899
Views
1
CrossRef citations to date
0
Altmetric
Editorial

Biologics in combination with chemotherapy for gastric cancer: is this the answer?

, , , , &

Abstract

Gastric cancer (GC) continues to be a significant problem worldwide and is the third leading cause of cancer death. Armamentarium to treat GC whether it is potentially curable or metastatic (incurable) has changed little over the last decades with only two new agents being approved (trastuzumab and ramucirumab). Many relatively healthy patients after second-line therapy have limited and generally ineffective options. The recent The Cancer Genome Atlas analysis has uncovered four genotypes of GC; however, it is not sufficient to change our treatment strategies and more work needs to be done. The popular front-line regimen containing a platinum compound and a fluoropyrimidine is widely used for drug development and has worked well globally. Thus, this combination appears suitable for adding a biologic agent. The search for new classes of cytotoxics has almost stopped, but it is clear that cytotoxic therapy continues to contribute and it is here to stay. Biologic agents that modulate the immune system of the host appear promising along with many other biologics that can potentially inhibit signaling pathways that are often employed by GC cells. We will briefly describe the efforts that have targeted EGFR, mTOR, angiogenesis and MET pathways.

1. Introduction

Gastric cancer (GC) continues to be a significant problem worldwide and is the third leading cause of cancer death Citation[1]. Armamentarium to treat GC whether it is potentially curable or metastatic (incurable) has changed little over the last decades with only two new agents being approved (trastuzumab and ramucirumab). Many relatively healthy patients after second-line therapy have limited and generally ineffective options. The recent The Cancer Genome Atlas (TCGA) analysis has uncovered four genotypes of GC Citation[2]. TCGA Research Network has classified GC into four subtypes based on the molecular characterization of 295 primary adenocarcinomas, but it is not clear if these genotypes will ultimately guide patient therapy. However, they clearly converged on four major genomic subtypes of GC with distinct features and classes of molecular alterations:

  1. Tumors containing EBV, along with recurrent mutations in the PIK3CA gene pathway, extreme DNA hypermethylation, amplification of JAK2 and extra copies of PD-L1 and PD-L2 genes, which are suppressors of immune response. This group makes up about 10% of the cancers, with nearly 80% harboring a protein-changing alteration in PIK3CA.

  2. Tumors showing microsatellite instability, in which malfunctioning DNA repair mechanisms cause a high rate of mutations, including mutations of genes encoding targetable oncogenic signaling proteins. About 20% of tumors fall into this subtype.

  3. The largest category (50%) of tumors, making up about half of the cancer specimens, is termed ‘chromosomally unstable.’ These contain a jumble of extra or missing pieces of genes and chromosomes (aneuploidy) and have a striking number of genomic amplifications of key receptor tyrosine kinases. This subtype of tumor is frequently found in the junction between the stomach and the esophagus, a type of GC that has been dramatically increasing in the US.

  4. The fourth group was termed ‘genomically stable’ since they lacked the molecular features of the other three types. These tumors, making up 20% of the specimens are largely those of a specific class of GC enriched for the diffuse-type histologic variant, with approximately 30% of these tumors having genomic alterations in the RHOA signaling pathway.

However, it is not sufficient to change our treatment strategies and more work needs to be done. The popular front-line regimen containing a platinum compound and a fluoropyrimidine is widely used for drug development and has worked well globally. Thus, this combination appears suitable for adding a biologic agent. The search for new classes of cytotoxics has almost stopped, but it is clear that cytotoxic therapy continues to contribute and it is here to stay. Biologic agents that modulate the immune system of the host appear promising along with many other biologics that can potentially inhibit signaling pathways that are often employed by GC cells. We will briefly describe the efforts that have targeted EGFR, angiogenesis and MET pathways.

2. EGFR-targeted therapy

EGFR gene is often amplified and its protein is overexpressed in upper gastrointestinal cancers. Overexpression is prognostic. With the advent of monoclonal antibodies and tyrosine kinase inhibitors (TKIs) against EGFR, many randomized clinical trials in patients with advanced or localized gastroesophageal cancers (squamous cell carcinoma (SCC) and adenocarcinoma) have been conducted; however, the results have been uniformly disappointing.

The most recent UK COG report on patients with Siewert type I/II advanced gastroesophageal cancers (adenocarcinoma or SCC) in the second-line setting Citation[3] randomized 449 patients to receive gefitinib, an anti-EGFR TKI or placebo. The primary end point was overall survival (OS). Secondary end points were progression-free survival (PFS) and quality-of-life outcomes. However, the median OS was 3.73 months for patients who received gefitinib and 3.63 months for those who received a placebo (HR = 0.9, 95% CI 0.74 – 1.09; p = 0.29). There was a minor prolongation of PFS by 0.4 months for patients who received gefitinib compared to those who received placebo (HR = 0.80, 95% CI 0.66 – 0.96; p = 0.02) (). Some recent data suggest that there may be a benefit in EGFR amplified patients; however, this needs further validation Citation[4].

Table 1. Major Phase III trials involving biologics in combination with chemotherapy in the advanced/metastatic gastric cancer setting.

Equally disappointing results were reported from two EGFR targeting trials (EXPAND and REAL-3), of patients with metastatic gastric or gastroesophageal cancer Citation[5,6]. The EXPAND trial randomized 904 patients to receive capecitabine and cisplatin, with or without cetuximab, a chimeric anti-EGFR mAb. This study did not achieve its primary end point, with the median PFS for capecitabine-cisplatin plus cetuximab being 4.4 months compared to 5.6 months for capecitabine-cisplatin alone (HR 1.09, 95% CI 0.92 – 1.29; p = 0.32) Citation[5]. The REAL-3 study was terminated prematurely because a statistically significantly lower OS was noted in patients who received epirubicin/oxaliplatin/capecitabine (EOC) and panitumumab, a fully human anti-EGFR mAb Citation[6]. Median OS of patients allocated to EOC was 11.3 months (95% CI 9.6 – 13.0) compared with 8.8 months (7.7 – 9.8) in 278 patients allocated to modified EOC and panitumumab (HR 1.37, 95% CI 1.07 – 1.76; p = 0.013). A molecular exploratory analysis of tumors of patients in the REAL-3 trial did not identify any predictive biomarkers for panitumumab Citation[7]. presents the major Phase III localized trials all of which were negative.

Table 2. Major Phase III trials involving biologics in combination with chemotherapy in the localized gastric cancer setting.

SCCs seem to overexpress EGFR at a higher frequency (60 – 70%) and have fairly high rate of EGFR amplification (28%) Citation[8]. These changes are associated with poor response to chemoradiotherapy and shorter OS Citation[9]. However, in the COG study, SCC patients formed a minority and there was a trend for improved OS for esophageal adenocarcinoma patients, highlighting the fact that overexpression of EGFR may not represent a therapeutic target. In GC, although EGFR amplification has been low, EGFR expression is similar to esophageal cancer and it is prognostic Citation[10].

3. VEGF-targeted therapy

Angiogenesis is recognized as a hallmark of several types of tumors, including gastric. VEGF is responsible for tumor-mediated angiogenesis, stimulating new blood vessel formation and higher levels of VEGF in tissues correlate with more advanced stage and poorer overall prognosis Citation[11]. Thus, efforts to block this pathway, either by inhibiting VEGF or its receptor, have emerged as attractive strategies for GC treatment.

Bevacizumab, the humanized mAb to VEGF, was investigated in locally advanced or metastatic GC in the AVAGAST trial Citation[12]. It was added to a combination of cisplatin and fluoropyrimidine. A total of 774 patients were randomized, and the median OS was 12.1 months with bevacizumab plus fluoropyrimidine-cisplatin and 10.1 months with placebo plus fluoropyrimidine-cisplatin (HR = 0.87, 95% CI: 0.73 – 1.03; p = 0.1002). A subsequent retrospective biomarker analysis of the AVAGAST trial showed that only Western patients with elevated baseline plasma VEGF-A levels and low baseline expression of neuropilin-1 seemed to have a statistically significant improvement of OS Citation[13]. It is important to note that neither of these biomarkers has been prospectively validated. Unlike the ToGA trial, the AVAGAST trial did not use an enriched patient population, underscoring the importance of appropriate patient population selection in randomized controlled trials and the use of predictive biomarkers to direct care.

Ramucirumab is a fully human IgG1 mAb receptor antagonist designed to bind the extracellular domain of VEGFR-2, thereby blocking the binding of VEGF ligands and inhibiting receptor activation, thus inhibiting angiogenesis Citation[14]. In the REGARD trial, 355 patients with advanced or metastatic GC that had progressed after first-line chemotherapy were randomized to receive ramucirumab or placebo Citation[15]. This study demonstrated a marginal improvement in median OS, 5.2 months in patients in the ramucirumab group and 3.8 months in those in the placebo group (HR = 0.776, 95% CI: 0.603 – 0.998; p = 0.047) with a disease control rate improved from 23 to 49% and very low toxicity – 8% grade ≥ 3 hypertension. In the recently published RAINBOW trial, ramucirumab was added to weekly paclitaxel as a second-line therapy in 665 patients with advanced or metastatic GC, demonstrating a significant improvement in both PFS and OS over paclitaxel alone, which was more impressive Citation[16]. A statistically significant prolongation of OS was demonstrated (HR = 0.81, 95% CI: 0.68 – 0.96, p = 0.017). Median OS was 9.6 and 7.4 months in the ramucirumab-plus-paclitaxel arm and placebo-plus-paclitaxel arm, respectively. This could imply that the use of ramucirumab mainly benefits patients when used in combination with paclitaxel and the combination has a modest safety profile.

Apatinib is a small-molecule multi-targeted TKI with activity against VEGFR-2 which was evaluated in a Phase III trial in 271 patients with advanced GC (after second line) Citation[17]. The median survival is 6.5 months for apatinib and 4.7 months for placebo (HR = 0.71, 95% CI: 0.54 – 0.94, p = 0.015) and the median PFS 2.6 months for apatinib and 1.8 months for placebo (HR = 0.44, 95% CI: 0.33 – 0.61, p < 0.0001). Currently, apatinib is only approved in China.

4. Anti-HER2 therapy

Trastuzumab has been a success in the first-line metastatic GC TOGA trial which demonstrated a benefit in OS in HER2+ metastatic gastric and GEJ patients treated with this antibody in addition to cisplatin and fluoropyrimidine Citation[18]. However, in the second-line setting targeted HER2 therapy with TKIs has been a failure.

Lapatinib, a HER1-2 TKI has been investigated in combination with capecitabine plus oxaliplatin in 545 HER2+ advanced or metastatic gastric and esophageal adenocarcinomas in the TRIO-013/LOGiC trial. Its addition to CapeOx did not improve efficacy (OS and PFS) among untreated HER2+ metastatic GC patients Citation[19]. In the second-line in a large 420 patient study (TyTAN Trial), HER2+ patients were randomized to lapatinib plus paclitaxel versus paclitaxel alone Citation[20]. Median OS was 11 months for the combination and 8.9 months for paclitaxel alone in the intent-to-treat (ITT) population (HR = 0.84; p = 0.2088). In a pre-planned subgroup analysis, median OS in HER2 immunohistochemistry (IHC) 3+ subgroup was 14 months for the combination therapy and 7.6 months for paclitaxel alone (HR = 0.59; p = 0.0176). Interestingly, it has recently been demonstrated that although the study mandated IHC HER2 positivity, 35% of patients in TyTAN had tumors classified as IHC0/1+. Identification of specific biomarkers for various patient subpopulations with advanced GC may help define those patients who would receive the most benefit from treatment.

5. MET-targeted therapy

Hepatocyte growth factor (HGF) and its receptor MET have been found to promote the proliferation, migration and survival of tumor cells and to play a role in GC. MET amplification and/or overexpression of its protein product have long been implicated in the pathogenesis of GC supporting its role as a poor prognostic factor Citation[21]. Rilotumumab (AMG 102), a fully human anti-HGF mAb targeting the c-MET ligand, demonstrated prolonged PFS for patients whose tumors had high total c-MET expression in a Phase Ib/II study but was associated with higher rates of grade 3/4 neutropenia and venous thromboembolism Citation[22]. Currently, two Phase III studies were testing the efficacy of rilotumumab in combination with epirubicin, cisplatin and capecitabine (RILOMET-1) and rilotumumab with cisplatin and capecitabine (RILOMET-2) as a first-line treatment of metastatic gastric and GEJ adenocarcinoma. However, the company recently terminated both its studies because they did not meet safety standards and the protocol-defined futility criteria would likely have been met at the planned interim analysis set for March 2015.

A recent Phase II trial testing the efficacy of onartuzumab, an anti c-MET mAb versus placebo combined with mFOLFOX6 in patients with metastatic HER2-negative and MET-positive GC, failed to show a significant difference in PFS – the primary end point – between the onartuzumab and placebo arms in either the ITT population or the subgroup of patients with MET-positive tumors Citation[23]. In contrast to the onartuzumab findings, the investigational oral MET TKI AMG 337 is generating excitement based on early-phase results in patients with gastrointestinal cancers. In a Phase I analysis of single-agent AMG 337 in 90 patients with advanced solid tumors, 13 individuals were found to have MET-amplified gastroesophageal adenocarcinomas. Eight of 13 patients had partial to near-complete responses to the small-molecule inhibitor AMG-337 Citation[24]. On the basis of these results, a Phase II study of AMG-337 in patients with MET-amplified gastroesophageal cancer or other MET-amplified solid tumors is currently recruiting participants.

6. Expert opinion

Establishing valid biomarkers in the clinic and then targeting them for therapeutic advantage is challenging. Most of the time, such efforts have failed. The sheer complexity of the genome is staggering, and structural alterations do not necessarily translate into functional/protein aberrations. For advancing research in GC, the clinical trial machinery has been well established. Many pivotal trials are being conducted; therefore, we can anticipate some advances in the near future. Much work remains to be done. We must spend our resources to establish reliable preclinical models that will assure success in the clinic. We must also delve deep in uncovering true drivers of GC in individual patients. One of the most challenging and exciting frontiers is the potential of host’s immune system. We should harness the power of the immune system, either through vaccines, antibodies, cell therapy and/or through programmed cell death inhibitors.

Acknowledgements

N Charalampakis has been awarded a scholarship from the Hellenic Society of Medical Oncology. N Charalampakis and E Elimova contributed equally to this work. Both these authors are co-first authors.

Declaration of interest

The authors have received generous grants from the Caporella, Dallas, Sultan, Park, Smith, Frazier, Oaks, Vanstekelenberg, Planjery, and Cantu Families. From the Schecter Private Foundation, Rivercreek Foundation, Kevin Fund, Myer Fund, Dio Fund, Milrod Fund, and Multidisciplinary Grants from the University of Texas M. D. Anderson Cancer Center, Houston, USA. The authors are supported in part by the National Cancer Institute awards CA138671, CA172741, CA129926 (JAA) and P30CA016672 and used the Biostatistics Resource Group (RS, H-CC). J Ajani has been part of the advisory board and contracted research for Amgen, Novartis and Lilly. He has acted as advisory committee for Celgene, he has also been involved in contracted research for Genetech, Roche and Taiho, as well as consulting agreements for Lilly. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Bibliography

  • Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136(5):E359-86
  • Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513(7517):202-9
  • Dutton SJ, Ferry DR, Blazeby JM, et al. Gefitinib for oesophageal cancer progressing after chemotherapy (COG): a phase 3, multicentre, double-blind, placebo-controlled randomised trial. Lancet Oncol 2014;15(8):894-904
  • Gefitinib extends survival in some esophageal cancers. Cancer Discov 2015;5(1):OF4
  • 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(6):490-9
  • 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(6):481-9
  • Okines AF, Gonzalez de Castro D, Cunningham D, et al. Biomarker analysis in oesophagogastric cancer: results from the REAL3 and TransMAGIC trials. Eur J Cancer 2013;49(9):2116-25
  • Hanawa M, Suzuki S, Dobashi Y, et al. EGFR protein overexpression and gene amplification in squamous cell carcinomas of the esophagus. Int J Cancer 2006;118(5):1173-80
  • 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(4):658-67
  • 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(7):1458-68
  • 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(1):69-79
  • 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(30):3968-76
  • Van Cutsem E, de Haas S, Kang YK, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol 2012;30(17):2119-27
  • Spratlin JL, Cohen RB, Eadens M, et al. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol 2010;28(5):780-7
  • 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(9911):31-9
  • Wilke H, Muro K, Van Cutsem E, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 2014;15(11):1224-35
  • Qin S. Phase III study of apatinib in advanced gastric cancer: a randomized, double-blind, placebo-controlled trial. ASCO Meeting Abstracts 32(15 suppl):4003
  • 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(9742):687-97
  • 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. ASCO Meeting Abstracts 2013;31(18 suppl):LBA4001
  • 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(19):2039-49
  • Lee HE, Kim MA, Lee HS, et al. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer 2012;107(2):325-33
  • 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(9):1007-18
  • Manish A, Shah JY. Iain Tan Bee Huat, et al. Randomized phase II study of FOLFOX +/- MET inhibitor, onartuzumab (O), in advanced gastroesophageal adenocarcinoma (GEC). GI ASCO; 2015; San Francisco. J Clin Oncol 2015;33(suppl 3; abstr 2):2015
  • Eunice Lee Kwak PL, Hamid O, Janku F, et al. Clinical activity of AMG 337, an oral MET kinase inhibitor, in adult patients (pts) with MET-amplified gastroesophageal junction (GEJ), gastric (G), or esophageal (E) cancer. GI ASCO; 2015; San Francisco [abstract 1]. J Clin Oncol 2015;33(Suppl 3):2015
  • Ohtsu A, Ajani JA, Bai YX, et al. Everolimus for previously treated advanced gastric cancer: results of the randomized, double-blind, phase III GRANITE-1 study. J Clin Oncol 2013;31(31):3935-43
  • Suntharalingam M, Winter K, Ilson DH, et al. The initial report of RTOG 0436: a phase III trial evaluating the addition of cetuximab to paclitaxel, cisplatin, and radiation for patients with esophageal cancer treated without surgery. ASCO Meeting Abstracts 32(3(suppl):LBA6
  • Crosby T, Hurt CN, Falk S, et al. Chemoradiotherapy with or without cetuximab in patients with oesophageal cancer (SCOPE1): a multicentre, phase 2/3 randomised trial. Lancet Oncol 2013;14(7):627-37
  • Okines AF, Langley RE, Thompson LC, et al. Bevacizumab with peri-operative epirubicin, cisplatin and capecitabine (ECX) in localised gastro-oesophageal adenocarcinoma: a safety report. Ann Oncol 2013;24(3):702-9

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.