60
Views
4
CrossRef citations to date
0
Altmetric
Review

Tracking the 2015 Gastrointestinal Cancers Symposium: bridging cancer biology to clinical gastrointestinal oncology

, , , , , , , & show all
Pages 1149-1156 | Published online: 22 May 2015

Abstract

The 2015 Gastrointestinal Cancers Symposium (San Francisco, CA, USA; January 15–17) is the world-class conference co-sponsored by the American Society of Clinical Oncology, the American Society for Radiation Oncology, the American Gastroenterological Association Institute, and the Society of Surgical Oncology, in which the most innovative research results in digestive tract oncology are presented and discussed. In its twelfth edition, the meeting has provided new insights focusing on the underpinning biology and clinical management of gastrointestinal malignancies. More than 3,400 health care professionals gathered from all over the world to share their experiences on how to bridge the recent novelties in cancer biology with everyday medical practice. In this article, the authors report on the most significant advances, didactically moving on three different anatomic tracks: gastroesophageal malignancies, pancreatic and biliary cancers, and colorectal adenocarcinomas.

Introduction

Significant studies have been presented at the 2015 Gastrointestinal Cancers Symposium, an outstanding appointment that is held every January in San Francisco, CA, USA. Although this year the practice-changing results were limited, results of many studies have markedly contributed to the expansion of our knowledge in the biology of gastrointestinal malignancies and to fine-tune the available treatment options.

Aims of this report are to recall and comment on the most significant preclinical and clinical studies that have been presented at the scientific venue, which will continue the major progress made in the past decades.Citation1 In the gastroesophageal track, we will present and reason on the available results of the studies testing mesenchymal epithelial transition (MET) factor inhibitors (AMG 337 and onartuzumab) and programmed cell death receptor 1 ligand (PD-L1) inhibitors (pembrolizumab) in patients with upper gastrointestinal tract diseases. In the second section of the manuscript, we will describe how a comprehensive molecular profiling of biliary tract cancers may be used as a tool for treatment and prognostic stratification, update on the method for refining optimal candidates for sorafenib in hepatocellular carcinoma (HCC), and discuss how to expand valuable treatment options in pancreatic carcinomas, including the novel MM-398 and PF-04136309. In the final section focused on colorectal cancers (CRCs), we will face the most recent advances in molecular selection for targeted therapies and understand that CRC has many underlying drivers, as well as the opportunity to identify targets responsible for drug resistance and the possibility of reshaping the paradigm of sequential approach in multimodality therapies. Notable advances that may challenge the current paradigms in the management of rectal cancers are also presented. Results are summarized in .

Table 1 Significant clinical trials presented at the Gastrointestinal Cancers Symposium 2015 investigating novel agents in gastric, liver, and colorectal carcinomas

Steps ahead in upper gastrointestinal malignancies

Recent advances in the understanding of gastric cancer biology and the comprehensive molecular analysis performed by the Cancer Genome Atlas (TCGA), which allowed recognizing four distinct disease subtypes,Citation2 have served as the rationale for the development of novel targeted agents.Citation3 Overexpression or amplification of the MET factor has been observed in gastric cancerCitation4 and been correlated with unfavorable clinical outcomes.Citation5 Compared to the proportion of genes identified by immunohistochemical (IHC) detection of overexpression, the rate of detection of activating mutations or amplifications of MET gene is higher,Citation6 defining a small group of tumors with aggressive clinical behavior regardless of disease stage. AMG 337 is a highly selective, orally available MET inhibitor that showed promising preclinical activity. In a multicenter, Phase I, open-label trial, 80 patients with MET-amplified tumors and good performance status (PS) received increasing doses of AMG 337 monotherapy, defining 300 mg/day as the maximum tolerated dose.Citation7 In the small subset of 13 heavily pretreated patients with MET-amplified gastroesophageal tumors exposed to AMG 337, the investigators observed a notable 62% rate of response. Interestingly, the response was fast and usually detectable within 4 weeks from treatment start, which may be a remarkable advantage for symptomatic patients. The experimental treatment had a favorable profile of tolerability. Dose-limiting toxicities were severe hypertension, headache, and increased amylase activity; the most common side effects were headache (45%), nausea (32%), vomiting (21%), fatigue (14%), and peripheral edema (12%); headache (9%) and fatigue (4.5%) were the most frequent severe adverse events.

Onartuzumab is a recombinant, fully humanized, monoclonal anti-MET antibody. Results of the randomized Phase II trial testing upfront FOLFOX6 with onartuzumab at the dose of 10 mg/kg or FOLFOX6 plus placebo were presented.Citation8 One hundred and twenty-three patients with advanced gastroesophageal cancer were enrolled from 25 centers; key eligibility criteria included no previous treatments for metastatic disease, Eastern Cooperative Oncology Group (ECOG) PS status of 0 or 1, and retention of organ function. MET-positive patients, defined as those with ≥50% of tumor with moderate–strong intensity staining by centrally reviewed IHC, were well balanced in the two arms: 28% in the experimental arm vs 33% in the control arm. Stratification factors were histological subtype and previous gastrectomy. At data cutoff, 96 out of 121 randomized patients had progression-free survival (PFS) events, 74% of those exposed to FOLFOX and onartuzumab and 82% of those receiving FOLFOX and placebo. The primary end point, PFS, in the intention-to treat population, was not met (6.77 months in the onartuzumab arm, 6.97 months in the placebo arm; hazard ratio [HR]: 1.08, 95% confidence interval [CI]: 0.71–1.63). In addition, the preplanned analyses in the MET-positive population generated similarly disappointing results: median PFS was 5.95 months for those exposed to FOLFOX6 and onartuzumab vs 6.8 months for those treated with FOLFOX6 and placebo (HR: 1.38). No differences were found despite the use of different definitions for MET positivity. The addition of onartuzumab was not beneficial, regardless of patients’ race, and produced more adverse events. It is unclear whether these negative results suggest a failure of the strategy or the unreliability of the current definition of MET positivity. In fact, while it is reasonable not to consider IHC MET overexpression a good driver for patient selection, it should also be recalled that a large Phase III trial testing onartuzumab was prematurely interrupted because of failure of the drug in lung cancers,Citation9 and another randomized study testing rilotumumab in the advanced disease setting failed to meet the primary trial end point.Citation10

Targeting the immune checkpoints in solid malignancies is becoming a major methodological approach. T-lymphocytes may recognize and eliminate cancer antigens, while immune checkpoints such as cytotoxic T-lymphocyte-associated antigen (CTLA)-4 and programmed cell death (PD-1) receptor and its ligands (PD-L1, PD-L2) are able to suppress the activity of T-lymphocytes. Therefore, enhancing antitumor immunity by blocking PD-1 is now an attractive reality. Pembrolizumab, a highly selective IgG4k, humanized monoclonal antibody against PD-1, has recently received approval from the US Food and Drug Administration for the treatment of advanced melanoma after the failure of ipilimumab administration or BRAF V600E-mutant melanoma in progression following BRAF inhibitor administration.Citation11 Because the high expression of PD-L1 on tumor gastric cellsCitation12 and macrophages can suppress immune surveillance and permit neoplastic growth,Citation13 the molecule has become an interesting target even in gastric cancer. Preliminary data of the KEYNOTE-012 gastric cohort study, in which pembrolizumab (MK-3475) was given at 10 mg/kg every 2 weeks to 39 patients with PD-L1-positive advance gastric cancer, were recently presented.Citation14 The trial enrolled heavily pretreated Asian (19) or non-Asian (20) patients, wherein 67% received ≥2 treatment lines. Overall response rate (RR) was 30.8% (95% CI: 17.0–46.6) and 41% of patients experienced a decrease in tumor burden. Aim of the abstract presented at the 2015 Gastrointestinal Cancers Symposium was to analyze the relationship between PD-L1 expression and clinical outcome in patients with advanced disease treated with pembrolizumab.Citation15 Muro et alCitation15 found a significant association between PD-L1 expression level and objective response rate (ORR; one-sided P=0.10). Median overall survival (OS) was not reached, but the 6-month OS rate was surprisingly high (69%). Though described as easily manageable by the authors, the toxicity profile appears a bit challenging, with five severe adverse events (peripheral sensory neuropathy, fatigue, decreased appetite, hypoxia, and pneumonitis) and one drug-related death (hypoxia). Because the immune-related response may not be fully captured by conventional response criteria,Citation16 it would be interesting to assess the response with immune-related response criteria to further confirm the activity of pembrolizumab. On the basis of the KEYNOTE results, a Phase III randomized trial that compares pembrolizumab to paclitaxel in patients with recurrent or metastatic gastric or gastroesophageal junction adenocarcinoma who progressed after first-line treatment has been planned.

Perioperative chemotherapy is a standard of care in locally advanced gastroesophageal cancer in Europe.Citation17,Citation18 In the MAGIC population, the human epidermal growth factor receptor 2 (HER2) positivity was not a prognostic or predictive factor.Citation19 In the NEOHX study,Citation20 36 patients with HER2-positive locally advanced gastric adenocarcinoma entered the Phase II trial to explore the combination of trastuzumab and chemotherapy in this setting. Patients were treated with three preoperative cycles of standard XELOX plus trastuzumab (loading dose 8 mg/kg, instead of 6 mg/kg), followed by definitive surgery. In the postoperative phase, patients received three other cycles of the same combination regimen and eventually 12 cycles of trastuzumab monotherapy. The primary end point of the study was 18 months disease-free survival (DFS). Median age at trial entry was 63 years. R0 resection was reached in 78% of the patients (three cases had a pathological complete response), median DFS rate at 18 months was 71%, 2-year OS rate was 75%, while median DFS and OS had not been reached. The trial results are very interesting in this biologically selected population and may promote the use of trastuzumab in resectable gastric cancer patients.

Significant advances in HCC, pancreatic cancer, and ciliary tract adenocarcinoma

Ramucirumab is a humanized monoclonal antibody that specifically blocks the vascular endothelial growth factor receptor 2 (VEGFR2).Citation21 The REACH trial is a randomized Phase III study investigating ramucirumab as a single agent for the treatment of advanced HCC after failure of first-line sorafenib.Citation22 Unfortunately, the trial did not meet its primary end point. In fact, median OS was 9.2 months for ramucirumab vs 7.6 months for placebo. Interestingly the prespecified subgroup analysis suggested that baseline alpha-fetoprotein (AFP) level might be a predictive marker for ramucirumab efficacy. Among 250 patients with baseline AFP ≥400 ng/mL, OS HR was 0.67 (95% CI: 0.51–0.90; P=0.0059) (). Median OS was 7.8 months for ramucirumab vs 4.2 months for placebo. Accordingly, in patients with a baseline AFP ≥1.5× the upper limit, OS was 8.6 months for ramucirumab vs 5.7 months for placebo and the HR was 0.749 (95% CI: 0.603–0.930; P=0.0088). In the REACH trial, AFP baseline levels seemed to be correlated with clinical outcome during ramucirumab treatment. Hopefully, the impact of this novel treatment approach will be proved prospectively in future clinical trials incorporating findings from the REACH study.

Casadei Gardini et alCitation23 presented a new prognostic factor for advanced HCC patients receiving sorafenib. The authors hypothesized that the antiangiogenic compound might induce an inhibition of endothelial nitric oxide synthase (eNOS) activity by blocking the VEGFRs, with a consequent decrease of the production of nitric oxide, which is correlated with tumor angiogenesis, invasion, and metastasis. On the basis of this assumption, eNOS polymorphisms were retrospectively investigated in 54 HCC patients receiving sorafenib. Patients carrying the b allele (five repetitions of 27 bp) of eNOS were found to have improved OS. The variants 4aa (four repeats of 27 bp in homozygosis), 4ab, and 4bb were associated with a median OS of 5.7 months, 13.9 months, and 23.6 months, respectively (P=0.016). For eNOS-786, the presence of the T allele was associated with a statistically significant, longer median OS (15.6 months vs 13.9 months, respectively; P=0.031).

Two Phase II randomized studies with novel antitumor agents were presented.Citation24,Citation25 Dovitinib was tested in patients with advanced HCC in an attempt to overcome the fibroblast growth factor receptors (FGFRs)-activated mechanism of resistance, which is considered an escape pathway for sorafenib activity. Nonetheless, dovitinib failed to prove superior to sorafenib in the first-line treatment of HCC. In the Phase II trial presented by Cheng et alCitation24 OS was 34.6 weeks for dovitinib and 36.7 weeks (23.3 weeks–49.3 weeks) for sorafenib (HR: 1.27; 95% CI: 0.89–1.80). Palmer et alCitation25 presented disappointing results for the triple angiokinase inhibitor nintedanib. In a randomized Phase II trial, time to progression (TTP) was similar between nintedanib and sorafenib (TTP: 5.5 months vs 3.8 months; HR: 1.05; 95% CI: 0.63–1.76). Accordingly, median OS was similar between the treatment arms (11.9 months vs 11.4 months; HR: 0.88; 95% CI: 0.52–1.47) ().Citation25

Prajapati et alCitation26 proposed a novel staging system for patients with advanced HCC treated with doxorubicin drug-eluting beads transarterial chemoembolization. In the multivariate analysis, the independent factors for survival were Child-Pugh class, ECOG PS, number of HCC lesions, index HCC (iHCC) size, site of portal or hepatic venous thrombosis, extrahepatic metastasis, and serum creatinine and AFP levels. Consequentially, the Clinical, Imaging, and Serum examination (CIS) staging system was proposed. According to the study findings, patients were grouped into different stages with different outcomes. The OS of stages I, II, III, and IV was 40.4 months, 24 months, 10.6 months, and 2.6 months, respectively (P<0.0001).

A prognosis model was also suggested for patients with locally advanced unresectable pancreatic cancer included in the LAP 07 trial.Citation27 In the multivariate analysis, the independent factors for OS were age, pain, albumin levels, and Response Evaluation Criteria in Solid Tumors (RECIST) size. Three risk groups were identified: lower risk (n=17; median OS: 18.8 months; group of reference); intermediate risk (n=166; median OS: 13.4 months); higher risk (n=187; median OS: 11.8 months). This easy-to-use prognostic model might be relevant for treatment decision in clinical practice and future trial design. Novel potential therapeutic options for metastatic pancreatic cancer have been suggested from analyses of randomized trials. In the three-arm NAPOLI-1 trial,Citation28 417 patients who had failed a gemcitabine-based first-line treatment were randomized to MM-398 (nanoliposomal encapsulation of irinotecan), 5-fluorouracil (FU) plus leucovorin (LV), or a combination of MM-398 and 5FU/LV. The primary end point of the trial was OS. The analysis in the per-protocol population (patients who received at least 80% of the target dose in the first 6 weeks and did not violate any inclusion/exclusion criteria) confirmed the favorable OS, PFS, and ORR for the combination MM-398+5FU/LV arm relative to the control 5FU/LV arm with a manageable safety profile, whereas single-agent MM-398 was not superior to 5FU/LV.

A Phase I trial investigated the role of PF-04136309 (a novel C–C chemokine receptor type 2 inhibitor) in combination with FOLFIRINOX for locally advanced pancreatic cancer.Citation29 Among 23 evaluable patients, 21 (91%) completed all six cycles; 12 (52.2%) obtained a RECIST-defined partial remission, whereas the remaining 11 (48%) had stable disease. Curative resections were achieved in four patients out of five with borderline resectable disease and in two with locally advanced pancreatic cancer. Globally, these very initial findings suggest that PF-04136309 and MM-398+5FU/LV should undergo further investigation in pancreatic cancer.

Relevant information about the genomic landscape of biliary tract cancer was derived from an analysis including >500 hundred patients.Citation30 The primary aim of the study was to identify crucial molecular markers as potential candidates for targeted agents. Findings from this analysis showed that intrahepatic cholangiocarcinoma (IHCCA), extrahepatic cholangiocarcinoma (EHCCA) and gall bladder carcinoma (GBCA) share genomic alterations in cell cycle regulation (CDKN2B) and chromatin remodeling (ARID1A). IHCCA features FGFR fusions, isocitrate dehydrogenase (IDH)-1/2 substitutions, BRAF substitutions, and MET amplification, with a low KRAS-mutation frequency. EHCCA and GBCA feature ERBB2 amplifications (GBCA > EHCCA) and PIK3CA/MTOR pathway alterations. EHCCA has a high KRAS-mutation frequency, whereas the KRAS genomic alteration in GBCA is low. Knowledge of relevant genomic alterations will be a fundamental starting point in designing future clinical trials in this setting.

We believe that new data deriving from the 2015 Gastrointestinal Cancers Symposium on HCC, pancreatic cancer, and biliary tract cancer will serve as relevant integration to our scientific knowledge about this heterogeneous group of neoplasms. Although the search for novel therapeutic targets represents a priority in this setting, we should remember that an improvement in the use of already available treatment options is equally important.Citation31,Citation32 Recent presentation of results from an analysis of polymorphisms in the angiogenic pathway of HCC patients receiving sorafenib should be integrated with already known data in this area and with clinical data defining prognostic groups.Citation32,Citation33 Similar considerations are applicable to data presented on patients with pancreatic cancer and biliary tract cancer.

Colorectal cancers: integrating novel drugs in innovative strategies

Results of the treatment with new antiangiogenic agents have been reported. Two studies suggested that ramucirumab and famitinib might soon be added to the armamentarium for metastatic CRC. The RAISE trial demonstrated that the addition of ramucirumab to second-line FOLFIRI chemotherapy resulted in a significant delay in disease progression and prolongation of survival in patients with metastatic CRC who had previously failed a first-line therapy containing bevacizumab.Citation34 The study included 1,072 patients with metastatic disease randomly assigned to ramucirumab (a human immunoglobulin G-1 monoclonal antibody that targets the extracellular domain of VEGFR2) plus FOLFIRI every 2 weeks per cycle (n=536) or placebo plus FOLFIRI every 2 weeks per cycle (n=536). The primary end point was OS. Ramucirumab reduced the risk for death by 16% (HR: 0.84; P=0.0219) and prolonged survival by a median of 1.6 months compared with FOLFIRI alone (median OS: 13.3 months vs 11.7 months). In addition, ramucirumab reduced the risk of disease progression by 21% (HR: 0.79; P=0.0005).

The efficacy and safety of famitinib, a small multi-target receptor tyrosine kinase inhibitor, which primarily inhibits VEGFR2, cKit, and PDGFR, were evaluated in a multicenter, randomized, double-blind, Phase II trial.Citation35 The study included 154 advanced CRC patients, who were randomly assigned in a 2:1 ratio to receive either famitinib or placebo. All patients had previously failed at least two lines of standard therapy. The primary end point was PFS. Famitinib increased median PFS from 1.5 months to 2.8 months (HR: 0.58; 95% CI: 0.41–0.86; P=0.0034). No statistical difference was observed in median OS with famitinib vs placebo (7.5 months vs 7.6 months; HR: 1.10; 95% CI: 0.76–1.60; P=0.605). The safety profile of famitinib was similar to that of other anti-VEGFR agents. Although these studies indicate that there will be alternative treatment options for metastatic CRC in the near future, they represent confirmations of the previously consolidated strategy of chemotherapy and antiangiogenic agent combination in unselected patients.Citation36 During this edition of the conference, a significant convergence was also demonstrated in refining molecular selection to increase the effectiveness of targeted therapies and particularly in developing a “beyond-RAS” strategy;Citation37 many works from all over the world have explored new screening, predictive biomarkers, and targeted therapies for metastatic CRC patients.

Preliminary data from two multinational screening platforms show that collaborative genomic analysis beyond the RAS is feasible and might be used to run next-generation trials with targeted agents. The European Organisation for Research and Treatment of Cancer (EORTC) Screening Patients for Efficient Clinical Trial Access in advanced colorectal cancer (SPECTAcolor), a pan-European network, has already analyzed 406 patients in the first year and has planned to use next-generation sequencing for 360 key cancer alterations.Citation38 A nationwide genomic screening project in Japan has evaluated a total of 361 tumor samples in the first 6 months to detect rare mutations such as PI3KCA and BRAF mutations.Citation39

Some other studies have shown that a strategy “from bench to bedside” is being pursued. The combination of the EGFR inhibitor panitumumab with the BRAF inhibitor vemurafenib for BRAF-mutant metastatic CRC has shown interesting hints of activity.Citation40 Two of 12 evaluable patients (13%) had confirmed long-lasting partial responses, whereas 8 patients (53%) had stable disease. The combination was well tolerated, with acneiform rash, fatigue, and arthralgia being the most frequent treatment-related adverse events. On the basis of the same assumption that activation of the EGFR pathway is the leading cause of failure of anti-BRAF monotherapy, a randomized Phase II trial of irinotecan plus cetuximab with or without vemurafenib is also currently ongoing.Citation41

Because another mechanism of resistance to anti-EGFR therapy is supposed to be HER2 amplification, in the Italian study HERACLES, heavily pretreated patients with HER2-positive, EGFR inhibitor-refractory advanced CRC have been treated with the combination of lapatinib and trastuzumab.Citation42 As expected from the literature,Citation43 the screening failure for HER2-positive patients was impressively high. Eighteen patients were enrolled (2.8% of the patients screened), and an ORR of 33% was achieved. Patients with HER2 3+ CRC and with a gene copy number ≥20 had the highest chance to respond to the dual inhibition.

Besides the new therapies and new strategies, the 2015 Gastrointestinal Cancers Symposium challenged some old paradigms. A retrospective review of 145 patients with locally advanced rectal cancer who received standard neoadjuvant chemoradiation suggests that patients who obtained a clinical complete response can avoid surgery with little compromise in the overall outcomes.Citation44 After a median follow-up of 3.5 years, there were no significant differences in disease-specific parameters or OS between patients who had a clinical complete response to preoperative therapy and skipped surgery on the one hand and patients who underwent rectal resection with a pathologic complete response (pCR) on the other. The study highlighted that a nonoperative approach may obtain preservation of rectal function in more than three-fourths of the patients.

The optimal timing of surgical resection of rectal cancer after preoperative chemoradiation was investigated in a retrospective analysis of 6,805 patients in the National Cancer Database.Citation45 A significant relationship between time delay (TD) and pCR was demonstrated (P=0.0002). At TD <30 days, 4.0% of patients achieved pCR, while 9.3% of patients achieved pCR by 75 days. At TD >75 days, the rate of pCR decreased. However, TD of >60 days was associated with 20% greater risk of mortality (95% CI: 1.07–1.36). Direct correlation between pCR and survival does not seem very strong in rectal cancer.

Conclusion

In the era of targeted therapies, trastuzumab and ramucirumab are the only biologic therapy agents approved for patients with advanced gastroesophageal cancers, the first being limited for use in the 15% of patients with HER2- positive tumors. There is a need for novel agents to be offered to these patients. In this landscape, MET inhibitors fostered the debate, with discordant results: while onartuzumab and rilotumumab did not fulfill their promises, AMG 337 provided interesting results. After several years of intense clinical and translational research, sorafenib still remains the only available therapeutic option for patients with advanced HCC. However, novel prognostic markers and interesting treatment strategies might, in the near future, change the way we approach this highly deadly disease. Besides TH-302, other novel drugs have emerged against pancreatic cancers. MM-398, an investigational nanocompound consisting of the chemotherapeutic irinotecan encapsulated in a liposomal sphere, and PF-04136309, a novel orally available human chemokine receptor 2 antagonist with immunomodulating and antineoplastic properties, may provide new hope for patients with advanced disease when combined with standard chemotherapy. With the expansion of the process of knowledge acquisition on CRC biology and, correspondingly, of growth of complexity, famitinib and ramucirumab will provide new options to patients with advanced disease. Once again, many studies have been presented. Hopefully, the time needed to transfer these scientific advances to clinical practice will be shorter than ever.

Disclosure

The authors report no conflicts of interest in this work.

References

  • MayerRJVenookAPSchilskyRLProgress against GI cancer during the American Society of Clinical Oncology’s first 50 yearsJ Clin Oncol201432151521153024752046
  • Cancer Genome Atlas Research NetworkComprehensive molecular characterization of gastric adenocarcinomaNature2014513751720220925079317
  • AprileGGiampieriRBonottoMThe challenge of targeted therapies for gastric cancer patients: the beginning of a long journeyExpert Opin Investig Drugs2014237925942
  • AnXWangFShaoQMET amplification is not rare and predicts unfavorable clinical outcomes in patients with recurrent/metastatic gastric cancer after chemotherapyCancer2014120567568224804300
  • LennerzJKKwakELAckermanAMET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinibJ Clin Oncol201129364803481022042947
  • GrazianoFGalluccioNLorenziniPGenetic activation of the MET pathway and prognosis of patients with high-risk, radically resected gastric cancerJ Clin Oncol201129364789479522042954
  • KwakELLoRussoPHamidOClinical activity of AMG 337, an oral MET kinase inhibitor, in adult patients (pts) with MET-amplified gastroesophageal junction (GEJ), gastric (G), or esophageal (E) cancerJ Clin Oncol201533suppl 3 abstr 1
  • ShahMAChoJYHuatITRandomized phase II study of FOLFOX +/− MET inhibitor, onartuzumab (O), in advanced gastroesophageal adenocarcinoma (GEC)J Clin Oncol201533suppl 3 abstr 2
  • Available from: http://www.roche.com/media/store/releases/med-cor-2014-03-03.htmAccessed on February 8th 2015
  • Available from: http://www.prnewswire.com/news-releases/amgen-announces-termination-of-all-amgen-sponsored-clinical-studies-of-rilotumumab-in-advanced-gastric-cancer-300000103.htmlAccessed on February 8th 2015
  • HerseyPGowrishankarKPembrolizumab joins the anti-PD-1 armamentarium in the treatment of melanomaFuture Oncol201511113314025572788
  • SunJXuKWuCPD-L1 expression analysis in gastric carcinoma tissue and blocking of tumor-associated PD-L1 signaling by two functional monoclonal antibodiesTissue Antigens2007691192717212704
  • PardollDMThe blockade of immune checkpoints in cancer immunotherapyNat Rev Cancer201212425226422437870
  • MuroKBangYShankaranVA phase 1B study of pembrolizumab (PEMBRO; MK-3475) in patients (PTS) with advanced gastric cancerAnn Oncol201425suppl 5v1v41
  • MuroKBangYJShankaranVRelationship between PD-L1 expression and clinical outcomes in patients (Pts) with advanced gastric cancer treated with the anti-PD-1 monoclonal antibody pembrolizumab (Pembro; MK-3475) in KEYNOTE-012J Clin Oncol201533suppl 3 abstr 3
  • WolchokJDHoosAO’DaySGuidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteriaClin Cancer Res200915237412747419934295
  • CunninghamDAllumWHStenningSPMAGIC Trial ParticipantsPerioperative chemotherapy versus surgery alone for resectable gastroesophageal cancerN Engl J Med20063551112016822992
  • YchouMBoigeVPignonJPPerioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trialJ Clin Oncol201129131715172121444866
  • OkinesAFThompsonLCCunninghamDEffect of HER2 on prognosis and benefit from peri-operative chemotherapy in early oesophago-gastric adenocarcinoma in the MAGIC trialAnn Oncol20132451253126123233651
  • RiveraFJimenez-FonsecaPGarcia AlfonsoPNEOHX study: perioperative treatment with trastuzumab in combination with capecitabine and oxaliplatin (XELOX-T) in patients with HER-2 resectable stomach or esophagogastric junction (EGJ) adenocarcinoma – 18 m DFS analysisJ Clin Oncol201533suppl 3 abstr 107
  • AprileGRijavecEFontanellaCRihawiKGrossiFRamucirumab: preclinical research and clinical developmentOnco Targets Ther201471997200625378934
  • ZhuAXRyooBYYenCJRamucirumab (RAM) as second-line treatment in patients (pts) with advanced hepatocellular carcinoma (HCC): analysis of patients with elevated α-fetoprotein (AFP) from the randomized phase III REACH studyJ Clin Oncol201533suppl 3 abstr 232
  • Casadei GardiniAMarisiGScarpiEeNOS polymorphisms in relation to outcome in advanced HCC patients receiving sorafenibJ Clin Oncol201533suppl 3 abstr 230
  • ChengALThongprasertSLimHYPhase II study of front-line dovitinib (TKI258) versus sorafenib in patients (Pts) with advanced hepatocellular carcinoma (HCC)J Clin Oncol201533suppl 3 abstr 237
  • PalmerDHMaYTPeck-RadosavljevicMRandomized phase II trial comparing the efficacy and safety of nintedanib versus sorafenib in patients with advanced hepatocellular carcinoma (HCC)J Clin Oncol201533suppl 3 abstr 238
  • PrajapatiHJSik KimHSNew prognostic staging system from the multivariate survival analysis (MVA) of the patients with unresectable hepatocellular carcinoma (HCC) treated with doxorubicin drug eluting beads transarterial chemoembolization (DEB TACE)J Clin Oncol201533suppl 3 abstr 236
  • VernereyDHammelPPaget-BaillySPrognosis model for overall survival in locally advanced unresecable pancreatic carcinoma: an ancillary study of the LAP 07 trialJ Clin Oncol201533suppl 3 abstr 235
  • ChenLTVon HoffDDPin LiCExpanded analyses of napoli-1: Phase 3 study of MM-398 (nal-IRI), with or without 5-fluorouracil and leucovorin, versus 5-fluorouracil and leucovorin, in metastatic pancreatic cancer (mPAC) previously treated with gemcitabine-based therapyJ Clin Oncol201533suppl 3 abstr 234
  • Wang-GillamANyweningTMSanfordDMPhase IB study of FOLFIRINOX plus PF-04136309 in patients with borderline resectable and locally advanced pancreatic adenocarcinoma (PC)J Clin Oncol201533suppl 3 abstr 338
  • RossJSWangKCatenacciDVTComprehensive genomic profiling of biliary tract cancers to reveal tumor-specific differences and genomic alterationsJ Clin Oncol201533suppl 3 abstr 231
  • PierantoniCPagliacciAScartozziMBerardiRBianconiMCascinuSPancreatic cancer: progress in cancer therapyCrit Rev Oncol Hematol2008671273818356073
  • FaloppiLScartozziMMaccaroniEEvolving strategies for the treatment of hepatocellular carcinoma: from clinical-guided to molecularly-tailored therapeutic optionsCancer Treat Rev2011373167177
  • ScartozziMFaloppiLSvegliati BaroniGVEGF and VEGFR genotyping in the prediction of clinical outcome for HCC patients receiving sorafenib: the ALICE-1 studyInt J Cancer201413551247125624510746
  • TaberneroJCohnALObermannovaRRAISE: a randomized, double-blind, multicenter phase III study of irinotecan, folinic acid, and 5-fluorouracil (FOLFIRI) plus ramucirumab (RAM) or placebo (PBO) in patients (pts) with metastatic colorectal carcinoma (CRC) progressive during or following first-line combination therapy with bevacizumab (bev), oxaliplatin (ox), and a fluoropyrimidine (fp)J Clin Oncol201533suppl 3 abstr 512
  • XuRShenLWangKA randomized, double-blind, parallel-group, placebo-controlled, multicenter, phase II clinical study of famitinib in the treatment of advanced metastatic colorectal cancerJ Clin Oncol201533suppl 3 abstr 513
  • AprileGLutrinoSEFerrariLEvidence-based appraisal of the upfront treatment for unresectable metastatic colorectal cancer patientsWorld J Gastroenterol201319468474848824379565
  • GiampieriRAprileGDel PreteMBeyond RAS: the role of epidermal growth factor receptor (EGFR) and its network in the prediction of clinical outcome during anti-EGFR treatment in colorectal cancer patientsCurr Drug Targets201415131225123025382208
  • FolprechtGAustDERothAImproving access to molecularly defined clinical trials for patients with colorectal cancer: the EORTC SPECTAcolor platformJ Clin Oncol201533suppl 3 abstr 575
  • ShitaraKFujiiSDendaTThe nationwide genomic screening project for gastrointestinal cancer in Japan (GI-SCREEN): simultaneous identification of KRAS, NRAS, BRAF, and PIK3CA mutation in advanced colorectal cancer (aCRC) (GI-SCREEN 2013-01)J Clin Oncol201533suppl 3 abstr 578
  • YaegerRDCercekAO’ReillyEMPilot study of vemurafenib and panitumumab combination therapy in patients with BRAF V600E mutated metastatic colorectal cancerJ Clin Oncol201533suppl 3 abstr 611
  • YaegerRCercekAO’ReillyEMS1406: randomized phase II study of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (mCRC)J Clin Oncol201533suppl 3 abstr TPS790
  • SienaSSartore-BianchiATrusolinoLTherapeutic dual inhibition of HER2 pathway for metastatic colorectal cancer (mCRC): the HERACLES trialJ Clin Oncol201533suppl 3 abstr 565
  • Ingold HeppnerBBehrensHMBalschunKHER2/neu testing in primary colorectal carcinomaBr J Cancer2014111101977198425211663
  • SmithJJChowOSEatonAOrgan preservation in patients with rectal cancer with clinical complete response after neoadjuvant therapyJ Clin Oncol201533suppl 3 abstr 509
  • HuntingtonCHBoselliDHillJSJonathanCOptimal timing of surgical resection after radiation therapy in locally advanced rectal adenocarcinoma: an analysis of the National Cancer Database (NCDB)J Clin Oncol201533suppl 3 abstr 510