related to metastasis and worse outcome in GE (49,50). Multiple agents have been developed to target the VEGF pathway, including monoclonal antibodies and TKIs.

Bevacizumab

Bevacizumab is a recombinant humanized IgG1 monoclonal antibody against VEGF, which has been shown to have efficacy in colorectal, lung, ovarian, and renal cell cancers (13,51-54). Side effects including thromboembolic events, gastrointestinal perforation, and hypertension have been demonstrated.

Promising phase II trial results in GE cancers led to AVAGAST (avastin in gastric cancer), a phase III multinational, randomized, placebo-controlled trial to evaluate the efficacy of adding bevacizumab to cisplatin based chemotherapy in the first-line treatment of advanced gastric cancer (55). Seven hundred and seventy-four patients from 17 countries were enrolled. Approximately 50% of patients were from Asia. Median OS was 12.1 months in the bevacizumab plus chemotherapy arm compared to 10.1 months with placebo plus chemotherapy arm [hazard ratio (HR) 0.87; 95% confidence interval (CI), 0.73 to 1.03; P=0.1002]. Though the trial did not meet its primary objective of OS, both median PFS and overall response rate (ORR) were significantly improved in the bevacizumab group. No bevacizumab-related safety signals were identified. The genetic heterogeneity of gastric cancer might explain the discordant results between the phase II and III trials. In addition, the patients with GEJ tumors on the AVAGAST study treated with bevacizumab arm had an exceptionally high response rate of 85% and improved OS. Asian patients showed better OS and PFS regardless of the treatment received when compared to European and Americans. Selection bias, sample size, and study design might have limited the conclusions of single-arm phase II studies.

In order to better select patients who might benefit from anti-VEGF therapy, a panel of tumor angiogenic factors was evaluated in the AVAGAST study, including EGFR, VEGF-A, VEGFR-1, VEGFR-2 and neuropilin (56). Low tumor neuropilin expression was associated with shorter OS in the placebo group. Adding bevacizumab seemed to correct this effect as patients with low tumor neuropilin had an OS treatment HR numerically better than those with high neuropilin in the bevacizumab group. Neuropilin thus appeared to be a promising prognostic biomarker candidate, with potential predictive properties for bevacizumab as well. In addition, lower baseline plasma VEGF-A correlated with longer OS. Further evaluation of these biomarkers is ongoing.

Bevacizumab is being evaluated in the neoadjuvant setting in the United Kingdom. The MAGIC-B study (medical research council adjuvant gastric infusional chemotherapy) is assessing the role of bevacizumab for peri-operative chemotherapy in operable adenocarcinoma of the stomach and GEJ.

Ramucirumab

Ramucirumab is a fully human IgG1 monoclonal antibody that specifically and potently inhibits VEGFR-2. Ramucirumab has demonstrated efficacy and tolerability in several studies. The phase III REGARD study (ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma) randomized second line gastric or GEJ adenocarcinoma patients to single agent ramucirumab or best supportive care (BSC). They found a median OS of 5.2 months in the treatment arm compared to 3.8 months, with a P value of 0.042 (57). Based on this study, the FDA approved ramucirumab in 2014 for use as a single agent in gastric and GEJ cancer after progression on a platinum or fluropyrimidine containing regimen (58). This is the first approval of a biologic agent in an unselected GEJ population. Biomarker studies to better delineate the population most likely to benefit are ongoing.

The phase III RAINBOW study (a global, phase III, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic GEJ and gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy rainbow) randomized 665 second line advanced gastric or GEJ adenocarcinoma patients to paclitaxel with or without ramucirumab. Median OS was 9.6 months in the combination arm versus 7.4 months for paclitaxel alone. Patients in the combination arm had more neutropenia and hypertension (59). These findings will likely lead to approval of ramucirumab in combination with paclitaxel by the FDA later this year. However, a front line phase II study of ramucirumab with or without FOLFOX did not show an improvement in the primary endpoint of PFS (60). The results of the major trials involving bevacizumab and ramucirumab are described in Table 2.

Table 2
Table 2 Phase II/III trials targeting VEGFR in gastroesophageal cancerse
Full table

Another approach to targeting the VEGF pathway is through so-called dirty kinase inhibitors, which inhibit the VEGF receptor as well as FLT-3, c-kit, and RET. Several TKIs are currently being evaluated and are described below.

Sunitinib

Sunitinib is an oral multi-targeted TKI of VEGFR, platelet-derived growth factor receptors (PDGFRs), c-kit, RET, and FLT3 that has been approved for the treatment of advanced renal cell carcinoma and imatinib resistant or intolerant gastrointestinal stromal tumors.

Several trials have evaluated single agent sunitinib in the treatment of GEJ. In a phase II second line trial of single agent sunitinib in 78 patients with advanced gastric and GEJ cancer, two patients had partial response and 25 patients had stable disease for ≥6 weeks. Median PFS was 2.3 months and median OS was 6.8 months (95% CI, 4.4-9.6 months) (62). Sunitinib has also been evaluated in combination with chemotherapy. A second line phase II trial randomized 107 patients to docetaxel with or without sunitinib. The time to progression was not significantly different (3.9 months in the sunitinib arm versus 2.6 months), but there was an increased response rate of 41.4% compared to 14.3% (63).

Similar to other TKIs, sunitinib has multiple drug interactions and can lead to QTc prolongation and changes in the metabolism of CYP3A4 substrates. Common toxicities include hypertension, hand-foot syndrome, and liver dysfunction.

Sorafenib

Sorafenib is a potent inhibitor of the Raf tyrosine kinase and several other receptor tyrosine kinases, including VEGFR-2, VEGFR-3, and PDGFR-β. Sorafenib has been approved for the treatment of both renal cell carcinoma and hepatocellular carcinoma based on the results of phase III trials (64,65). In tumor xenograft models, sorafenib effectively inhibited tumor growth and angiogenesis in gastric tumors (66).

Sorafenib has been evaluated for the treatment of advanced GEJ in several studies, both in combination with chemotherapy and as a single agent. Though one phase II study of 44 second line gastric cancer patients which combined sorafenib with docetaxel and cisplatin showed an impressive median PFS of 5.8 months and median OS of 13.6 months (67), other studies have not found these results and have been terminated early because of low response rates (68,69).

Pazopanib

Pazopanib is an oral agent which inhibits angiogenesis through multiple pathways, including the VEGFR, the PDGFR, as well as c-kit. It has been approved by the FDA for use in the treatment of metastatic renal cell carcinoma as well as metastatic soft tissue sarcoma based on the results of phase III trials (70,71). Pazopanib has also been shown to have activity in metastatic thyroid cancer (72).

Pazopanib is currently being evaluated with chemotherapy in two GEJ trials. The phase II PaFLO trial (FLO ± pazopanib as first-line treatment in advanced gastric cancer) randomized first line advanced gastric cancer patients to 5-fluorouracil, leucovorin, and oxaliplatin with or without pazopanib and is currently accruing patients (73). Another first line phase II trial adds pazopanib to capecitabine and oxaliplatin in advanced gastric cancer patients and is also recruiting (74). The results of these studies will help determine if pazopanib has a role in the treatment of advanced GE cancer.


IGF-1 inhibition

The IGF-1 receptor belongs to the insulin receptor family. IGF-1R is expressed on the cell surface and phosphorylation of intracellular substrates leads to activation of the MAPK and PI3K/Akt pathways which promotes tumor growth, progression and invasion in several cancers, including GE (75).

In GE, IGF-1R expression in resected tumors correlates with poorer clinical outcomes (76). IGF-1R signaling has been associated with resistance to cytotoxic therapy and inhibition of IGF-1R enhances tumor cell apoptosis in numerous models (77). The IGF-1R pathway can be targeted through monoclonal antibodies, IGF-1R antisense/small interfering ribonucleic acid (siRNA), and receptor tyrosine kinases.

In a study of 86 patients with resected gastric tumors, patients with low expression of both IGF-1R and EGFR had significantly longer OS compared to those who lack the low co-expression (76). A phase I trial of docetaxel combined with CP-751,871, an IGF-1R antibody, has demonstrated promising results and warrants further investigation (78).


Fibroblast growth factor (FGF) TKIs

FGF and its signaling receptors have multiple biological properties including cell proliferation, differentiation, motility, and transformation (79,80). FGFR2 is amplified in poorly differentiated gastric cancer (scirrhous cancer) with malignant phenotypes (81), which makes it a potential molecular target for treatment.

In preclinical models, AZD2171, a highly potent oral VEGF, FGFR1, PDGFRB, and VEGFR-2 TKIs, led to tumor inhibition in gastric cancer xenografts in a dose-dependent fashion. The most potent antitumor activity was seen in xenografts over-expressing FGFR2. These results suggest that AZD2171 might be clinically beneficial in patients with FGFR2 expressing gastric tumors (82).

Ki23057, a broad-range TKI of FGFR2, also inhibits FGFR1, FGFR2, and VEGF2 tyrosine kinases. It inhibits the proliferation of gastric scirrhous cancer cells with FGFR2 gene amplification only. Oral administration of Ki23057 inhibits the growth and peritoneal dissemination of gastric cancer cells through FGFR2-RAS/extracellular-regulated kinase (ERK) inhibition, rather than through FGFR2-PI3k-AKT signaling inhibition (83). To our knowledge, no clinical trials are currently available for this compound in GE.


c-Met TKIs

C-Met is a receptor tyrosine kinase that is expressed in epithelial and endothelial cells. Overexpression of c-Met and activating c-Met mutations have been widely documented in many tumor types including GE and have been correlated with poorer outcomes (84,85). Hepatocyte growth factor (HGF), its ligand, is expressed by cells of the mesenchymal lineage.

A phase II study examined the safety and efficacy of two dosing schedules of foretonib (GSK1363089), an oral small-molecule inhibitor of c-Met and VEGFR-2, as a single agent in patients with metastatic gastric adenocarcinoma. Foretonib was well tolerated but demonstrated minimal antitumor activity in a c-Met unselected population (86).

A phase II study of rilotumumab (a human monoclonal antibody directed against HGF) showed more efficacy in a subset of patients with increased MET expression by IHC (87). Based on this data, the phase III RILOMET-1 trial [an international phase III multicenter, randomized, double-blind, placebo-controlled trial of rilotumumab plus epirubicin, cisplatin, and capecitabine (ECX) as first-line therapy in patients with advanced MET-positive gastric or GEJ adenocarcinoma] is currently recruiting (88). However, a double blind randomized first line phase II study of this combination (ECX with or without rilotumumab) was recently found to be negative for improved PFS (89).

Onartuzumab (a humanized monovalent antibody directed against MET) is also being evaluated in a first line, randomized phase III trial in MET-positive, HER2-negative GE patients in combination with FOLFOX. This study is ongoing and should be complete in 2015 (90).


PI3 kinase pathway inhibition

The PI3K enzymes are involved in the phosphorylation of membrane inositol lipids (91). The activation of PI3K generates the second messenger phosphatidylinositol 3,4,5-trisphosphate (PIP3) from phosphatidylinositol 4,5-bisphosphate (PIP2). This recruits proteins to the cell membrane, including the Akt/PKB kinases, resulting in their phosphorylation by phosphoinositide-dependent kinase 1 (PDK1) and by PDK2 (92,93).

Dysregulation of the PIP3/Akt/mTOR pathway can occur secondary to oncogenic mutations of PIK3CA (94), loss of phosphatase and tensin homolog (PTEN) function (95,96), mutation of Akt/PKB isoforms (97), or upstream activation through other pathways like IGF-1R. Abnormal expression of the PTEN protein in gastric cancer is found in 11% of tumors and is related to the tumor differentiation, advanced staging, and chemoresistance (98). Upregulation of the PI3k/Akt/mTOR downstream pathway correlates with a worse prognosis and may contribute to the resistance to chemotherapy (99).

Everolimus is an oral mTOR inhibitor that has shown anticancer activity both in phase I and II studies (100,101). The phase III GRANITE-1 trial (safety and efficacy of everolimus monotherapy plus BSC in patients with advanced gastric cancer) was performed for further evaluation. Six hundred and fifty-six second or third line advanced gastric cancer patients were randomized to everolimus as monotherapy or placebo with BSC. The median OS was not significantly different, at 5.39 months in the everolimus group compared to 4.34 months in the placebo group (102).


PARP Inhibitors

The function of PARP is to repair single stranded breaks (SSBs). If these SSBs are not repaired, they become double stranded breaks (DSBs) at the next fork replication, which leads to cell death. As cancer therapeutics, the PARP inhibitors prevent the cancer cell’s SSB repair mechanism and ultimately allow tumor cell death to occur (103). These agents have shown activity in ovarian and breast cancer, particularly in patients with BRCA1 or BRCA2 gene mutation.

The PARP inhibitor olaparib was studied in a second line phase II trial for metastatic or recurrent GE. Patients received paclitaxel with or without olaparib (104). Though PFS was not significantly different, OS was improved in the olaparib group. Because preclinical data had shown more olaparib sensitivity in patients with low ataxia telangiectasia mutated (ATM) protein (105), this study performed a subset analysis in which low ATM patients were found to have improved OS with olaparib. Based on these results, an ongoing phase III study of second line GE randomizes patients to paclitaxel with or without olaparib (106). A phase I study of another PARP inhibitor veliparib with FOLFIRI is also currently recruiting (107).


Immunotherapy

Cancer evades host immune recognition through multiple mechanisms acquired during tumor evolution (108). By blocking negative immune regulatory pathways and thereby allowing increased immune activity, cancer immunotherapy is a novel way to attack tumor cells. With the approval of therapies like ipilumamb for melanoma, there has been increased interest in immunotherapy for other diseases. Ipilumamb releases negative immune regulatory pathway by blocking cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), an inhibitory receptor.

The immunotherapy agent nivolumab has also been recently evaluated in cancer. This drug function by blocking binding of receptor inhibitor programmed cell death 1 (PD-1) that is expressed on T-cells to programmed cell death ligand 1 (PDL-1) which prevents T cell death. A phase I trial of nivolumab included patients with gastric adenocarcinoma. Unfortunately, these patients were not included in the efficacy analysis (109).

Pembrolizumab is another agent that blocks the binding of PD-1 and PDL-1 (as well as PDL-2). A phase IB study of pembrolizumab in recurrent and metastatic gastric and GEJ adenocarcinoma patients with PD-L1 tumor positivity by IHC was presented at ESMO 2014 (110). Tolerability as well as anti-tumor activity was demonstrated. Another anti-PDL-1 agent, MEDI4736, has shown activity in gastric cancer (111).

The combination of CTLA-4 and PDL-1 blocking agents has also been investigated. In melanoma, this grouping has been shown to improve response rate and survival in melanoma compared to each drug alone, suggesting synergistic activity of these agents (112). Based on promising pre-clinical data, the combination of MEDI4736 and tremilumumab (an anti-CTLA-4 agent) is being investigated in patients with advanced solid tumors, including gastric cancer (113). Immunotherapy could provide an unmet clinical need to patients with advanced GE cancers who might not benefit or be able to tolerate further traditional chemotherapy.


Guanylyl cyclase C (GCC) inhibitor

GCC, a trans-membrane cell surface receptor, is expressed on normal intestinal tissue but also expressed on the tumor cells of patients with gastrointestinal malignancies. Expression has been shown to be a good prognostic marker (39). Based on preclinical data that GCC on tumor cells has alterations in epithelial junctions, an antibody-drug conjugate MLN0264 was developed to preferentially target tumor cells. Based on promising phase I results (114), a phase II study of MLN 0264 in previously treated patients with gastric and GEJ cancers whose tumors express GCC by IHC is currently recruiting patients (115).


Conclusions

Together, GE cancers are among the most common malignancies worldwide (116). At diagnosis, approximately 50 percent of patients have disease that extends beyond locoregional confines. Cytotoxic agents have been the mainstay of systemic treatment for decades but carry significant toxicity.

During recent years, several molecular abnormalities underlying GE carcinogenesis have been identified. This has stimulated the search for targeted therapeutic approaches, and many studies are incorporating these agents with chemotherapy as described in this review.

The highly complex nature of the underlying molecular abnormalities and concurrent aberrations in multiple signaling pathways in GE cancers has been established (117). Because of the inherent redundancies in tumor molecular pathways, targeted agents used as monotherapy or even added to a chemotherapy backbone are unlikely to result in dramatic improvements in efficacy. However, pursuing multiple targets simultaneously might be logistically difficult given the current limited understanding of how to combine targeted agents, the issue of designing multi-sponsor trials, as well as the potential for additional toxicities. In the future, molecular profiling will play a role in identifying the specific patient who might benefit from targeted therapy, validate whether the drug inhibits the target, and determine if the tumor having the target is of functional importance.

To better achieve this goal of personalized cancer care, biomarkers should be utilized to predict the efficacy and toxicity of anticancer agents, as with HER2 overexpression prior to trastuzumab use. However, though selecting patients based on predictive factors is ideal, the lack of validated biomarkers in GE and the diversity of molecular alterations acquired during malignant transformation, recurrence or metastasis makes biomarker incorporation into clinical trials difficult.

Finally, the failure of phase III trials to demonstrate survival benefit despite promising results from phase II studies indicates the need to change the current evaluation system. Targeted agents often result in stable disease rather than disease response, which make assessment more challenging. OS should remain the primary end point of clinical trials because of the short survival in GE cancers.

Apart from the molecular targeted therapies described in this article, many other agents are currently being evaluated in GE cancers. Adequately powered, randomized trials are necessary to define the role of targeted therapies in advanced GE. Further work is needed to determine the optimal use of targeted therapy, validate biomarkers, and bring personalized medicine to GE adenocarcinomas.


Acknowledgements

Disclosure: The authors declare no conflict of interest.


References

  1. Siegel R, Ma J, Zou Z, et al. Cancer statistics, 2014. CA Cancer J Clin 2014;64:9-29. [PubMed]
  2. Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2009. CA Cancer J Clin 2009;59:225-49. [PubMed]
  3. Ajani JA, Moiseyenko VM, Tjulandin S, et al. Clinical benefit with docetaxel plus fluorouracil and cisplatin compared with cisplatin and fluorouracil in a phase III trial of advanced gastric or gastroesophageal cancer adenocarcinoma: the V-325 Study Group. J Clin Oncol 2007;25:3205-9. [PubMed]
  4. Coussens L, Yang-Feng TL, Liao YC, et al. Tyrosine kinase receptor with extensive homology to EGF receptor shares chromosomal location with neu oncogene. Science 1985;230:1132-9. [PubMed]
  5. King CR, Kraus MH, Aaronson SA. Amplification of a novel v-erbB-related gene in a human mammary carcinoma. Science 1985;229:974-6. [PubMed]
  6. Bilous M, Osamura RY, Rüschoff J, et al. HER-2 amplification is highly homogenous in gastric cancer. Hum Pathol 2010;41:304-5; author reply 305-6. [PubMed]
  7. Marx AH, Tharun L, Muth J, et al. HER-2 amplification is highly homogenous in gastric cancer. Hum Pathol 2009;40:769-77. [PubMed]
  8. Bozzetti C, Negri FV, Lagrasta CA, et al. Comparison of HER2 status in primary and paired metastatic sites of gastric carcinoma. Br J Cancer 2011;104:1372-6. [PubMed]
  9. Stintzing S, Jung A, Rossius L, et al. Mutations within the EGFR signaling pathway: Influence on efficacy in FIRE-3—A randomized phase III study of FOLFIRI plus cetuximab or bevacizumab as first-line treatment for wild-type (WT) KRAS (exon 2) metastatic colorectal cancer (mCRC) patients. J Clin Oncol 2014;32(suppl 3; abstr 445).
  10. Tanner M, Hollmén M, Junttila TT, et al. Amplification of HER-2 in gastric carcinoma: association with Topoisomerase IIalpha gene amplification, intestinal type, poor prognosis and sensitivity to trastuzumab. Ann Oncol 2005;16:273-8. [PubMed]
  11. Matsui Y, Inomata M, Tojigamori M, et al. Suppression of tumor growth in human gastric cancer with HER2 overexpression by an anti-HER2 antibody in a murine model. Int J Oncol 2005;27:681-5. [PubMed]
  12. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687-97. [PubMed]
  13. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350:2335-42. [PubMed]
  14. A study of trastuzumab emtansine versus taxane in patients with advanced gastric cancer [Internet]. 2014 [cited 2014 June 12]. Available online: http://clinicaltrials.gov/show/NCT01641939
  15. A study of capecitabine [xeloda] in combination with trastuzumab [herceptin] and oxaliplatine in patients with resectable gastric cancer [Internet]. 2014 [cited 2014 June 12]. Available online: http://clinicaltrials.gov/show/NCT01130337
  16. A study of the combination of oxaliplatin, capecitabine and herceptin (trastuzumab) and chemoradiotherapy in the adjuvant setting in operated patients with HER2+ gastric or gastro-esophageal junction cancer (TOXAG study) [Internet]. 2014 [cited 2014 June 12]. Available online: http://www.clinicaltrials.gov/show/NCT01748773
  17. Explorative phase II study of perioperative treatment in patients with adenocarcinoma of the gastroesophageal junction or stomach (HerFLOT) [Internet]. 2014 [cited 2014 June 12]. Available online: http://www.clinicaltrials.gov/ct2/show/NCT01472029?term=NCT01472029&rank=1
  18. Radiation therapy, paclitaxel, and carboplatin with or without trastuzumab in treating patients with esophageal cancer [Internet]. 2014 [cited 2014 June 12]. Available online: http://clinicaltrials.gov/show/NCT01196390
  19. Baselga J, Cortés J, Kim SB, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med 2012;366:109-19. [PubMed]
  20. Hoff P, Tabernero J, Shen L, et al. P-0111 Pertuzumab, trastuzumab and chemotherapy in HER2-positive metastatic gastric or gastro-oesophageal junction cancer: an international phase III study (JACOB). Ann Oncol 2013;24:iv67.
  21. Cameron D, Casey M, Press M, et al. A phase III randomized comparison of lapatinib plus capecitabine versus capecitabine alone in women with advanced breast cancer that has progressed on trastuzumab: updated efficacy and biomarker analyses. Breast Cancer Res Treat 2008;112:533-43. [PubMed]
  22. Geyer CE, Forster J, Lindquist D, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med 2006;355:2733-43. [PubMed]
  23. Administration US FDA. Lapatinib 2014 [Internet]. [cited 2014 June 14]. Available online: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/022059s016s017lbl.pdf
  24. Hecht JR, Bang YJ, Qin S, et al. Lapatinib in combination with capecitabine plus oxaliplatin (CapeOx) in HER2-positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma (AC): The TRIO-013/LOGiC Trial. J Clin Oncol 2013;31:abstr LBA4001.
  25. Satoh T, Xu RH, Chung HC, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol 2014;32:2039-49. [PubMed]
  26. Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys 2004;59:21-6. [PubMed]
  27. Oda K, Matsuoka Y, Funahashi A, et al. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol Syst Biol 2005;1:2005.0010.
  28. Wang KL, Wu TT, Choi IS, et al. Expression of epidermal growth factor receptor in esophageal and esophagogastric junction adenocarcinomas: association with poor outcome. Cancer 2007;109:658-67. [PubMed]
  29. Galizia G, Lieto E, Orditura M, et al. Epidermal growth factor receptor (EGFR) expression is associated with a worse prognosis in gastric cancer patients undergoing curative surgery. World J Surg 2007;31:1458-68. [PubMed]
  30. Lieto E, Ferraraccio F, Orditura M, et al. Expression of vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) is an independent prognostic indicator of worse outcome in gastric cancer patients. Ann Surg Oncol 2008;15:69-79. [PubMed]
  31. Martinelli E, De Palma R, Orditura M, et al. Anti-epidermal growth factor receptor monoclonal antibodies in cancer therapy. Clin Exp Immunol 2009;158:1-9. [PubMed]
  32. Saltz LB, Lenz HJ, Kindler HL, et al. Randomized phase II trial of cetuximab, bevacizumab, and irinotecan compared with cetuximab and bevacizumab alone in irinotecan-refractory colorectal cancer: the BOND-2 study. J Clin Oncol 2007;25:4557-61. [PubMed]
  33. Vermorken JB, Mesia R, Rivera F, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med 2008;359:1116-27. [PubMed]
  34. Lordick F, Kang YK, Chung HC, et al. Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:490-9. [PubMed]
  35. Ilson DH, Moughan J, Suntharalingam M, et al. RTOG 0436: A phase III trial evaluating the addition of cetuximab to paclitaxel, cisplatin, and radiation for patients with esophageal cancer treated without surgery. J Clin Oncol 2014;32:abstr 4007.
  36. Park SR, Kook MC, Choi IJ, et al. Predictive factors for the efficacy of cetuximab plus chemotherapy as salvage therapy in metastatic gastric cancer patients. Cancer Chemother Pharmacol 2010;65:579-87. [PubMed]
  37. Luber B, Deplazes J, Keller G, et al. Biomarker analysis of cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric and oesophago-gastric junction cancer: results from a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie (AIO). BMC Cancer 2011;11:509. [PubMed]
  38. Moehler M, Mueller A, Trarbach T, et al. Cetuximab with irinotecan, folinic acid and 5-fluorouracil as first-line treatment in advanced gastroesophageal cancer: a prospective multi-center biomarker-oriented phase II study. Ann Oncol 2011;22:1358-66. [PubMed]
  39. Pinto C, Di Fabio F, Siena S, et al. Phase II study of cetuximab in combination with FOLFIRI in patients with untreated advanced gastric or gastroesophageal junction adenocarcinoma (FOLCETUX study). Ann Oncol 2007;18:510-7. [PubMed]
  40. Lordick F, Luber B, Lorenzen S, et al. Cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric cancer: a phase II study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Br J Cancer 2010;102:500-5. [PubMed]
  41. Waddell T, Chau I, Cunningham D, et al. Epirubicin, oxaliplatin, and capecitabine with or without panitumumab for patients with previously untreated advanced oesophagogastric cancer (REAL3): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:481-9. [PubMed]
  42. Waddell T, Chau I, Barbachano Y, et al. A randomized multicenter trial of epirubicin, oxaliplatin, and capecitabine (EOC) plus panitumumab in advanced esophagogastric cancer (REAL3). J Clin Oncol 2012;30:abstr LBA4000.
  43. Lockhart AC, Reed CE, Decker PA, et al. Phase II study of neoadjuvant therapy with docetaxel, cisplatin, panitumumab, and radiation therapy followed by surgery in patients with locally advanced adenocarcinoma of the distal esophagus (ACOSOG Z4051). Ann Oncol 2014;25:1039-44. [PubMed]
  44. Rojo F, Tabernero J, Albanell J, et al. Pharmacodynamic studies of gefitinib in tumor biopsy specimens from patients with advanced gastric carcinoma. J Clin Oncol 2006;24:4309-16. [PubMed]
  45. Gefitinib in treating patients with esophageal cancer that is progressing after chemotherapy [Internet]. 2014 [cited 2014 June 14]. Available online: http://www.clinicaltrials.gov/ct2/show/NCT01243398?term=NCT01243398&rank=1
  46. Dragovich T, McCoy S, Fenoglio-Preiser CM, et al. Phase II trial of erlotinib in gastroesophageal junction and gastric adenocarcinomas: SWOG 0127. J Clin Oncol 2006;24:4922-7. [PubMed]
  47. Carmeliet P. Angiogenesis in health and disease. Nat Med 2003;9:653-60. [PubMed]
  48. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9:669-76. [PubMed]
  49. Karayiannakis AJ, Bolanaki H, Syrigos KN, et al. Serum vascular endothelial growth factor levels in pancreatic cancer patients correlate with advanced and metastatic disease and poor prognosis. Cancer Lett 2003;194:119-24. [PubMed]
  50. Maeda K, Chung YS, Takatsuka S, et al. Clinical significance of angiogenesis in gastric carcinoma as a predictive marker for recurrence. Gan To Kagaku Ryoho 1994;21:1283-5. [PubMed]
  51. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006;355:2542-50. [PubMed]
  52. Cannistra SA, Matulonis UA, Penson RT, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol 2007;25:5180-6. [PubMed]
  53. Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 2007;370:2103-11. [PubMed]
  54. Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 2007;357:2666-76. [PubMed]
  55. Ohtsu A, Shah MA, Van Cutsem E, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 2011;29:3968-76. [PubMed]
  56. Shah M, Kang Y, Ohtsu A, et al. Tumor and blood plasma biomarker analyses in the AVAGAST phase III randomized study of first-line bevacizumab + capecitabine/cisplatin in patients with advanced gastric cancer. European Society for Medical Oncology (ESMO) 2010.
  57. Fuchs CS, Tomasek J, Yong CJ, et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014;383:31-9. [PubMed]
  58. Administration US FDA. Ramucirumab 2014 [Internet]. [2014 June 15]. Available online: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/125477lbl.pdf
  59. Wilke H, Van Cutsem E, Oh SC, et al. RAINBOW: A global, phase 3, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy: Results of a multiple Cox regression analysis adjusting for prognostic factors. J Clin Oncol 2014;32:abstr 4076.
  60. Yoon HH, Bendell JC, Braiteh FS, et al. Ramucirumab (RAM) plus FOLFOX as front-line therapy (Rx) for advanced gastric or esophageal adenocarcinoma (GE-AC): Randomized, double-blind, multicenter phase 2 trial. J Clin Oncol 2014;32:abstr 4004.
  61. Shah MA, Ramanathan RK, Ilson DH, et al. Multicenter phase II study of irinotecan, cisplatin, and bevacizumab in patients with metastatic gastric or gastroesophageal junction adenocarcinoma. J Clin Oncol 2006;24:5201-6. [PubMed]
  62. Bang YJ, Kang YK, Kang WK, et al. Phase II study of sunitinib as second-line treatment for advanced gastric cancer. Invest New Drugs 2011;29:1449-58. [PubMed]
  63. Yi JH, Lee J, Lee J, et al. Randomised phase II trial of docetaxel and sunitinib in patients with metastatic gastric cancer who were previously treated with fluoropyrimidine and platinum. Br J Cancer 2012;106:1469-74. [PubMed]
  64. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007;356:125-34. [PubMed]
  65. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359:378-90. [PubMed]
  66. Yang S, Ngo VC, Lew GB, et al. AZD6244 (ARRY-142886) enhances the therapeutic efficacy of sorafenib in mouse models of gastric cancer. Mol Cancer Ther 2009;8:2537-45. [PubMed]
  67. Sun W, Powell M, O'Dwyer PJ, et al. Phase II study of sorafenib in combination with docetaxel and cisplatin in the treatment of metastatic or advanced gastric and gastroesophageal junction adenocarcinoma: ECOG 5203. J Clin Oncol 2010;28:2947-51. [PubMed]
  68. Martin-Richard M, Gallego R, Pericay C, et al. Multicenter phase II study of oxaliplatin and sorafenib in advanced gastric adenocarcinoma after failure of cisplatin and fluoropyrimidine treatment. A GEMCAD study. Invest New Drugs 2013;31:1573-9. [PubMed]
  69. Sorafenib as a second line treatment in patients with advanced or metastatic gastric cancer [Internet]. 2014 [cited 15 June 2014]. Available online: http://www.clinicaltrials.gov/ct2/show/NCT00595985
  70. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 2010;28:1061-8. [PubMed]
  71. van der Graaf WT, Blay JY, Chawla SP, et al. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2012;379:1879-86. [PubMed]
  72. Bible KC, Suman VJ, Molina JR, et al. Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study. Lancet Oncol 2010;11:962-72. [PubMed]
  73. FLO +/- pazopanib as first-line treatment in advanced gastric cancer (PaFLO) [Internet]. 2014 [cited 2014 June 22]. Available online: http://clinicaltrials.gov/ct2/show/NCT01503372
  74. A study of pazopanib with CAPEOX in AGC patients [Internet]. 2014 [cited 2014 June 22]. Available online: http://clinicaltrials.gov/ct2/show/NCT01130805
  75. Foulstone E, Prince S, Zaccheo O, et al. Insulin-like growth factor ligands, receptors, and binding proteins in cancer. J Pathol 2005;205:145-53. [PubMed]
  76. Matsubara J, Yamada Y, Nakajima TE, et al. Clinical significance of insulin-like growth factor type 1 receptor and epidermal growth factor receptor in patients with advanced gastric cancer. Oncology 2008;74:76-83. [PubMed]
  77. Baserga R, Peruzzi F, Reiss K. The IGF-1 receptor in cancer biology. Int J Cancer 2003;107:873-7. [PubMed]
  78. Attard G, Fong PC, Molife R, et al. Phase I trial involving the pharmacodynamic (PD) study of circulating tumour cells, of CP-751,871 (C), a monoclonal antibody against the insulin-like growth factor 1 receptor (IGF-1R), with docetaxel (D) in patients (p) with advanced cancer. J Clin Oncol 2006;24:abstr 3023.
  79. Grose R, Dickson C. Fibroblast growth factor signaling in tumorigenesis. Cytokine Growth Factor Rev 2005;16:179-86. [PubMed]
  80. Moffa AB, Tannheimer SL, Ethier SP. Transforming potential of alternatively spliced variants of fibroblast growth factor receptor 2 in human mammary epithelial cells. Mol Cancer Res 2004;2:643-52. [PubMed]
  81. Hattori Y, Itoh H, Uchino S, et al. Immunohistochemical detection of K-sam protein in stomach cancer. Clin Cancer Res 1996;2:1373-81. [PubMed]
  82. Takeda M, Arao T, Yokote H, et al. AZD2171 shows potent antitumor activity against gastric cancer over-expressing fibroblast growth factor receptor 2/keratinocyte growth factor receptor. Clin Cancer Res 2007;13:3051-7. [PubMed]
  83. Nakamura K, Yashiro M, Matsuoka T, et al. A novel molecular targeting compound as K-samII/FGF-R2 phosphorylation inhibitor, Ki23057, for Scirrhous gastric cancer. Gastroenterology 2006;131:1530-41. [PubMed]
  84. Lee JH, Han SU, Cho H, et al. A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene 2000;19:4947-53. [PubMed]
  85. Nakajima M, Sawada H, Yamada Y, et al. The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer 1999;85:1894-902. [PubMed]
  86. Jhawer M, Kindler HL, Wainberg Z, et al. Assessment of two dosing schedules of GSK1363089 (GSK089), a dual MET/VEGFR2 inhibitor, in metastatic gastric cancer (GC): Interim results of a multicenter phase II study. J Clin Oncol 2009;27:abstr 4502.
  87. Oliner KS, Tang R, Anderson A, et al. Evaluation of MET pathway biomarkers in a phase II study of rilotumumab (R, AMG 102) or placebo (P) in combination with epirubicin, cisplatin, and capecitabine (ECX) in patients (pts) with locally advanced or metastatic gastric (G) or esophagogastric junction (EGJ) cancer. J Clin Oncol 2012;30:abstr 4005.
  88. First-line treatment for locally advanced or metastatic mesenchymal epithelial transition factor (MET)-positive gastric, lower esophageal, or gastroesophageal junction (GEJ) adenocarcinoma (RILOMET-1) [Internet]. 2014 [cited 2014 August 23]. Available online: https://clinicaltrials.gov/ct2/show/NCT01697072
  89. Iveson T, Donehower RC, Davidenko I, et al. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: an open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study. Lancet Oncol 2014;15:1007-18. [PubMed]
  90. A study of onartuzumab (MetMAb) in combination with mFOLFOX6 in patients with metastatic HER2-negative and Met-positive gastroesophageal cancer (MetGastric) [Internet]. 2014 [cited 2014 August 23]. Available online: http://clinicaltrials.gov/ct2/show/NCT01662869
  91. Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer 2002;2:489-501. [PubMed]
  92. Yap TA, Garrett MD, Walton MI, et al. Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Curr Opin Pharmacol 2008;8:393-412. [PubMed]
  93. Yang ZZ, Tschopp O, Baudry A, et al. Physiological functions of protein kinase B/Akt. Biochem Soc Trans 2004;32:350-4. [PubMed]
  94. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004;304:554. [PubMed]
  95. Suzuki H, Freije D, Nusskern DR, et al. Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues. Cancer Res 1998;58:204-9. [PubMed]
  96. Yoshimoto M, Cunha IW, Coudry RA, et al. FISH analysis of 107 prostate cancers shows that PTEN genomic deletion is associated with poor clinical outcome. Br J Cancer 2007;97:678-85. [PubMed]
  97. Bellacosa A, Kumar CC, Di Cristofano A, et al. Activation of AKT kinases in cancer: implications for therapeutic targeting. Adv Cancer Res 2005;94:29-86. [PubMed]
  98. Oki E, Baba H, Tokunaga E, et al. Akt phosphorylation associates with LOH of PTEN and leads to chemoresistance for gastric cancer. Int J Cancer 2005;117:376-80. [PubMed]
  99. Yu HG, Ai YW, Yu LL, et al. Phosphoinositide 3-kinase/Akt pathway plays an important role in chemoresistance of gastric cancer cells against etoposide and doxorubicin induced cell death. Int J Cancer 2008;122:433-43. [PubMed]
  100. Okamoto I, Doi T, Ohtsu A, et al. Phase I clinical and pharmacokinetic study of RAD001 (everolimus) administered daily to Japanese patients with advanced solid tumors. Jpn J Clin Oncol 2010;40:17-23. [PubMed]
  101. Doi T, Muro K, Boku N, et al. Multicenter phase II study of everolimus in patients with previously treated metastatic gastric cancer. J Clin Oncol 2010;28:1904-10. [PubMed]
  102. Van Cutsem E, Yeh KH, Bang YJ, et al. Phase III trial of everolimus (EVE) in previously treated patients with advanced gastric cancer (AGC): GRANITE-1. J Clin Oncol 2012;30:abstr LBA3.
  103. Underhill C, Toulmonde M, Bonnefoi H. A review of PARP inhibitors: from bench to bedside. Ann Oncol 2011;22:268-79. [PubMed]
  104. Bang YJ, Im SA, Lee KW, et al. Olaparib plus paclitaxel in patients with recurrent or metastatic gastric cancer: A randomized, double-blind phase II study. J Clin Oncol 2013;31:abstr 4013.
  105. Kubota E, Williamson CT, Ye R, et al. Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines. Cell Cycle 2014;13:2129-37. [PubMed]
  106. Efficacy and safety study of olaparib in combination with paclitaxel to treat advanced gastric cancer [Internet]. 2014 [cited 2014 June 22]. Available online: http://clinicaltrials.gov/ct2/show/NCT01924533
  107. Evaluating the safety and tolerability of the Poly-ADP ribose (PARP) inhibitor with FOLFIRI in subjects with solid tumor [Internet]. 2014 [cited 2014 June 22]. Available online: http://clinicaltrials.gov/ct2/show/NCT01123876
  108. Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013;369:134-44. [PubMed]
  109. Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455-65. [PubMed]
  110. Muro K, Bang Y, Shankaran V, et al. LBA15 A phase 1b study of pembrolizumab (PEMBRO; MK-3475) in patients (PTS) with advanced gastric cancer. Ann Oncol 2014;25:v1-v41.
  111. Lutzky J, Antonia SJ, Blake-Haskins A, et al. A phase 1 study of MEDI4736, an anti–PD-L1 antibody, in patients with advanced solid tumors. J Clin Oncol 2014;32:abstr 3001.
  112. National Comprehensive Cancer Network. Gastric Cancer (Version 1.2014). Available online: http://www.debbiesdream.org/portal/documents/33005/671772/NCCN+Gastric+Cancer+Guidelines+2014.pdf
  113. A phase 1 study to evaluate MEDI4736 in combination with tremelimumab [Internet]. 2014 [cited 2014 November 2].
    Available online: http://clinicaltrials.gov/ct2/show/NCT01975831
  114. Messersmith W, Almhanna K, Rodon J, et al. PD-0032MLN0264, an investigational, first-in-class antibody-drug conjugate targeting guanylyl cyclase C (GCC): first-in-human study in patients with advanced gastrointestinal malignancies. Ann Oncol 2013;24:piv36.
  115. A study of MLN0264 in patients with cancer of the stomach or gastroesophageal junction [Internet]. 2014 [cited 2014 August 24]. Available online: http://clinicaltrials.gov/ct2/show/NCT02202759
  116. Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA Cancer J Clin 2011;61:113. [PubMed]
  117. Wang K, Yuen ST, Xu J, et al. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet 2014;46:573-82. [PubMed]
Cite this article as: Kothari N, Almhanna K. Current status of novel agents in advanced gastroesophageal adenocarcinoma. J Gastrointest Oncol 2015;6(1):60-74. doi: 10.3978/j.issn.2078-6891.2014.098

Download Citation