Skip to main content
F1000Research logoLink to F1000Research
. 2018 Aug 30;7:F1000 Faculty Rev-1365. [Version 1] doi: 10.12688/f1000research.15133.1

Recent advances in the management of gastric adenocarcinoma patients

Kazuto Harada 1,2, Anthony Lopez 1,3, Namita Shanbhag 1, Brian Badgwell 4, Hideo Baba 2, Jaffer Ajani 1,a
PMCID: PMC6117861  PMID: 30228868

Abstract

Gastric adenocarcinoma (GAC) is one of the most aggressive malignancies and has a dismal prognosis. Therefore, multimodality therapies to include surgery, chemotherapy, targeted therapy, immunotherapy, and radiation therapy are needed to provide advantage. For locally advanced GAC (>cT1B), the emerging strategies have included preoperative chemotherapy, postoperative adjuvant chemotherapy, and (occasionally) postoperative chemoradiation in various regions. Several novel therapies have been assessed in clinical trials, but only trastuzumab and ramucirumab (alone and in combination with paclitaxel) have shown overall survival advantage. Pembrolizumab has been approved by the US Food and Drug Administration on the basis of response rate only for patients with microsatellite instability (MSI-H) or if PD-L1 expression is positive (≥1% labeling index in tumor/immune cells in the presence of at least 100 tumor cells in the specimen). Nivolumab has been approved in Japan on the basis of a randomized trial showing significant survival advantage for patients who received nivolumab compared with placebo in the third or later lines of therapy. The cure rate of patients with localized GAC in the West is only about 40% and that for metastatic cancer is very poor (only 2–3%). At this stage, much more target discovery is needed through molecular profiling. Personalized therapy of patients with GAC remains a challenge.

Keywords: Gastric adenocarcinoma, preoperative therapy, targeted therapy

Introduction

Gastric adenocarcinoma (GAC) is considered the fifth most common cancer in the world (1,313,000 cases) and the third leading cause of cancer death globally (819,000 deaths) 1. Its incidence varies according to the region: high incidence in East Asia and South America but low incidence in the West 1. In East Asia, especially Japan and Korea, the incidence of distal GAC is high, whereas proximal GAC has a higher incidence in the West 2. GAC located in the cardia or the gastroesophageal junction (or both) has dramatically increased in incidence in the USA 3. This trend of proximal migration of GAC is also being observed in Asia and South America along with Europe. Based on American Joint Committee on Cancer 8 (AJCC 8), gastroesophageal junction adenocarcinoma that has its epicenter in the proximal 2 to 5 cm of the stomach (Siewert type III) should be staged and treated as GAC. For locally advanced GAC, various strategies have been developed in different regions of the world and these have evolved on the basis of practice preferences and types of clinical trials performed. For example, preoperative chemotherapy is favored in the European Union and the USA, whereas postoperative adjuvant chemotherapy is preferred in Asia. Postoperative chemoradiation has a diminishing role while the quality of surgery appears to be improving. Adjunctive therapy (preoperative or postoperative) seems to increase the cure rate by about 10%. Multidisciplinary evaluation is essential to improve patient outcomes and for the initial treatment decision process.

For metastatic GAC, the standard-of-care therapies have a limited impact on patient outcome. The median survival of patients with advanced GAC is less than 12 months. Only a limited number of therapies are approved, and many of these therapies are done empirically. Here, we summarize recent advances in the management of GAC.

Molecular features of gastric adenocarcinoma

Two groups—The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group (ACRG)—have reported multiplatform sequencing of primary GACs (TCGA analysis being more comprehensive than ACRG) and, out of these efforts, four genotypes of GAC have emerged 4, 5. TCGA data were more comprehensive and basically GACs have been divided into those with microsatellite instability (MSI), chromosomal instability (CIN), genome stability (GS), and Epstein–Barr virus (EBV) association 5. The MSI cohort represented about 22% and was more frequent in the distal GAC than the proximal GAC. In contrast, CIN is more frequent in the proximal GAC. Compared with other gastrointestinal adenocarcinomas, CIN in GAC tends to have focal region alterations 6. GS and EBV had frequencies of 20% and 9%, respectively.

Certain molecular subtypes are associated with shorter survival; for example, GS and CIN have poor prognosis 4, 7. The GS subtype of GAC is enriched in diffuse-type histology and is molecularly characterized by less mutation and overexpression of epithelial–mesenchymal transition-related genes 4, 5. On the other hand, the CIN subtype of GAC is enriched in intestinal histology and is molecularly characterized by TP53 mutation and RTK-RAS activation/amplifications 4, 5. MSI subtyped GAC harbors numerous somatic mutations, leading to a large number of neoantigens that can activate T cells 8. Thus, GACs with MSI respond well to immune checkpoint blockade 9. However, the frequency of MSI-H and EBV-related GACs in the metastatic setting is low (<3%).

Resectable gastric adenocarcinoma

Primary resection

GAC with clinical T1N0 can be treated by either endoscopic therapy or surgery without any adjunctive therapy, while advanced localized GAC should undergo either preoperative therapy or surgery first followed by adjuvant chemotherapy. Adequate lymphadenectomy (D2 dissection) is desired with gastrectomy 10. If the depth of invasion suggests eusT1a, then endoscopic resection is preferred according to the Japanese guidelines 10. When surgery is performed first, postoperative adjuvant chemotherapy should be considered on the basis of the pathological stage or quality of surgery.

Postoperative treatment

Postoperative adjuvant chemotherapy is the most common strategy in East Asia. The ACTS-GC trial, a phase III trial in Japan, showed that postoperative adjuvant therapy with S-1 for 12 months improved overall survival (OS) (5-year OS 72% versus 61%, hazard rate [HR] 0.67, 95% confidence interval [CI] 0.52–0.82) and relapse-free survival (RFS) (5-year RFS 65% versus 53%, HR 0.65, 95% CI 0.54–0.79) in GAC patients with stage II/III who underwent D2 gastrectomy 11. The CLASSIC trial, a phase III trial performed in South Korea, China, and Taiwan, showed that adjuvant capecitabine plus oxaliplatin combination given for 6 months after D2 gastrectomy improved OS (5-year OS 78% versus 69%, HR 0.66, 95% CI 0.51–0.85) and RFS (5-year RFS 68% versus 53%, HR 0.58, 95% CI 0.47–0.72) 12. These trials are the basis for recommending postoperative chemotherapy after optimal surgery. More intensive regimens have no advantage for postoperative chemotherapy 13, 14. Recently, the JACCRO GC-07 trial suggests that S-1 plus docetaxel has significant advantage over S-1 alone and it has become the standard of care 15.

Efficacy of postoperative chemoradiation was shown in the INT-0116 trial; compared with the surgery-alone group, the postoperative chemoradiotherapy group had longer OS (median OS 27 versus 36 months, HR 1.35, 95% CI 1.09–1.66) and RFS (median RFS 19 versus 30 months, HR 1.52, 95% CI 1.23–1.86) 16, 17. However, the quality of surgery was suboptimum in patients enrolled in this trial; D0, D1, and D2 lymph node dissection rates were 54%, 36%, and 10%, respectively. Importantly, the ARTIST and CRITICS trials demonstrated the lack of efficacy of postoperative chemoradiation after D2 or D1+ nodal dissection 18, 19. Therefore, postoperative chemoradiation is not useful if optimal or near-optimal surgery is performed.

Preoperative treatment

The MAGIC trial provided evidence that perioperative chemotherapy (three preoperative and three postoperative cycles of epirubicin, cisplatin, and fluorouracil [ECF]) for resectable GAC could improve cure rates (5-year OS 23% versus 36%, HR 0.75, 95% CI 0.60–0.93) 20. However, a follow-up analysis of the MAGIC results has not yet been presented. The results overall were suboptimal. Moreover, epirubicin is not necessary 21. The FNCLCC/FFCD trial showed that surgery plus preoperative cisplatin and fluorouracil (FP) improved OS compared with surgery alone (5-year OS 24% versus 38%, HR 0.69, 95% CI 0.50–0.95) 22. Recently, the MRC-OEO5 trial compared two cycles of FP and four cycles of ECF as perioperative chemotherapy and showed that the two regimens had similar OS (3-year rate 42% versus 39%, HR 0.92, 95% CI 0.79–1.08) 23. These results suggest that the addition of epirubicin and longer duration of chemotherapy do not provide any advantage.

The FLOT4 trial compared perioperative (predominantly preoperative) chemotherapy with docetaxel, oxaliplatin, and fluorouracil/leucovorin (FLOT) and ECF/ECX 24, 25. A total of 716 patients were randomly assigned to the ECF/ECX group (n = 360) or the FLOT group (n = 356). FLOT improved median RFS (30 versus 18 months, HR 0.75, p = 0.001) and median OS (50 versus 35 months, HR 0.77, p = 0.012) compared with ECF/ECX. A total of 50% of patients in the FLOT group completed the planned postoperative treatments, while 37% of patients in ECF/ECX completed them. Perioperative complications were similar across the two groups and unacceptably high 24, 25. Grade 3–4 diarrhea, infections, sensory disorder, and neutropenia were more frequent in the FLOT group, while vomiting, nausea, thromboembolism, and anemia were more frequent in the ECF/ECX group. FLOT should not be recommended to every patient. FLOT might be more suitable for only very fit patients with GAC rather than patients with esophageal or gastroesophageal junction adenocarcinoma where chemoradiation followed by surgery is the preferred strategy. FLOT resulted in 90-day mortality of 5% and grade 3–4 diarrhea (10%) and infections (18%). The follow-up in this trial is also short, and it is likely that, with further follow-up, the differences between the two regimens will narrow. If the 5-year OS rate of FLOT is about 40%, then it would not be a major advance.

The efficacy of targeted therapy for perioperative treatment was assessed. The ST03 trial evaluated whether adding bevacizumab to perioperative chemotherapy could improve survival 26. A total of 1,063 patients were randomly assigned to receive chemotherapy alone (n = 533) or chemotherapy plus bevacizumab (n = 530), and prognosis was similar in the two groups (3-year OS 50.3% versus 48.1%, HR 1.08, 95% CI 0.91–1.29, p = 0.36) 26. Moreover, anastomotic leak was more frequent in the chemotherapy-plus-bevacizumab group 26. To date, there is no place for targeted therapy in the preoperative setting. PETRARCA/FLOT6 (ClinicalTrials.gov Identifier: NCT02581462) is evaluating the efficacy of adding herceptin/pertuzumab to perioperative chemotherapy for human epidermal growth factor receptor 2 (HER2)-positive GAC. RAMSES/FLOT7 (ClinicalTrials.gov Identifier: NCT02661971) is evaluating the efficacy of adding ramucirumab (VEGFR2 monoclonal antibody) for HER2-negative GAC.

For esophageal or gastroesophageal junction adenocarcinoma, the CROSS trial documented benefit of preoperative chemoradiation 27. For GAC, preoperative chemoradiation is an attractive but non-standard option. Several trials have evaluated the efficacy of preoperative chemoradiation 2831, and a retrospective study at the M.D. Anderson Cancer Center showed excellent prognosis of GAC patients who had preoperative chemoradiation 32. To date, phase III trials assessing the value of preoperative chemoradiation in GAC are ongoing. The TOPGEAR trial is assessing the efficacy of adding preoperative radiation to the MAGIC trial regimen 33. The CRITICS-II trial is comparing three arms: preoperative chemotherapy followed by surgery, preoperative chemotherapy and subsequent chemoradiation followed by surgery, and preoperative chemoradiation followed by surgery (ClinicalTrials.gov Identifier: NCT02931890). Result of these trials are expected.

Standard treatment for metastatic patients

Standard first-line therapy

The recommended first-line therapy for patients with HER2-negative GAC is a two-drug combination of oxaliplatin (preferred) or cisplatin plus 5-FU or capecitabine. For HER2-positive GAC, the ToGA study showed that trastuzumab should be added to the first-line cytotoxic therapy 34. Irinotecan or taxane should be considered when platinum-based chemotherapy cannot be tolerated in the first-line setting 3537. DCF (docetaxel, cisplatin, and 5-FU) provides marginal OS advantage but its toxicity is significant 38, 39. Therefore, a modified regimen has been evaluated 4043. Modified DCF is still one of the options in selected cases, but the routine use of a taxane in the first-line setting should be avoided. Also, ECF is not recommended for metastatic GAC 44.

Standard second-/third-line therapy

In the second-line therapy setting, ramucirumab is the only molecular-targeted drug with a confirmed but marginal survival benefit (as a single agent) in a global phase III trial. The RAINBOW study showed that OS as a result of ramucirumab plus paclitaxel was significantly longer than in the placebo-plus-paclitaxel group (median OS 9.6 versus 7.4 months) and thus this regimen is the preferred choice for second-line therapy 45. Ramucirumab monotherapy is not recommended 46, 47. Docetaxel, irinotecan, and paclitaxel have significantly prolonged OS compared with best supportive care (BSC) 4850. However, the data on these molecules are based on a few small trials.

Treatment for peritoneal metastatic gastric adenocarcinoma

There is no established therapy for peritoneal carcinomatosis, which is a common site of metastases in patients with advanced GAC. Systemic chemotherapy or BSC is often recommended 28. Recently, intraperitoneal chemotherapy or hyperthermic intraperitoneal chemoperfusion (HIPEC) has been assessed as a potential therapy for peritoneal metastases. PHOENIX-GC, a Japanese phase III trial, compared S-1 in combination with intravenous/intraperitoneal paclitaxel and S-1 in combination with intravenous cisplatin (standard therapy in Japan) 51. Primary results showed that there was no significant difference between the two groups (median OS 17.7 and 15.2 months), suggesting lack of superiority of the intraperitoneal chemotherapy 51. A subgroup appeared to benefit, but this would only be hypothesis generating 52, 53. Also, perioperative intraperitoneal chemotherapy is being assessed 54, 55. The CY-PHOENIX trial showed that intraperitoneal chemotherapy for CY-positive patients (no other site of metastasis) resulted in conversion to CY-negative in 36 patients (94.7%) and 84.2% of the 1-year OS rate 55. Neoadjuvant laparoscopic HIPEC (mitomycin C 30 mg and cisplatin 200 mg) was assessed in a phase II study, which showed that seven patients (37%) had negative peritoneal cytology after HIPEC 56.

Molecularly targeted drug for metastatic gastric adenocarcinoma

HER2

HER2 is the only molecularly targeted drug accepted in first-line therapy. The ToGA study showed that adding trastuzumab in chemotherapy for patients with HER2-positive GAC resulted in longer median OS (13.8 versus 11.1 months, HR 0.74, 95% CI 0.60–0.91) 34. However, the subsequent analysis of the ToGA study showed that all of these differences have decreased by 40%. HER2 status should be tested according to guidance in all candidates for HER2-targeted therapy 57. The randomized phase III GATSBY trial compared T-DM1, conjugated with trastuzumab and emtansine, and taxane in patients with HER2-positive GAC in a second-line setting 58. However, T-DM1 failed to show benefit in OS (median OS 7.9 versus 8.6 months, HR 1.15, 95% CI 0.87–1.51) 58.

TRIO-013/LOGiC, a randomized phase III trial, assessed lapatinib, a dual inhibitor of HER2 and epidermal growth factor receptor (EGFR) 59. However, the addition of lapatinib to chemotherapy did not show benefit in OS for patients with HER2-positive GAC in the first-line setting (median OS 12.2 versus 10.5 months, HR 0.91, 95% CI 0.73–1.12) 59. TyTAN, a randomized phase III study, also showed that lapatinib was not effective for patients with HER2-positive GAC in the second-line setting (median OS 11.0 versus 8.9 months, HR 0.84, 95% CI 0.64–1.11) 60.

EGFR

EGFR-targeted therapies showed no benefit in any trials for GAC. Two trials—the EXPAND trial and the REAL3 study—failed to show a benefit from the addition of EGFR inhibitor to first-line chemotherapy 61, 62. To date, EGFR inhibitor is evaluated in selected patients who have EGFR overexpression with immunohistochemistry (IHC) 2+ or 3+ (ClinicalTrials.gov Identifier: NCT01813253).

VEGFR

Ramucirumab is marginally effective for second-line therapy based on REGARD and performs somewhat better, as shown in the RAINBOW study 45, 46. The RAINFALL trial assessed whether adding ramucirumab to first-line standard chemotherapy could be effective 63. A total of 645 patients were randomly assigned to two groups—ramucirumab plus chemotherapy (n = 326) or placebo plus chemotherapy (n = 319)—and adding ramucirumab marginally prolonged progression-free survival (PFS) (median PFS 5.7 versus 5.4 months, HR 0.75, 95% CI 0.61–0.94) but not OS (median OS 11.2 versus 10.7 months, HR 0.96, 95% CI 0.80–1.16) 63. These results strongly suggest that ramucirumab should not be used as first-line therapy. Bevacizumab, in the first-line setting, was found to have no efficacy when added to chemotherapy in the AVAGAST study or perioperative chemotherapy in the STO3 study 26, 64.

MET

The phase III RILOMET-1 trial compared ECX with and without MET-positive gastroesophageal adenocarcinoma, but this trial was stopped early because of a higher number of deaths in the rilotumumab group; the median OS was 8.8 months in the rilotumumab group (n = 304) compared with 10.7 months in the placebo group (n = 304) 65. The MET gastric trial assessed adding onartuzumab to FOLFOX in HER2-negative GAC but did not significantly benefit OS (median OS 11.0 versus 11.3 months, HR 0.82, 95% CI 0.59–1.15) 66. These results suggest that MET inhibitors are not particularly efficacious in most patients with MET-positive GAC.

mTOR

The GRANITE-1 study compared everolimus, an inhibitor of mammalian target of rapamycin (mTOR), and placebo in patients with GAC previously treated with standard care 67. Unfortunately, everolimus showed no benefit for OS (median OS 5.9 versus 4.3 months, HR 0.90, 95% CI 0.75–1.08) 67. The PADPAC study assessed adding everolimus to paclitaxel in the second-line setting but did not show significant benefit; the median OS was 6.1 months in the everolimus group (n = 150) compared with 5.1 months in the placebo group (n = 150) 68.

FGFR2

Fibroblast growth factor receptor (FGFR) inhibitor failed to show a benefit in a phase II trial, the SHINE study 69. GAC patients with FGFR2 amplification were randomly assigned to an AZD4547 (an FGFR inhibitor) group (n = 41) or a paclitaxel group (n = 30) and showed similar median PFS: 1.8 months in the AZD4547 group and 3.5 months in the paclitaxel group 69.

PARP

Poly (ADP-ribose) polymerase (PARP) inhibitor is potentially useful in cancers with a deficiency in the repair of double-strand breaks 70. In patients with GAC, a phase II trial showed that lower/absent expression level of ATM, which has a key role in activating DNA damage response to double-strand breaks, seemed to benefit from PARP inhibitor 71. However, the phase III GOLD study did not show an advantage from the addition of olaparib 72. A total of 643 patients were assigned to the olaparib-plus-paclitaxel arm (n = 263) or the placebo-plus-paclitaxel arm (n = 262), and the median OS values in the olaparib and placebo groups were 8.8 and 6.9 months, respectively (not significant) 72. Even in the ATM-negative population, olaparib was not effective 72.

Targeted therapies against stem cell pathway

Cancer stem cells are often resistant to therapy that is non-specific and thus can be targeted to overcome resistance 73. Thus far, few clinical trials have assessed the effect of inhibiting stemness-related pathways, such as the Hedgehog and signal transducer and activator of transcription 3 (STAT3) pathway. Vismodegib, an inhibitor of Hedgehog signal by binding smoothened (SMO), was assessed in a phase II randomized study, but adding vismodegib to FOLFOX did not prolong PFS (11.5 versus 9.3 months, p = 0.34) 74. Moreover, the BRIGHTER study showed that adding napabucasin, a STAT3 inhibitor, to paclitaxel did not prolong OS 75. The trial did not enrich patients by expression of stem cell markers in the tumor cells. It may be that selected patients should be assessed 76. STAT3 inhibitors may be better suited for immune modulation as well.

Other targets

The isoform 2 of the tight junction molecule claudin-18 (CLDN18.2) is an important component of the tight cell junctions and overexpressed significantly in GAC 77. IMAB362, an inhibitor of CLDN18.2, was assessed in clinical trials. In a randomized phase II trial, IMAB362 plus epirubicin, oxaliplatin, and capecitabine (EOX) significantly prolonged PFS (median 5.7 versus 7.9 months, HR 0.5, 95% CI 0.35–0.78) 78. Moreover, the benefit of IMAB362 was more pronounced in the patients with CLDN18.2 high-expression (70% labeling index) GAC (PFS 6.1 versus 9.1 months, HR 0.46; OS 9.3 versus 16.6 months, HR 0.44) 78. A phase III trial is ongoing.

Matrix metalloproteinase 9 (MMP9) has a key role in extracellular matrix remodeling and angiogenesis, and its inhibition in combination with FOLFOX led to potential improvement of prognosis in a phase I study 79. A phase III trial assessing potential benefit of MMP9 inhibition has completed accrual (ClinicalTrials.gov Identifier: NCT02545504) 80.

Immunotherapy

Immunotherapy checkpoint therapy has dramatically improved the prognosis of patients with metastatic melanoma or non-small cell lung cancer 81, 82, and the concept/approach has been adopted in many tumor types, including gastrointestinal malignancies. Cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and programmed death protein 1 (PD-1) and its ligand (PD-L1) are the key proteins that have the capacity to inhibit the responses of T cells that can be tumor promoting. A few phase III trials assessing the efficacy of immune checkpoint inhibitors in gastrointestinal malignancies have been conducted in patients with advanced gastric or gastroesophageal junction cancer 83. After two or more prior therapy failures, 493 Asian patients were randomly assigned to receive nivolumab, a PD-1 antibody, at 3 mg/kg (n = 330) or placebo (n = 163) every 2 weeks. The median OS was 5.3 months in the nivolumab group versus 4.1 months in the control group ( p <0.0001). The median PFS was also slightly longer with nivolumab compared with placebo (HR 0.60, 95% CI 0.49–0.75). The safety profile was excellent; the grade 3 or 4 treatment-related adverse event rate was 10% in the nivolumab group. The combination of different immune checkpoint inhibitors may also be promising.

Ipilimumab is an anti-CTLA4 antibody, tested in the phase I/II study, in combination with nivolumab 84. Patients (n = 160) were randomly assigned to one of three groups: nivolumab alone (3 mg/kg), nivolumab plus ipilimumab (1 and 3 mg/kg, respectively), and nivolumab plus ipilimumab (3 and 1 mg/kg, respectively). Overall response rates (ORRs) were 12%, 24%, and 8% in the first, second, and third groups, respectively, and seemed better in the case of PD-L1 expression. Corresponding median OS values were 6.2, 6.9, and 4.8 months (not reached in PD-L1 + subgroups). Toxicity was increased with combination therapy (47% of grade 3 or 4 adverse events and treatment discontinuation due to toxicity in 20% of the cases in the second group). A phase III trial is ongoing.

Pembrolizumab is another anti-PD-1 antibody, currently tested alone or in combination in three different cohorts in the phase II KEYNOTE-059 study. In cohort 1, patients with advanced gastric and gastroesophageal junction cancer who were progressive after two or more chemotherapy lines received pembrolizumab monotherapy at 200 mg every 3 weeks 85. In total, 259 patients were included (52% in third-line and 48% in fourth-line or more). ORR, the primary endpoint, was 11.6%; interestingly, durable responses were observed in patients with PD-L1-positive cancer (15.5% in PD-L1 positive and 6.4% in PD-L1 negative). Based on these results, pembrolizumab has been approved by the US Food and Drug Administration (FDA) for patients with GAC positive for PD-L1 expression (≥1% labeling index in tumor/immune cells in the presence of at least 100 tumor cells in the specimen) to be used in the third (or later) line. Cohort 2 included treatment-naïve patients who received a combination of pembrolizumab (200 mg/3 weeks) and chemotherapy (cisplatin plus 5-FU or capecitabine). Preliminary results in 25 patients were promising, showing an ORR of 60% and a median PFS of 6.6 months 86. These results suggest that anti-PD1 antibodies can potentiate conventional cytotoxic chemotherapy in the first-line setting. The results from the phase III trial are expected (ClinicalTrials.gov Identifier: NCT02494583) 87.

Avelumab is an anti-PD-L1 antibody and was evaluated in the phase Ib JAVELIN trial as a first-line maintenance (group 1) or second-line (group 2) therapy 88. ORRs were 9.0% and 9.7% in groups 1 and 2, respectively. The corresponding median PFS values were 12.0 and 6.0 weeks. Two phase III trials are ongoing with avelumab in metastatic GAC or gastroesophageal junction adenocarcinoma. The JAVELIN Gastric 300 evaluated avelumab as a third-line treatment, and 371 patients were randomly assigned to receive avelumab plus BSC or chemotherapy (paclitaxel or irinotecan) plus BSC (ClinicalTrials.gov Identifier: NCT02625623). This trial failed to meet its primary outcome of superior survival, and detailed results are awaited. The JAVELIN Gastric 100 has been evaluating avelumab as a maintenance therapy (ClinicalTrials.gov Identifier: NCT02625610). This trial has completed accrual and results are awaited.

The modest efficacy and high cost of immune checkpoint inhibitors suggest that we need a reliable predictive/prognostic biomarker to select these agents. In studies described above, ORR was two to three times higher in the case of PD-L1 positivity in tumor cells. However, results are conflicting, and there is no consensus on the best way to assess PD-L1 status. MSI refers to the replicative error phenotype caused by mutations in the mismatch repair (MMR) system. Recently, Le et al. reported an ORR of 53% in 86 patients with MSI-H tumors, of whom 76% had gastrointestinal cancers 89. After a 2-year follow-up, 53% of the patients had not had tumor progression and 64% were still alive (medians not reached). In the KEYNOTE-059 study (cohort 1), the ORR was 57% in patients with MSI tumors compared with 9% in the case of MSS tumors 85. Based on five trials (KEYNOTE-016, KEYNOTE-164, KEYNOTE-012, KEYNOTE-028, and KEYNOTE-158), pembrolizumab has been approved by the FDA for MSI-H or deficient MMR solid tumors. In preliminary studies, a new generation of immunotherapy drugs is being evaluated for downregulating immunosuppressive pathways (for example, VISTA, an immunosuppressive molecule expressed on regulatory T cells, and indoleamine 2,3-dioxygenase (IDO), an enzyme leading to decreased tryptophan level which then suppresses T-cell proliferation) or stimulating immune tumor response (for example, inducible T-cell co-stimulator [ICOS] agonists) 90.

Conclusions

In summary, in addition to high-quality surgery, preoperative or postoperative chemotherapy is recommended for localized advanced GAC, depending on local preferences ( Table 1). Preoperative chemoradiation is a non-standard option with potential and is being studied in phase III trials. Only trastuzumab and ramucirumab (in combination) are effective targeted drugs for GAC ( Table 2). PD-1 inhibitor is highly active against MSI-H GACs. Clearly, more basic research is needed to identify novel targets and drugs.

Table 1. Preoperative treatment trial for localized gastric adenocarcinoma.

Study Number Treatment Survival HR
(95% CI)
p value Reference
ACTS-GC n = 529
n = 530
Surgery → S-1
Surgery
5-year OS: 72%
5-year OS: 61%
0.67
(0.54–0.82)
- 11
CLASSIC n = 520
n = 515
Surgery → XP
Surgery
5-year OS: 78%
5-year OS: 69%
0.66
(0.51–0.85)
0.0015 12
ITACA-S n = 562
n = 538
Surgery → FOLFIRI → DP
Surgery → 5-FU/LV
5-year OS: 51%
5-year OS: 51%
0.98
(0.82–1.18)
0.87 13
INT-0116 n = 281
n = 275
Surgery → 5-FU/45 Gy
Surgery
Median OS: 36 months
Median OS: 27 months
1.35
(1.09–1.66)
0.005 16
ARTIST n = 228
n = 230
Surgery → XP
Surgery → XP/45 Gy
3-year DFS: 74%
3-year DFS: 78%
- 0.86 18
CRITICS n = 393
n = 395
ECC → Surgery → ECC
ECC → Surgery → ECC/45 Gy
5-year OS: 41%
5-year OS: 41%
- 0.99 19
FNCLCC/FFCD n = 113
n = 111
CF → Surgery (n = 113)
Surgery (n = 111)
5-year OS: 38%
5-year OS: 24%
0.69
(0.50–0.95)
0.02 22
MAGIC n = 250
n = 253
ECF → Surgery → ECF
Surgery
5-year OS: 36%
5-year OS: 23%
0.75
(0.60–0.93)
0.009 20
MRC
OEO-5
n = 446
n = 451
ECF → Surgery
CF → Surgery
3-year OS: 39%
3-year OS: 42%
0.92
(0.79–1.08)
0.30 23
FLOT4 n = 360
n = 356
ECF → Surgery → ECF
FLOT → Surgery → FLOT
3-year OS: 48%
3-year OS: 57%
0.77
(0.63–0.94)
0.012 25
ST03 n = 533
n = 530
ECF → Surgery → ECF
ECF+Bev → Surgery → ECF+Bev
3-year OS: 50%
3-year OS: 48%
1.08
(0.91–1.29)
0.36 26

5-FU, 5-fluorouracil; Bev, bevacizumab; CF, cisplatin and 5-fluorouracil; CI, confidence interval; ECC, epirubicin, cisplatin, and capecitabine; ECF, epirubicin, cisplatin, and 5-fluorouracil; FLOT, docetaxel, oxaliplatin, leucovorin, and 5-fluorouracil; Gy, Gray; HR, hazard rate; OS, overall survival; XP, cisplatin and capecitabine.

Table 2. Targeted therapy trial for metastatic gastric adenocarcinoma.

Study Number Target Treatment Survival HR
(95% CI)
Reference
First-line setting
ToGA n = 298
n = 296
HER2 Trastuzumab + XP
Placebo + XP
mOS: 13.8 months
mOS: 11.1 months
0.74
(0.60–0.91)
34
TRIO-013/LOGiC n = 249
n = 238
HER2/EGFR Lapatinib + CapeOx
Placebo + CapeOx
mOS: 12.2 months
mOS: 10.5 months
0.91
(0.73–1.12)
59
EXPAND n = 455
n = 449
EGFR Cetuximab + XP
Placebo + XP
mOS: 9.4 months
mOS: 10.7 months
1.00
(0.87–1.17)
61
REAL3 n = 278
n = 275
EGFR Panitumumab + EOC
Placebo + EOC
mOS: 8.8 months
mOS: 11.3 months
1.37
(1.07–1.76)
62
AVAGAST n = 387
n = 387
VEGF Bevacizumab + FP
Placebo + FP
mOS: 12.1 months
mOS: 10.1 months
0.87
(0.73–1.03)
64
RAINFALL n = 326
n = 319
VEGFR Ramucirumab + Cape/Cis
Placebo + Cape/Cis
mOS: 11.2 months
mOS: 10.7 months
0.68
(0.80–1.16)
63
RILOMET-1 n = 304
n = 305
MET Rilotumumab + ECX
Placebo + ECX
mOS: 8.8 months
mOS: 10.7 months
1.34
(1.10–1.63)
65
METGastric n = 279
n = 283
MET Onartuzumab + FOLFOX
Placebo + FOLFOX
mOS: 11.0 months
mOS: 11.3 months
0.82
(0.59–1.15)
66
Cohen et al. n = 60
n = 63
Hedgehog
pathway
Vismodegib + FOLFOX
Placebo + FOLFOX6
mOS: 11.5 months
mOS: 14.9 months
- 74
FAST n =84
n =77
Claudin18.2 IMAB362 + EOX
Placebo + EOX
mOS: 13.2 months
mOS: 8.4 months
0.51
(0.36–0.73)
78
Beyond second-line setting
REGARD n = 238
n = 117
VEGFR Ramucirumab
Placebo
mOS: 5.2 months
mOS: 3.8 months
0.78
(0.60–0.99)
46
RAINBOW n = 330
n = 335
VEGFR Ramucirumab + paclitaxel
Placebo + paclitaxel
mOS: 9.6 months
mOS: 7.4 months
0.81
(0.68–0.96)
45
GATSBY n = 228
n = 117
HER2 Trastuzumab emtansine
Taxane
mOS: 7.9 months
mOS: 8.6 months
1.15
(0.87–1.51)
58
TyTAN n = 132
n = 129
HER2/EGFR Lapatinib + paclitaxel
Placebo + paclitaxel
mOS: 11.0 months
mOS: 8.9 months
0.84
(0.64–1.11)
60
GRANITE-1 n = 439
n = 217
mTOR Everolimus
Placebo
mOS: 5.9 months
mOS: 4.3 months
0.90
(0.75–1.08)
67
RADPAC n = 150
n = 150
mTOR Everolimus + paclitaxel
Placebo + paclitaxel
mOS: 6.1 months
mOS: 5.1 months
0.92
-
68
SHINE n = 41
n = 30
FGFR AZD4547
paclitaxel
mOS: 5.5 months
mOS: 6.6 months
1.31
(0.89–1.95) a
69
GOLD n = 263
n = 262
PARP Olaparib + paclitaxel
Placebo + paclitaxel
mOS: 8.8 months
mOS: 6.9 months
0.79
(0.63–1.00)
72

a80% confidence interval. Cape/Cis, capecitabine and cisplatin; CapeOx, capecitabine and oxaliplatin; CI, confidence interval; ECX, epirubicin, cisplatin, and capecitabine; EGFR, epidermal growth factor receptor; EOC/EOX, epirubicin, oxaliplatin, and capecitabine; FGFR, fibroblast growth factor receptor; FOLFOX, leucovorin, 5-fluorouracil, and oxaliplatin; FP, cisplatin and 5-fluorouracil; HER2, human epidermal growth factor receptor 2; HR, hazard rate; MET, mesenchymal–epithelial transition; mOS, median overall survival; mTOR, mammalian target of rapamycin; PARP, poly (ADP-ribose) polymerase; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; XP, cisplatin and capecitabine.

Editorial Note on the Review Process

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are:

  • Rupert Langer, University of Bern, Institute of Pathology, Bern, Switzerland

  • Rebecca KS Wong, Princess Margaret Cancer Center, Dept Radiation Oncology, University of Toronto, 610 University Avenue, Toronto, Canada

Funding Statement

This research was supported by generous grants from the Caporella, Dallas, Sultan, Park, Smith, Frazier, Oaks, Vanstekelenberg, Planjery, and Cantu families as well as from the Schecter Private Foundation, Rivercreek Foundation, Kevin Fund, Myer Fund, Dio Fund, Milrod Fund, and the University of Texas M.D. Anderson Cancer Center (Houston, TX, USA) multidisciplinary grant program. This research was also supported in part by the National Cancer Institute and Department of Defense awards CA138671, CA172741, CA129926, and CA150334 (JAA) and by a grant from the Japan Society for the Promotion of Science Overseas Research Fellowships and Program for Advancing Strategic International Networks to Accelerate the Circulation of Talented Researchers (KH).

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

[version 1; referees: 2 approved]

References

  • 1. Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Allen C, et al. : Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2017;3(4):524–48. 10.1001/jamaoncol.2016.5688 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Crew KD, Neugut AI: Epidemiology of gastric cancer. World J Gastroenterol. 2006;12(3):354–62. 10.3748/wjg.v12.i3.354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Blot WJ, Devesa SS, Kneller RW, et al. : Rising incidence of adenocarcinoma of the esophagus and gastric cardia. JAMA. 1991;265(10):1287–9. 10.1001/jama.1991.03460100089030 [DOI] [PubMed] [Google Scholar]
  • 4. Cristescu R, Lee J, Nebozhyn M, et al. : Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21(5):449–56. 10.1038/nm.3850 [DOI] [PubMed] [Google Scholar]
  • 5. Cancer Genome Atlas Research Network: Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513(7517):202–9. 10.1038/nature13480 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 6. Liu Y, Sethi NS, Hinoue T, et al. : Comparative Molecular Analysis of Gastrointestinal Adenocarcinomas. Cancer Cell. 2018;33(4):721–735.e8. 10.1016/j.ccell.2018.03.010 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 7. Sohn BH, Hwang JE, Jang HJ, et al. : Clinical Significance of Four Molecular Subtypes of Gastric Cancer Identified by The Cancer Genome Atlas Project. Clin Cancer Res. 2017;23(15):4441–9. 10.1158/1078-0432.CCR-16-2211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Saeterdal I, Bjørheim J, Lislerud K, et al. : Frameshift-mutation-derived peptides as tumor-specific antigens in inherited and spontaneous colorectal cancer. Proc Natl Acad Sci U S A. 2001;98(23):13255–60. 10.1073/pnas.231326898 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Colli LM, Machiela MJ, Myers TA, et al. : Burden of Nonsynonymous Mutations among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. Cancer Res. 2016;76(13):3767–72. 10.1158/0008-5472.CAN-16-0170 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 10. Japanese Gastric Cancer Association: Japanese gastric cancer treatment guidelines 2014 (ver. 4). Gastric Cancer. 2017;20(1):1–19. 10.1007/s10120-016-0622-4 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 11. Sasako M, Sakuramoto S, Katai H, et al. : Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol. 2011;29(33):4387–93. 10.1200/JCO.2011.36.5908 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 12. Noh SH, Park SR, Yang HK, et al. : Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15(12):1389–96. 10.1016/S1470-2045(14)70473-5 [DOI] [PubMed] [Google Scholar]
  • 13. Bajetta E, Floriani I, Di Bartolomeo M, et al. : Randomized trial on adjuvant treatment with FOLFIRI followed by docetaxel and cisplatin versus 5-fluorouracil and folinic acid for radically resected gastric cancer. Ann Oncol. 2014;25(7):1373–8. 10.1093/annonc/mdu146 [DOI] [PubMed] [Google Scholar]
  • 14. Cascinu S, Labianca R, Barone C, et al. : Adjuvant treatment of high-risk, radically resected gastric cancer patients with 5-fluorouracil, leucovorin, cisplatin, and epidoxorubicin in a randomized controlled trial. J Natl Cancer Inst. 2007;99(8):601–7. 10.1093/jnci/djk131 [DOI] [PubMed] [Google Scholar]
  • 15. Kodera Y, Yoshida K, Kochi M, et al. : A randomized phase III study comparing S-1 plus docetaxel with S-1 alone as a postoperative adjuvant chemotherapy for curatively resected stage III gastric cancer (JACCRO GC-07 trial). J Clin Oncol. 2018;36(15_suppl): abstr 4007. Reference Source [Google Scholar]
  • 16. Macdonald JS, Smalley SR, Benedetti J, et al. : Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345(10):725–30. 10.1056/NEJMoa010187 [DOI] [PubMed] [Google Scholar]
  • 17. Smalley SR, Benedetti JK, Haller DG, et al. : Updated analysis of SWOG-directed intergroup study 0116: A phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol. 2012;30(19):2327–33. 10.1200/JCO.2011.36.7136 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 18. Lee J, Lim DH, Kim S, et al. : Phase III trial comparing capecitabine plus cisplatin versus capecitabine plus cisplatin with concurrent capecitabine radiotherapy in completely resected gastric cancer with D2 lymph node dissection: The ARTIST trial. J Clin Oncol. 2012;30(3):268–73. 10.1200/JCO.2011.39.1953 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 19. Verheij M, Jansen EP, Cats A, et al. : A multicenter randomized phase III trial of neo-adjuvant chemotherapy followed by surgery and chemotherapy or by surgery and chemoradiotherapy in resectable gastric cancer: First results from the CRITICS study. J Clin Oncol. 2016;34(15_suppl):4000 Reference Source 27646943 [Google Scholar]
  • 20. Cunningham D, Allum WH, Stenning SP, et al. : Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355(1):11–20. 10.1056/NEJMoa055531 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 21. Elimova E, Janjigian YY, Mulcahy M, et al. : It Is Time to Stop Using Epirubicin to Treat Any Patient With Gastroesophageal Adenocarcinoma. J Clin Oncol. 2017;35(4):475–7. 10.1200/JCO.2016.69.7276 [DOI] [PubMed] [Google Scholar]
  • 22. Ychou M, Boige V, Pignon JP, et al. : Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: An FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29(13):1715–21. 10.1200/JCO.2010.33.0597 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 23. Alderson D, Langley RE, Nankivell MG, et al. : Neoadjuvant chemotherapy for resectable oesophageal and junctional adenocarcinoma: Results from the UK Medical Research Council randomised OEO5 trial (ISRCTN 01852072). J Clin Oncol. 2015;33(15_suppl):4002 Reference Source [Google Scholar]
  • 24. Al-Batran SE, Pauligk C, Homann N, et al. : LBA27_PRDocetaxel, oxaliplatin, and fluorouracil/leucovorin (FLOT) for resectable esophagogastric cancer: Updated results from multicenter, randomized phase 3 FLOT4-AIO trial (German Gastric Group at AIO). Ann Oncol. 2017;28(suppl_5):mdx440.019 10.1093/annonc/mdx440.019 [DOI] [Google Scholar]
  • 25. Al-Batran SE, Homann N, Schmalenberg H, et al. : Perioperative chemotherapy with docetaxel, oxaliplatin, and fluorouracil/leucovorin (FLOT) versus epirubicin, cisplatin, and fluorouracil or capecitabine (ECF/ECX) for resectable gastric or gastroesophageal junction (GEJ) adenocarcinoma (FLOT4-AIO): A multicenter, randomized phase 3 trial. J Clin Oncol. 2017;35(15_suppl):4004 Reference Source [Google Scholar]
  • 26. Cunningham D, Stenning SP, Smyth EC, et al. : Peri-operative chemotherapy with or without bevacizumab in operable oesophagogastric adenocarcinoma (UK Medical Research Council ST03): Primary analysis results of a multicentre, open-label, randomised phase 2-3 trial. Lancet Oncol. 2017;18(3):357–70. 10.1016/S1470-2045(17)30043-8 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 27. van Hagen P, Hulshof MC, van Lanschot JJ, et al. : Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366(22):2074–84. 10.1056/NEJMoa1112088 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 28. Ajani JA, D'Amico TA, Almhanna K, et al. : Gastric Cancer, Version 3.2016, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2016;14(10):1286–312. 10.6004/jnccn.2016.0137 [DOI] [PubMed] [Google Scholar]
  • 29. Ajani JA, Mansfield PF, Crane CH, et al. : Paclitaxel-based chemoradiotherapy in localized gastric carcinoma: Degree of pathologic response and not clinical parameters dictated patient outcome. J Clin Oncol. 2005;23(6):1237–44. 10.1200/JCO.2005.01.305 [DOI] [PubMed] [Google Scholar]
  • 30. Ajani JA, Mansfield PF, Janjan N, et al. : Multi-institutional trial of preoperative chemoradiotherapy in patients with potentially resectable gastric carcinoma. J Clin Oncol. 2004;22(14):2774–80. 10.1200/JCO.2004.01.015 [DOI] [PubMed] [Google Scholar]
  • 31. Ajani JA, Winter K, Okawara GS, et al. : Phase II trial of preoperative chemoradiation in patients with localized gastric adenocarcinoma (RTOG 9904): quality of combined modality therapy and pathologic response. J Clin Oncol. 2006;24(24):3953–8. 10.1200/JCO.2006.06.4840 [DOI] [PubMed] [Google Scholar]
  • 32. Elimova E, Slack RS, Chen HC, et al. : Patterns of relapse in patients with localized gastric adenocarcinoma who had surgery with or without adjunctive therapy: costs and effectiveness of surveillance. Oncotarget. 2017;8(46):81430–40. 10.18632/oncotarget.19226 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Leong T, Smithers BM, Michael M, et al. : TOPGEAR: a randomised phase III trial of perioperative ECF chemotherapy versus preoperative chemoradiation plus perioperative ECF chemotherapy for resectable gastric cancer (an international, intergroup trial of the AGITG/TROG/EORTC/NCIC CTG). BMC Cancer. 2015;15:532. 10.1186/s12885-015-1529-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. 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. 10.1016/S0140-6736(10)61121-X [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 35. Guimbaud R, Louvet C, Ries P, et al. : Prospective, randomized, multicenter, phase III study of fluorouracil, leucovorin, and irinotecan versus epirubicin, cisplatin, and capecitabine in advanced gastric adenocarcinoma: a French intergroup (Fédération Francophone de Cancérologie Digestive, Fédération Nationale des Centres de Lutte Contre le Cancer, and Groupe Coopérateur Multidisciplinaire en Oncologie) study. J Clin Oncol. 2014;32(31):3520–6. 10.1200/JCO.2013.54.1011 [DOI] [PubMed] [Google Scholar]
  • 36. Ajani JA, Baker J, Pisters PW, et al. : CPT-11 plus cisplatin in patients with advanced, untreated gastric or gastroesophageal junction carcinoma: results of a phase II study. Cancer. 2002;94(3):641–6. 10.1002/cncr.10279 [DOI] [PubMed] [Google Scholar]
  • 37. Dank M, Zaluski J, Barone C, et al. : Randomized phase III study comparing irinotecan combined with 5-fluorouracil and folinic acid to cisplatin combined with 5-fluorouracil in chemotherapy naive patients with advanced adenocarcinoma of the stomach or esophagogastric junction. Ann Oncol. 2008;19(8):1450–7. 10.1093/annonc/mdn166 [DOI] [PubMed] [Google Scholar]
  • 38. Van Cutsem E, Moiseyenko VM, Tjulandin S, et al. : Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. J Clin Oncol. 2006;24(31):4991–7. 10.1200/JCO.2006.06.8429 [DOI] [PubMed] [Google Scholar]
  • 39. Ajani JA, Fodor MB, Tjulandin SA, et al. : Phase II multi-institutional randomized trial of docetaxel plus cisplatin with or without fluorouracil in patients with untreated, advanced gastric, or gastroesophageal adenocarcinoma. J Clin Oncol. 2005;23(24):5660–7. 10.1200/JCO.2005.17.376 [DOI] [PubMed] [Google Scholar]
  • 40. Blum Murphy MA, Qiao W, Mewada N, et al. : A Phase I/II Study of Docetaxel, Oxaliplatin, and Fluorouracil (D-FOX) Chemotherapy in Patients With Untreated Locally Unresectable or Metastatic Adenocarcinoma of the Stomach and Gastroesophageal Junction. Am J Clin Oncol. 2018;41(4):321–5. [DOI] [PubMed] [Google Scholar]
  • 41. Al-Batran SE, Hartmann JT, Hofheinz R, et al. : Biweekly fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) for patients with metastatic adenocarcinoma of the stomach or esophagogastric junction: a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie. Ann Oncol. 2008;19(11):1882–7. 10.1093/annonc/mdn403 [DOI] [PubMed] [Google Scholar]
  • 42. Van Cutsem E, Boni C, Tabernero J, et al. : Docetaxel plus oxaliplatin with or without fluorouracil or capecitabine in metastatic or locally recurrent gastric cancer: a randomized phase II study. Ann Oncol. 2015;26(1):149–56. 10.1093/annonc/mdu496 [DOI] [PubMed] [Google Scholar]
  • 43. Shah MA, Janjigian YY, Stoller R, et al. : Randomized Multicenter Phase II Study of Modified Docetaxel, Cisplatin, and Fluorouracil (DCF) Versus DCF Plus Growth Factor Support in Patients With Metastatic Gastric Adenocarcinoma: A Study of the US Gastric Cancer Consortium. J Clin Oncol. 2015;33(33):3874–9. 10.1200/JCO.2015.60.7465 [DOI] [PubMed] [Google Scholar]
  • 44. Elimova E, Ajani JA: Time-to-Treatment Failure As the Primary End Point of a First-Line Advanced Gastric Cancer Randomized Trial: How Confused Would You Want Us to Be? J Clin Oncol. 2015;33(21):2410. 10.1200/JCO.2014.59.8847 [DOI] [PubMed] [Google Scholar]
  • 45. 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. 10.1016/S1470-2045(14)70420-6 [DOI] [PubMed] [Google Scholar]
  • 46. 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. 10.1016/S0140-6736(13)61719-5 [DOI] [PubMed] [Google Scholar]
  • 47. Ajani JA, Lee J, Sano T, et al. : Gastric adenocarcinoma. Nat Rev Dis Primers. 2017;3: 17036. 10.1038/nrdp.2017.36 [DOI] [PubMed] [Google Scholar]
  • 48. Thuss-Patience PC, Kretzschmar A, Bichev D, et al. : Survival advantage for irinotecan versus best supportive care as second-line chemotherapy in gastric cancer--a randomised phase III study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Eur J Cancer. 2011;47(15):2306–14. 10.1016/j.ejca.2011.06.002 [DOI] [PubMed] [Google Scholar]
  • 49. Kang JH, Lee SI, Lim DH, et al. : Salvage chemotherapy for pretreated gastric cancer: a randomized phase III trial comparing chemotherapy plus best supportive care with best supportive care alone. J Clin Oncol. 2012;30(13):1513–8. 10.1200/JCO.2011.39.4585 [DOI] [PubMed] [Google Scholar]
  • 50. Hironaka S, Ueda S, Yasui H, et al. : Randomized, open-label, phase III study comparing irinotecan with paclitaxel in patients with advanced gastric cancer without severe peritoneal metastasis after failure of prior combination chemotherapy using fluoropyrimidine plus platinum: WJOG 4007 trial. J Clin Oncol. 2013;31(35):4438–44. 10.1200/JCO.2012.48.5805 [DOI] [PubMed] [Google Scholar]
  • 51. Ishigami H, Fujiwara Y, Fukushima R, et al. : Phase III study of intraperitoneal paclitaxel plus s-1/paclitaxel compared with s-1/cisplatin in gastric cancer patients with peritoneal metastasis: PHOENIX-GC trial. J Clin Oncol. 2016;34(15_suppl):4014 Reference Source [DOI] [PubMed] [Google Scholar]
  • 52. Ishigami H, Yamaguchi H, Yamashita H, et al. : Surgery after intraperitoneal and systemic chemotherapy for gastric cancer with peritoneal metastasis or positive peritoneal cytology findings. Gastric Cancer. 2017;20(Suppl 1):128–34. 10.1007/s10120-016-0684-3 [DOI] [PubMed] [Google Scholar]
  • 53. Kodera Y, Ito S, Mochizuki Y, et al. : Long-term follow up of patients who were positive for peritoneal lavage cytology: final report from the CCOG0301 study. Gastric Cancer. 2012;15(3):335–7. 10.1007/s10120-012-0156-3 [DOI] [PubMed] [Google Scholar]
  • 54. Kodera Y, Takahashi N, Yoshikawa T, et al. : Feasibility of weekly intraperitoneal versus intravenous paclitaxel therapy delivered from the day of radical surgery for gastric cancer: a preliminary safety analysis of the INPACT study, a randomized controlled trial. Gastric Cancer. 2017;20(1):190–9. 10.1007/s10120-016-0598-0 [DOI] [PubMed] [Google Scholar]
  • 55. Aizawa M, Ishigami H, Yabusaki H, et al. : Phase II study of intraperitoneal paclitaxel plus S-1/paclitaxel for gastric cancer with positive peritoneal cytology: CY-PHOENIX trial. J Clin Oncol. 2017;35(4_suppl):96 10.1200/JCO.2017.35.4_suppl.96 28034065 [DOI] [Google Scholar]
  • 56. Badgwell B, Blum M, Das P, et al. : Phase II Trial of Laparoscopic Hyperthermic Intraperitoneal Chemoperfusion for Peritoneal Carcinomatosis or Positive Peritoneal Cytology in Patients with Gastric Adenocarcinoma. Ann Surg Oncol. 2017;24(11):3338–44. 10.1245/s10434-017-6047-4 [DOI] [PubMed] [Google Scholar]
  • 57. Bartley AN, Washington MK, Colasacco C, et al. : HER2 Testing and Clinical Decision Making in Gastroesophageal Adenocarcinoma: Guideline From the College of American Pathologists, American Society for Clinical Pathology, and the American Society of Clinical Oncology. J Clin Oncol. 2017;35(4):446–64. 10.1200/JCO.2016.69.4836 [DOI] [PubMed] [Google Scholar]
  • 58. Thuss-Patience PC, Shah MA, Ohtsu A, et al. : Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): an international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol. 2017;18(5):640–53. 10.1016/S1470-2045(17)30111-0 [DOI] [PubMed] [Google Scholar]
  • 59. Hecht JR, Bang YJ, Qin SK, et al. : Lapatinib in Combination With Capecitabine Plus Oxaliplatin in Human Epidermal Growth Factor Receptor 2-Positive Advanced or Metastatic Gastric, Esophageal, or Gastroesophageal Adenocarcinoma: TRIO-013/LOGiC--A Randomized Phase III Trial. J Clin Oncol. 2016;34(5):443–51. 10.1200/JCO.2015.62.6598 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 60. 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. 10.1200/JCO.2013.53.6136 [DOI] [PubMed] [Google Scholar]
  • 61. 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. 10.1016/S1470-2045(13)70102-5 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 62. 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. 10.1016/S1470-2045(13)70096-2 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 63. Fuchs CS, Shitara K, Bartolomeo MD, et al. : RAINFALL: A randomized, double-blind, placebo-controlled phase III study of cisplatin (Cis) plus capecitabine (Cape) or 5FU with or without ramucirumab (RAM) as first-line therapy in patients with metastatic gastric or gastroesophageal junction (G-GEJ) adenocarcinoma. J Clin Oncol. 2018;36(4_suppll):5 Reference Source [Google Scholar]
  • 64. 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. 10.1200/JCO.2011.36.2236 [DOI] [PubMed] [Google Scholar]
  • 65. Catenacci DVT, Tebbutt NC, Davidenko I, et al. : Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18(11):1467–82. 10.1016/S1470-2045(17)30566-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Shah MA, Bang YJ, Lordick F, et al. : Effect of Fluorouracil, Leucovorin, and Oxaliplatin With or Without Onartuzumab in HER2-Negative, MET-Positive Gastroesophageal Adenocarcinoma: The METGastric Randomized Clinical Trial. JAMA Oncol. 2017;3(5):620–7. 10.1001/jamaoncol.2016.5580 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. 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. 10.1200/JCO.2012.48.3552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Al-Batran SE, Riera-Knorrenschild J, Pauligk C, et al. : A randomized, double-blind, multicenter phase III study evaluating paclitaxel with and without RAD001 in patients with gastric cancer who have progressed after therapy with a fluoropyrimidine/platinum-containing regimen (RADPAC). J Clin Oncol. 2017;35(4_suppl):4 10.1200/JCO.2017.35.4_suppl.4 28034060 [DOI] [Google Scholar]
  • 69. Van Cutsem E, Bang YJ, Mansoor W, et al. : A randomized, open-label study of the efficacy and safety of AZD4547 monotherapy versus paclitaxel for the treatment of advanced gastric adenocarcinoma with FGFR2 polysomy or gene amplification. Ann Oncol. 2017;28(6):1316–24. 10.1093/annonc/mdx107 [DOI] [PubMed] [Google Scholar]
  • 70. Farmer H, McCabe N, Lord CJ, et al. : Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434(7035):917–21. 10.1038/nature03445 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 71. Bang YJ, Im SA, Lee KW, et al. : Randomized, Double-Blind Phase II Trial With Prospective Classification by ATM Protein Level to Evaluate the Efficacy and Tolerability of Olaparib Plus Paclitaxel in Patients With Recurrent or Metastatic Gastric Cancer. J Clin Oncol. 2015;33(33):3858–65. 10.1200/JCO.2014.60.0320 [DOI] [PubMed] [Google Scholar]
  • 72. Bang YJ, Xu RH, Chin K, et al. : Olaparib in combination with paclitaxel in patients with advanced gastric cancer who have progressed following first-line therapy (GOLD): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18(12):1637–51. 10.1016/S1470-2045(17)30682-4 [DOI] [PubMed] [Google Scholar]
  • 73. Bekaii-Saab T, El-Rayes B: Identifying and targeting cancer stem cells in the treatment of gastric cancer. Cancer. 2017;123(8):1303–12. 10.1002/cncr.30538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Cohen DJ, Christos PJ, Kindler HL, et al. : Vismodegib (V), a hedgehog (HH) pathway inhibitor, combined with FOLFOX for first-line therapy of patients (pts) with advanced gastric and gastroesophageal junction (GEJ) carcinoma: A New York Cancer Consortium led phase II randomized study. J Clin Oncol. 2013;31(15_suppl):4011 Reference Source [Google Scholar]
  • 75. Sonbol MB, Bekaii-Saab T: A clinical trial protocol paper discussing the BRIGHTER study. Future Oncol. 2018;14(10):901–6. 10.2217/fon-2017-0406 [DOI] [PubMed] [Google Scholar]
  • 76. Yoon C, Park DJ, Schmidt B, et al. : CD44 expression denotes a subpopulation of gastric cancer cells in which Hedgehog signaling promotes chemotherapy resistance. Clin Cancer Res. 2014;20(15):3974–88. 10.1158/1078-0432.CCR-14-0011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Sahin U, Koslowski M, Dhaene K, et al. : Claudin-18 splice variant 2 is a pan-cancer target suitable for therapeutic antibody development. Clin Cancer Res. 2008;14(23):7624–34. 10.1158/1078-0432.CCR-08-1547 [DOI] [PubMed] [Google Scholar]
  • 78. Al-Batran SE, Schuler MH, Zvirbule Z, et al. : FAST: An international, multicenter, randomized, phase II trial of epirubicin, oxaliplatin, and capecitabine (EOX) with or without IMAB362, a first-in-class anti-CLDN18.2 antibody, as first-line therapy in patients with advanced CLDN18.2+ gastric and gastroesophageal junction (GEJ) adenocarcinoma. J Clin Oncol. 2016;34(18_suppl):LBA4001–LBA. 10.1200/JCO.2016.34.18_suppl.LBA4001 [DOI] [Google Scholar]
  • 79. Shah MA, Starodub A, Sharma S, et al. : Andecaliximab/GS-5745 Alone and Combined with mFOLFOX6 in Advanced Gastric and Gastroesophageal Junction Adenocarcinoma: Results from a Phase I Study. Clin Cancer Res. 2018;24(16):3829–3837. 10.1158/1078-0432.CCR-17-2469 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 80. Bendell JC, Starodub A, Wainberg ZA, et al. : A phase 3 randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of GS-5745 combined with mFOLFOX6 as first-line treatment in patients with advanced gastric or gastroesophageal junction adenocarcinoma. J Clin Oncol. 2016;34(15_suppl):TPS4132–TPS. Reference Source [Google Scholar]
  • 81. Luke JJ, Flaherty KT, Ribas A, et al. : Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol. 2017;14(18):463–82. 10.1038/nrclinonc.2017.43 [DOI] [PubMed] [Google Scholar]
  • 82. Giroux Leprieur E, Dumenil C, Julie C, et al. : Immunotherapy revolutionises non-small-cell lung cancer therapy: Results, perspectives and new challenges. Eur J Cancer. 2017;78:16–23. 10.1016/j.ejca.2016.12.041 [DOI] [PubMed] [Google Scholar]
  • 83. Kang YK, Boku N, Satoh T, et al. : Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71. 10.1016/S0140-6736(17)31827-5 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 84. Janjigian YY, Ott PA, Calvo E, et al. : Nivolumab ± ipilimumab in pts with advanced (adv)/metastatic chemotherapy-refractory (CTx-R) gastric (G), esophageal (E), or gastroesophageal junction (GEJ) cancer: CheckMate 032 study. J Clin Oncol. 2017;35(15_suppl):4014 Reference Source [Google Scholar]
  • 85. Fuchs CS, Doi T, Jang RW, et al. : Safety and Efficacy of Pembrolizumab Monotherapy in Patients With Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol. 2018;4(5):e180013. 10.1001/jamaoncol.2018.0013 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 86. Bang YJ, Muro K, Fuchs CS, et al. : KEYNOTE-059 cohort 2: Safety and efficacy of pembrolizumab (pembro) plus 5-fluorouracil (5-FU) and cisplatin for first-line (1L) treatment of advanced gastric cancer. J Clin Oncol. 2017;35(15_suppl):4012 Reference Source 28934000 [Google Scholar]
  • 87. Tabernero J, Bang YJ, Fuchs CS, et al. : KEYNOTE-062: Phase III study of pembrolizumab (MK-3475) alone or in combination with chemotherapy versus chemotherapy alone as first-line therapy for advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma. J Clin Oncol. 2016;34(4_suppl):TPS185–TPS. 10.1200/jco.2016.34.4_suppl.tps185 [DOI] [Google Scholar]
  • 88. Chung HC, Arkenau HT, Wyrwicz L, et al. : Avelumab (MSB0010718C; anti-PD-L1) in patients with advanced gastric or gastroesophageal junction cancer from JAVELIN solid tumor phase Ib trial: Analysis of safety and clinical activity. J Clin Oncol. 2016;34(15_suppl):4009 Reference Source [Google Scholar]
  • 89. Le DT, Durham JN, Smith KN, et al. : Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–13. 10.1126/science.aan6733 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 90. Dempke WCM, Fenchel K, Uciechowski P, et al. : Second- and third-generation drugs for immuno-oncology treatment-The more the better? Eur J Cancer. 2017;74:55–72. 10.1016/j.ejca.2017.01.001 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation

Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES