Abstract

Background

Platinum and fluoropyrimidine combinations typically comprise first-line (1L) therapy in advanced gastric cancer or gastroesophageal junction adenocarcinoma (G/GEA), although controversy exists regarding the use of 5doublet versus triplet cytotoxic regimens. Historically, second-line (2L) and third-line or later (3L+) therapy has been fragmented. Recent trials have increased the need for optimal treatment sequencing in advanced G/GEA.

Materials and Methods

We conducted a systematic search of peer-reviewed manuscripts of randomized clinical trials examining 1L, 2L, and 3L+ therapy for advanced G/GEA published from 2009 through November 19, 2019. When available, overall survival, progression-free survival, time to progression, overall response rate, and toxicity were extracted from each and compared descriptively.

Results

In 1L therapy, chemotherapy triplets demonstrated variable efficacy improvements with invariable increased toxicity compared with platinum/fluoropyrimidine doublets. Currently, the only published report of positive outcomes using biologics in 1L describes adding trastuzumab in HER2-overexpressing advanced G/GEA. In 2L, doublet chemotherapy regimens are not uniformly more efficacious than single-agent taxanes or irinotecan, and ramucirumab has demonstrated improved outcomes both as monotherapy and in combination.

Conclusion

For advanced G/GEA, review of trial results from 2009–2019 support 1L therapy with platinum and fluoropyrimidine and sequencing with taxanes or irinotecan in combination with biologics as effective 2L options. Escalating to a triplet may add some efficacy at the expense of added toxicity.

Implications for Practice

The rapidly changing treatment landscape for advanced gastric cancer includes increasing options for refractory disease. With multiple first-line platinum-based regimens, identification of those with the best benefit-to-risk ratio may provide guidance on treatment sequencing strategies. This article presents findings from the published literature of randomized controlled trials that included a first-line platinum/fluoropyrimidine combination and, for second-line trials, patients with platinum/fluoropyrimidine-refractory disease. This guiding summary could be a tool for clinicians to identify the optimal first-line regimen(s) followed by a strategy for subsequent regimens.

Introduction

Gastric cancer is the fifth most common cancer and third leading cause of cancer-related deaths worldwide [1]. Gastric cancer is a histologically and molecularly diverse disease encompassing the stomach and gastroesophageal junction. Adenocarcinoma is the most common histological type, and gastric cancer or gastroesophageal junction adenocarcinoma (G/GEA), with or without esophageal adenocarcinoma, are commonly studied within the same clinical trials [2]. Developments in treatment for locally advanced and unresectable/metastatic G/GEA lag behind other solid malignancies, with a median survival of less than 1 year [35].

Despite multiple options, there is no single standard of care for first-line (1L), second-line (2L), or third-line (3L) and beyond (3L+) treatment of G/GEA [6,7]. Current guidelines do not address optimizing sequence. The Cochrane reviews by Wagner evaluated the efficacy of chemotherapy versus best supportive care (BSC), combination versus single-agent chemotherapy, and different chemotherapy combinations [8,9]. However, the question of treatment sequencing was not addressed.

In the 1L setting, current options include platinum agents, fluoropyrimidines, taxanes, irinotecan, and anthracyclines in doublet or triplet regimens, whereas epirubicin has fallen out of favor [10,11]. The most commonly used 1L treatment combinations include fluoropyrimidine plus platinum, with or without a third agent [8,12], although addition of a third cytotoxic agent to established doublet regimens is likely to increase toxicities as reported in 2006 [13]. Unfortunately, the majority of patients who respond to 1L chemotherapy will relapse or experience disease progression [8]. It is unclear if there is a significant benefit with doublet therapies versus monotherapies, intravenous versus oral formulations of fluorouracil (5-FU), cisplatin versus oxaliplatin, or irinotecan versus docetaxel.

There is disagreement regarding the preferred treatment regimen in the 2L and 3L+ settings. The treatment landscape is fragmented, particularly in the U.S. [14]. Current recommended 2L therapies include the anti–vascular endothelial growth factor receptor-2 monoclonal antibody, ramucirumab, as monotherapy or combined with paclitaxel, or single chemotherapy agents (irinotecan, docetaxel, or paclitaxel) [12,15]. The diverse array of regimens is counterproductive to developing clear, standardized, evidence-based guidelines. Moreover, with the recent publication of several randomized controlled trials (RCTs) investigating novel therapies and chemotherapy combinations, a new evaluation of existing evidence is needed that might better inform physicians and guide treatment recommendations.

We conducted a systematic review from published RCTs to evaluate and synthesize evidence and provide insights into an evidence-based treatment sequencing strategy for advanced G/GEA. To this end, the review focused on RCTs in which the commonly recommended platinum/fluoropyrimidine-backbone was used in 1L and, for 2L, RCTs that included a prior platinum and/or fluoropyrimidine. Given the recent changes to the G/GEA landscape, we have discussed top-line data from seminal trials and approvals in this report.

Materials and Methods

Search Strategy

The systematic literature review (SLR) search, selection, and data extraction were conducted and reported using PRISMA guidelines [16]. The databases MEDLINE, MEDLINE In-Process, Embase, and the Cochrane Library were searched to identify English-language publications of RCTs, SLRs, and meta-analyses since the Cochrane review by Wagner et al. [9]. The search for RCTs was limited to 2009 through November 19, 2019, and the search for SLRs and meta-analyses was limited to 2015 through November 19, 2019. The review only included RCTs of larger populations: ≥200 and ≥ 40 patients in 1L and 2L or later settings, respectively. Although outside the original SLR parameters, recent phase III RCT data are also discussed in relevant sections.

The RCT search, SLR, and meta-analyses were structured as follows: study type search terms, disease search terms, treatment search terms, population search terms, and exclusionary search terms. Further details regarding inclusion and exclusion criteria, screening, and study quality assessment methodology from the SLR are available in the supplemental online data.

Synthesis Methods

Overall survival (OS), progression-free survival (PFS), time to progression (TTP), and overall response rate (ORR) were the primary efficacy endpoints considered. Overviews of adverse events (AEs) were summarized. Included studies were heterogeneous in terms of study design; therefore, results are presented descriptively.

Results

Literature Search Results

The screening process and number of identified articles are detailed in Figure 1. Literature searches identified a total of 920 nonduplicate records, of which 647 and 212 records were excluded during level 1 and 2 screenings, respectively. Seventy publications meeting eligibility criteria were included (Fig. 1). Of these, 27 articles assessed 1L, 34 assessed 2L, and 8 assessed 3L+.

Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) diagram for study inclusion and exclusion. The PRISMA flow chart details the number of articles identified in the literature search and the number of articles included and excluded at each stage. Note that articles from the SLR-MA search that met the inclusion criteria for reference list review to identify potential primary RCT publications are listed in the PRISMA diagram as excluded at level 2 for reason “other” (as these articles are not primary RCTs). Abbreviations: 2L, second line; 3L, third line; MA, meta-analysis; RCT, randomized controlled trial; SLR, systematic literature review.
Figure 1

Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) diagram for study inclusion and exclusion. The PRISMA flow chart details the number of articles identified in the literature search and the number of articles included and excluded at each stage. Note that articles from the SLR-MA search that met the inclusion criteria for reference list review to identify potential primary RCT publications are listed in the PRISMA diagram as excluded at level 2 for reason “other” (as these articles are not primary RCTs). Abbreviations: 2L, second line; 3L, third line; MA, meta-analysis; RCT, randomized controlled trial; SLR, systematic literature review.

Risk of Bias

The quality of each study was evaluated using the bias assessment tool detailed in supplemental online Table 4.

Description of Included Studies

An overview of the studies is provided in the supplemental online data. Patient demographics and disease characteristics are summarized in Figure 2 and supplemental online Table 1. A summary of treatment interventions for each line of therapy is provided in Figure 3 and supplemental online Table 3.

First-line, second-line, and third-line study overview. Abbreviations: ph1, phase I; ph2, phase II; ph3, phase III.
Figure 2

First-line, second-line, and third-line study overview. Abbreviations: ph1, phase I; ph2, phase II; ph3, phase III.

First-line, second-line, and third-line and beyond interventions. (A): First-line interventions. Targeted therapies include bevacizumab, cetuximab, lapatinib, onartuzumab, panitumumab, rilotumumab, trastuzumab, pertuzumab, and ramucirumab. Chemotherapy includes capecitabine, cisplatin, docetaxel, epirubicin, oxaliplatin, paclitaxel, S-1, tegafur, 5-fluorouracil, and leucovorin (folinic acid). Eleven studies compared the efficacy of chemotherapy doublets. (B): Second-line interventions. In the center of the diagram, “12 studies TT vs. CM/PB” include 12 studies with single-agent (SA) chemotherapy in both arms: six assessing the efficacy of SA versus SA and six assessing SA plus targeted therapy versus control. (C): Third-line interventions. Targeted therapies include avelumab, TAS-102, nivolumab (ICI), and ipilimumab (ICI). Chemotherapy includes irinotecan and paclitaxel. Abbreviations: ↑, increased/higher dose; BSC, best supportive care; CM, chemotherapy; DB, doublet; ICI, immune checkpoint inhibitor; Pac, paclitaxel; PB, placebo; Pembro, pembrolizumab; TP, triplet; Traz, trastuzumab; TT, targeted therapy; TX, taxane; VP, valproic acid.
Figure 3

First-line, second-line, and third-line and beyond interventions. (A): First-line interventions. Targeted therapies include bevacizumab, cetuximab, lapatinib, onartuzumab, panitumumab, rilotumumab, trastuzumab, pertuzumab, and ramucirumab. Chemotherapy includes capecitabine, cisplatin, docetaxel, epirubicin, oxaliplatin, paclitaxel, S-1, tegafur, 5-fluorouracil, and leucovorin (folinic acid). Eleven studies compared the efficacy of chemotherapy doublets. (B): Second-line interventions. In the center of the diagram, “12 studies TT vs. CM/PB” include 12 studies with single-agent (SA) chemotherapy in both arms: six assessing the efficacy of SA versus SA and six assessing SA plus targeted therapy versus control. (C): Third-line interventions. Targeted therapies include avelumab, TAS-102, nivolumab (ICI), and ipilimumab (ICI). Chemotherapy includes irinotecan and paclitaxel. Abbreviations: ↑, increased/higher dose; BSC, best supportive care; CM, chemotherapy; DB, doublet; ICI, immune checkpoint inhibitor; Pac, paclitaxel; PB, placebo; Pembro, pembrolizumab; TP, triplet; Traz, trastuzumab; TT, targeted therapy; TX, taxane; VP, valproic acid.

Efficacy and Safety of 1L Interventions

First-line studies varied with respect to trial design and patient populations (supplemental online Table 1). Of 27 RCTs, 22 reported OS and/or PFS data. Fourteen RCTs reported statistically significant findings for OS, 12-month survival, PFS, TTP, time to treatment failure (TTF), or ORR (Table 1; Table 2; supplemental online Table 2) [1729]. An overview of AEs is summarized in supplemental online Table 3.

Table 1

Overview of efficacy results

Table 1

Overview of efficacy results

Table 2

Overall survival, progression-free survival, and overall response rate of included first-, second-, or third-line and beyond randomized controlled trials ordered by study publication year

TrialTreatmentPatients, nOverall survivalacProgression-free survivalacORRac
Median (95% CI), moEffect size(95% CI)Median (95% CI), moEffect size (95% CI)%Effect size
First-line studies
Kang 2009 [24]d,eCis + Cap13910.4 (9.1–11.0)HR, 0.85 (0.65–1.11); p = .005 vs. noninferiority margin of 1.255.6 (4.8–6.9)HR, 0.80 (0.63–1.03); p < .001 vs. noninferiority margin of 1.25
Cis + FU1378.9 (7.3–10.2)5.0 (3.9–5.7)
FLAGS
Ajani 2010 [26]f,g
S-1 + Cis5278.6HR, 0.92 (0.80–1.05); p = .19834.8 (4.0–5.5)HR, 0.99 (0.86–1.14); p = .915829.1p = .40
FU + Cis5267.95.5 (4.4–5.8)31.9
ToGA
Bang 2010 [17]h,i
Tras + Chemoj29413.8HR, 0.74 (0.60–0.91); p = .00466.7HR, 0.71 (0.59–0.85); p = .000247kp = .0017
Chemo29011.15.535k
AVAGAST
Ohtsu 2011 [25]d,l
BEV + Cis + Cap/FUm25212.1 (11.1–13.8)HR, 0.87 (0.73–1.03); p = .10026.7 (5.9–7.1)HR, 0.80 (0.68–0.98); p = .0037
PBO + Cis + Cap/FUm26510.1 (9.0–11.3)5.3 (4.4–5.6)
REAL3
Waddell 2013 [18]d
Epir + Ox + Cap27511.3 (9.6–13.0)HR, 1.37 (1.07–1.76); p = .0136.0 (5.5–6.5)HR, 1.22 (0.98–1.52); p = .068NRNR
Epir + Ox + Cap + PAN2788.8 (7.7–9.8)7.4 (6.3–8.5)NR
EXPAND
Lordick 2013 [114]d,n
CTX + Cap + Cis4559.4 (8.3–10.6)HR, 1.00 (0.87–1.17); p = .954.4 (4.2–5.5)HR, 1.09 (0.92–1.29); p = .32
Cap + Cis44910.7 (9.4–11.3)5.6 (5.1–5.7)
Van Cutsem 2015 [37]iDoc + Ox798.97 (7.79–10.87)NR4.5 (3.7–5.3)NRNRNR
Doc + Ox + FU/FOL8914.59 (11.70–21.78)7.7 (7.0–9.4)
Doc + Ox + Cap8611.30 (8.08–14.03)5.6 (4.3–6.4)NR
Guimbaud 2014 [22]d,oEpir + Cis + Cap2099.49HR, 1.01 (0.82–1.24); p = .955.3HR, 0.99 (0.81–1.21); p = .9690.4NS
FOLFIRI2079.725.895.7
Kim 2014 [112]d,pSIM + Cap + Cis12011.6 (9.2–13.9)HR, 0.966 (0.722–1.293); p = .8185.2 (4.3–6.1)HR, 0.930 (0.684–1.264); p = .664
PBO + Cap + Cis12411.5 (9.9–13.1)4.6 (3.5–5.7)
SOS
Ryu 2015 [109]h,q
S-1 (D1–14) + Cis (D1)30614.1 (11.4–15.8)HR, 0.99 (0.81–1.21); p = .915.5 (4.7–6.6)HR, 0.82 (0.68–0.99); p = .041860p = .065
S-1 (D1–21) + Cis (D1 or 8)30913.9 (11.6–15.9)4.9 (4.2–5.5)50
AVATAR
Shen 2015 [110]d
PBO + Cap + Cis10211.4 (8.6–16.0)HR, 1.11 (0.79–1.56); p = .566.0 (4.9–7.4)HR, 0.89 (0.66–1.21); p = .47NRNR
BEV + Cap + Cis10010.5 (8.9–14.1)6.3 (5.7–7.4)NRNR
JapicCTI-101021
Yamada 2015 [20]d,r
S-1 + Ox31814.1 (13.0–15.8)HR, 0.958 (0.803–1.142) (S-1 + Ox noninferior to S-1 + Cis)5.5 (4.4–5.7)HR, 1.004 (0.840–1.199); p = .0044 (noninferiority)55.7NR
S-1 + Cis32413.1 (12.1–15.1)5.4 (4.2–5.7)52.2
Li 2015 [113]eS-1 + Cis12010.0 (8.59–14.52)p = .82
FU + Cis11610.46 (8.92–13.84)
Wang 2016 [19]d,sDoc + Cis + FU12110.2 (8.6–11.9)HR, 0.71 (0.52–0.97); p = .03197.2 (5.5–8.8)HR, 0.58 (0.42–0.80); p = .0008 (log-rank test)48.7p = .0244
Cis + FU1228.5 (7.1–9.5)4.9 (4.5–6.0)33.9
G-SOX
Bando 2016 [21]d
S-1 + Ox (≥70 yr)11617.5≥70 yr: HR, 0.857 (0.629–1.167); p = .3255.7HR, 0.805 (0.588–1.102); p = .174NRNR
S-1 + Cis (≥70 yr)10413.55.5NR
S-1 + Ox (<70 yr)22713.3<70 yr: HR, 0.984 (0.800–1.209); p = .8774.4HR, 1.019 (0.827–1.256); p = .862NR
S-1 + Cis (<70 yr)23813.15.3NR
TRIO-013/LOGiC
Hecht 2016 [23]d,n
LAP + Cap + Ox24912.2HR, 0.91 (0.73–1.12); p = .34926.0HR, 0.82 (0.68–1.00); p = .0381NRNR
PBO + Cap + Ox23810.55.4NR
DIGEST
Ajani 2017 [27]
S-1 + Cis2397.5 (6.7–9.3)Unstratified: HR, 0.99 (0.76–1.28); p = .9312
Stratified: HR, 0.90 (0.68–1.19); p = .4631
4.4HR, 0.86 (0.65–1.14); p = .303934.7p = .0122
FU + Cis1226.6 (5.7–8.1)3.919.8
RILOMET-1
Catenacci 2017 [28]
Rilotumumab + Epir + Cis + Cap2628.8 (7.7–10.2)HR, 1.34 (1.10–1.63); p = .0035.6 (5.3–5.9)HR, 1.26 (1.04–1.51); p = .01629.8p = .0005
PBO + Epir + Cis + Cap26710.7 (9.6–12.4)6.0 (5.7–7.2)44.6
METGastric
Shah 2017 [39]
Onartuzumab + mFOLFOX621711.0HR, 0.82 (0.59–1.15); p = .246.7HR, 0.90 (0.71–1.16); p = .4346.1p = .25
PBO + mFOLFOX620711.36.840.6
MET 2+/3+ subgroup:
onartuzumab + mFOLFOX6
7811.0HR, 0.64 (0.40–1.03); p = .066.9HR, 0.79 (0.54–1.15); p = .2253.8p = .23
MET 2+/3+ subgroup:
PBO + mFOLFOX6
929.75.744.6
Shu 2017 [32]Ox + tegafur16413.4 (12.2–15.1)HR, 0.96 (0.80–1.39)5.5 (4.6–6.2)HR, 1.02 (0.82–1.31)44.5p = .702
Ox + Cis16814.2 (13.1–16.0)6.1 (5.6–6.4)48.8
HELOISE
Shah 2017 [115]
SoC Tras + Cap + Cis12412.5HR, 1.24 (0.86–1.78); p = .24015.7HR, 1.04 (0.76–1.40); p = .822258.9p = .76
Higher-dose Tras + Cap + Cis12410.65.656.9
Lu 2018 [29]Pac + Cap16012.5 (11.5–14.5)HR, 0.878 (0.685–1.125); p = .304.994 (4.304–6.275)HR, 0.906 (0.706–1.164); p = .4443.1p = .01
Cis + Cap16011.8 (10.0–13.7)5.257 (4.665–5.815)28.8
Lu 2019 [35]S-1 + Doc150405 daysp = .5127180 daysp > .05NRNR
S-1 + Cis150378 days171 daysNR
RAINFALL
Fuchs 2019 [40]t,u
RAM + Cis + Cap/FUv32611.2 (9.9–11.9)HR, 0.962 (0.801–1.156); p = .68Investigator-assessed: 5.7 (5.5–6.5)
Independent review: 5.5 (4.2–5.8)
Investigator-assessed:
HR, 0.753 (0.607–0.935); p = .011
Independent review:
HR, 0.961 (0.78–1.203); p = .74
41.1p = .17
PBO + Cis + Cap/FUo31910.7 (9.5–11.9)Investigator-assessed: 5.4 (4.5–5.7)
Independent review: 5.4 (4.4–5.7)
36.4
JACOB
Tabernero 2018 [38]
Pertuzumab + Tras/Chemo38817.5 (16.2–19.3)HR,0.84 (0.71–1.00); p = .0578.5 (8.2–9.7)HR, 0.73 (0.62–0.86); p = .000156.7p = .026
PBO + Tras/Chemo39214.2 (12.9–15.5)7.0 (6.4–8.2)48.3
JCOG1013
Yamada 2019 [36]
Doc + Cis/S-137014.2 (12.9–15.9)HR, 0.99 (0.85–1.16); p = .477.4 (6.7–7.8)HR, 0.99 (0.86–1.15); p = .9259p = .50
Cis/S-137115.3 (14.2–16.2)6.5 (5.9–7.4)56
Second-line studies
Thuss-Patience 2011 [47]wy,asIRI + BSC214.0 (3.6–7.5)All patients died:
HR, 0.48 (0.25–0.92); p = .012
ITT population: 2.5 (1.6–3.9)
Per-protocol population: 2.6 (1.7–4.3)
NR (results provided for IRI arm only)0zNR
BSC192.4 (1.7–4.9)NR (results provided for IRI arm only)NR
Yi 2012 [43]x,asDoc + sunitinib568.0 (5.4–10.6)HR, 0.94 (0.60–1.49); p = .802NRNR41.1p = .002
Doc496.6 (3.6–9.7)NR14.3
Sym 2013 [55]x,aa,asIRI295.8 (3.0–8.7)HR, 1.21 (0.69–2.11); p = .5142.2 (0.2–4.3)HR, 1.20 (0.72–2.02); p = .48117.2p = .525
FOLFIRI306.7 (5.3–8.2)3.0 (2.0–3.7)20.0
WJOG 4007
Hironaka 2013 [53]aa,ab,as
Pac1089.5 (8.4–10.7)HR, 1.13 (0.86–1.49); p = .383.6 (3.3–3.8)HR, 1.14 (0.88–1.49); p = .3320.9p = .24
IRI1118.4 (7.6–9.8)2.3 (2.2–3.1)13.6
Roy 2013 [54]x,aa,asPEP02ac447.3 (3.84–9.17)NR2.7 (1.54–3.65)NR13.6NR
IRI447.8 (4.90–9.20)2.6 (1.48–4.34)6.8
Doc447.7 (5.32–12.32)2.7 (1.41–5.45)15.9
TyTAN
Satoh 2014 [46]x,aa,as
LAP + Pac13211.0adHR, 0.84 (0.64–1.11); p = .10445.5adHR, 0.85 (0.63–1.13); p = .2441ad27adEstimated OR, 3.85 (1.80–8.87); p < .001
Pac1298.9ad4.4ad9ad
COUGAR-02
Ford 2014 [49]at
Doc845.2 (4.1–5.9)HR, 0.67 (0.49–0.92); p = .01NRNRNRNR
Active symptom control843.6 (3.3–4.4)NRNR
TCOG GI-0801/BIRIP
Higuchi 2014 [44]x,aa,as
IRI + Cis6410.7HR, 1.00 (0.69–1.44); p = .98233.8HR, 0.68 (0.47–0.98); p = .039822p = .4975
IRI6310.12.816
REGARD
Fuchs 2014 [42]x,aa,av
RAM2385.2 (IQR, 2.3–9.9)HR, 0.776 (0.603–0.998); p = .0472.1 (IQR, 1.3–4.2)HR, 0.483 (0.376–0.620); p < .00013p = .76
PBO1173.8 (IQR, 1.7–7.1)1.3 (IQR, 1.1–2.1)3
RAINBOW
Wilke 2014 [41]ae,af,av
RAM + Pac3309.6 (8.5–10.8)HR, 0.807 (0.678–0.962); p = .0174.4 (4.2–5.3)HR, 0.635 (0.536–0.752); p < .000128p = .0001
PBO + Pac3357.4 (6.3–8.4)2.9 (2.8–3.0)16
JACCRO GC-05
Tanabe 2015 [56]aa,ag,as
S-1 + IRI1458.8 (IQR, 5.6–15.7)p = .92
HR for death, 0.99 (0.78–1.25)
3.8 (IQR, 1.9–6.6)HR for disease progression or death, 0.85 (0.67–1.07); p = .167.6NS
IRI1489.5 (IQR, 5.6–14.1)3.4 (IQR, 1.6–5.3)7.4
JapicCTI-090849
Satoh 2015 [62]as
Nimotuzumab + IRI40250.5 daysah (171.0–306.0)HR, 0.994 (0.618–1.599)ah; p = .977873.0 days (55.0–112.0)HR, 0.860 (0.516–1.435); p = .566818.4p = .3060
IRI42232.0 daysah (148.0–319.0)85.0 days (37.0–93.0)10.3
Study 39
Bang 2015 [48]ae,ah,au,ax
Olaparib + Pac6213.1HR, 0.56 (0.35–0.87); p = .0103.9HR, 0.80 (80% CI, 0.62–1.03); p = .13126.4NS
PBO + Pac628.33.619.1
TRICS
Nishikawa 2015 [57]ai,as
IRI + Cis8413.9 (10.8–17.6)HR, 0.834 (0.596–1.167); p = .2884.6 (3.4–5.9)HR, 0.860 (0.610–1.203); p = .376NRNR
IRI8412.7 (10.3–17.2)4.1 (3.3–4.9)NR
Kim 2015 [45]x,aeDoc277.2 (6.0–8.4)p = .3532.0 (1.2–2.9)p = .00214.8p = .40
Doc + Ox258.1 (7.6–8.6)4.9 (3.6–6.6)24.0
RAINBOW subgroup analysis (East Asia)
Muro 2016 [63]ae,aj,as
RAM + Pac10912.1HR, 0.986 (0.727–1.337); p = .9295.5HR, 0.628 (0.473–0.834)34OR, 2.24 (1.18–4.24); p = .0134
PBO + Pac11410.52.820
RAINBOW subgroup analysis (Japan)
Shitara 2016 [64]ae,ak,aw
Japanese: RAM + Pac6811.4HR, 0.880 (0.603–1.284); p = .51135.6HR, 0.503 (0.348–0.728); p = .000241.2p = .0035
Japanese: PBO + Pac7211.52.819.4
Western: RAM + Pac1988.6HR, 0.7326 (0.580–0.909); p = .0054.2HR, 0.631 (0.506–0.786); p < .000126.8p = .0004
Western: PBO + Pac2005.92.813.0
CCOG0701
Nakanishi 2016 [58]aa,al,as
Pac4010.0 (0.4–74.1)HR, 0.834 (0.511–1.359)4.6 (0.4–74.1)HR, 0.862 (0.543–1.367); difference NSb27p = .767
Pac + S-14910.0 (1.3–72.0)4.6 (0.4–59.6)22
JCOG0407
Nishina 2016 [50]w,as
Best available FU497.7 (6.7–9.0)HR, 0.89 (0.57–1.38); p = .2982.4 (1.7–3.6)HR, 0.58 (0.38–0.88); p = .005NRNR
Pac517.7 (6.0–9.7)3.7 (2.6–3.7)NR
Bang 2017b [70]w,aeIpilimumab5712.7 (10.5–18.9)NRirPFS: 2.92 (1.61–5.16)
PFS (mWHO criteria): 2.73 (1.45–2.96)
irPFS: HR, 1.44 80% CI, 1.09–1.91); p = .097
mWHO criteria: HR, 1.59 (80% CI, 1.20–2.10); p = .034 (“consistent with irPFS” [improvement was not observed])
1.8NR
BSC5712.1 (9.3–NE)irPFS: 4.90 (3.45–6.54)
PFS (mWHO criteria): 4.90 (3.45–6.08)
7.0NR
GOLD
Bang 2017a [69]x
Olaparib + Pac2638.8 (7.4–9.6)HR, 0.79 (97.5% CI, 0.63–1.00); p = .026 (NS)an3.7 (3.7–4.2)HR, 0.84 (97.5% CI, 0.67–1.04); p = .06517 (adjusted, 24)aeOR, 1.69 (97.5% CI, 0.92–3.17); p = .055
PBO + Pac2626.9 (6.3–7.9)3.2 (2.2–3.5)11 (adjusted, 16)ae
ATM-negative tumors subgroup: Olaparib + Pac4812.0 (7.8–18.1)HR, 0.73 (97.5% CI, 0.40–1.34); p = .255.3 (3.5–9.0)HR, 0.74 (97.5% CI, 0.42–1.29); p = .2225 (adjusted, 38)aeOR, 4.24 (0.95–23.23); p = .031
ATM-negative tumors subgroup: PBO + Pac4610.0 (6.4–13.3)3.7 (1.9–5.3)11 (adjusted, 16)ae
GATSBY
Thuss-Patience 2017 [66]x,ae
Taxane1178.6 (7.1–11.2)HR, 1.15 (0.87–1.51); p = .862.9 mo (2.8–4.0)HR, 1.13 (0.89–1.43); p = .3119.6p = .8406
Tras emtansine2287.9 (6.7–9.5)2.7 (1.6–2.7)20.6
SHINE
Van Cutsem 2017 [72]x,ae
AZD4547415.5 (95% CI, NR)HR, 1.31 (80% CI, 0.89–1.95); p = .81561.8HR, 1.57 (80% CI, 1.12–2.21); p = .95812.6OR, 0.09 (80% CI, 0.02–0.35); p = .9970
Pac306.6 (95% CI, NR)3.523.3
Lee 2017 [116]aaDoc2310.0 (7.8–12.2)All vs. Doc + S-1: p = .023
Doc + S-1 vs. Doc: p = .421
Doc + Cis vs. Doc: p = .035
1.3 (1.0–1.5)All vs. Doc + S-1: p = .072
Doc + Cis vs. Doc: p = .804
Doc + S-1 vs. Doc: p = .072
4.3p > .990 vs. Doc
Doc + Cis235.6 (4.4–6.7)1.8 (0.8–2.9)4.3
Doc + S-1236.9 (2.1–11.7)2.7 (1.0–4.4)8.7
ABSOLUTE
Shitara 2017 [52]
Nab-Pac Q3W24310.3 (8.7–11.4)Nab-Pac Q3W vs. Pac: HR, 1.06 (0.87–1.31); p = .0623.8 (3.5–4.4)Nab-Pac Q3W vs. Pac: HR, 1.03 (0.85–1.24); p = .77825(18.6–33.1); p = .897 vs. Pac
Nab-Pac QW24011.1 (9.9–13.0)Nab-Pac QW vs. Pac: HR, 0.97 (97.5% CI, 0.76–1.23); p = .00855.3 (4.0–5.6)Nab-Pac Q3W vs. Pac: HR, 0.88 (0.73–1.06); p = .17633(25.2–40.8); p = .106 vs. Pac
Pac24310.9 (9.4–11.8)3.8 (3.7–3.9)24(18.0–31.4)
DREAM
Kang 2018 [51]x,ae,ai
DHP107 (oral Pac)1189.7 (7.1–11.5)HR, 1.04 (0.76–1.41); p = .8243.0 (1.7–4.0)HR, 0.85 (0.64–1.13)NRNR
IV Pac1188.9 (7.1–12.2)2.6 (1.8–2.8)NR
KEYNOTE-061
Shitara 2018a [71]ae,ay
Pembrolizumab296NRNRNRNR12.2NR
Pac296NRNRNRNR15.2NR
PD-L1 CPS ≥1 subgroup: Pembrolizumab1969.1 (6.2–10.7)HR, 0.82 (0.66–1.03); p = .0421 (NS)ao1.5 (1.4–2.0)HR, 1.27 (1.03–1.57)16NR
PD-L1 CPS ≥1 subgroup: Pac1998.3 (7.6–9.0)4.1 (3.1–4.2)14NR
KCSG ST10-01
Lee 2019 [117]x,ae,am
Pac548.57 (7.1–10.0)HR, 1.39 (0.91–2.11); p = .1263.47 (2.2–4.7)HR, 1.27 (0.86–1.88); p = .23415.8p = .355
IRI587.03 (5.6–8.4)2.10 (1.4–2.8)13.6
Combined second- and third-line populations
Kang 2012 [61]asDoc or IRI + BSC1335.3 (4.1–6.5)HR, 0.657 (0.485–0.891); p = .007NRNRNRNR
BSC693.8 (3.1–4.5)NRNR
GRANITE-1
Ohtsu 2013 [73]aa,ap,as
Everolimus + BSC4395.4 (4.8–6.0)HR, 0.90 (0.75–1.08); p = .1241.7 (1.5–1.9)HR, 0.66 (0.56–0.78); p < .0014.5(95% CI for the rate, 2.6–7.1)
PBO + BSC2174.3 (3.8–5.5)1.4 (1.4–1.5)2.1(95% CI for the rate, 0.6–5.3)
Shitara 2014 [59]ae,aq,asDose-escalated Pac4411.8 (7.6–16.3)HR, 0.75 (0.45–1.22); p = .124.3 (3.0–5.7)HR, 0.55 (0.34–0.90); p = .01730.3(95% CI for the rate, 15.6–48.7)
Pac459.6 (7.4–11.7)2.5 (1.8–3.7)17.1(95% CI for the rate, 6.6–33.7)
Fushida 2016 [60]ae,asPac339.8HR, 1.19 (0.702–2.026); p = .514.5HR, 1.29 (0.753–2.211); p = .35NRNR
Pac + valproic acid318.73.0NR
Moehler 2016 [118]x,asNa-FOLFIRI + sunitinib4510.4 (4.5–10.9)HR, 0.82 (0.50–1.34); p = .423.5 (1.4–5.6)HR, 1.11 (0.70–1.74); p = .6620NR
Na-FOLFIRI + placebo458.9 (5.9–11.8)3.3 (1.5–5.2)29NR
INTEGRATE
Pavlakis 2016 [74]ae,ar,as
Regorafenib + BSC975.8 (4.4–6.8)HR, 0.74 (0.51–1.08); p = .1472.6 (1.8–3.1)HR, 0.40 (0.28–0.59); p < .0013.0(95% CI for the rate, 1–9)
Placebo + BSC504.5 (3.4–5.2)0.9 (0.9–0.9)2(95% CI for the rate, 0–11)
Third-line and beyond studies
Li 2013 [77]Apatinib 850 mg once daily474.83 (4.03–5.97)HR, 0.37 (0.22–0.62); p < .0013.67 (2.17–6.80)HR, 0.18 (0.10–0.34); p < .001)0(95% CI for the rate, 1.3–17.5)
Apatinib 425 mg twice daily464.27 (3.83–4.77)HR, 0.41 (0.24–0.72); p = .00173.20 (2.37–4.53)HR, 0.21 (0.11–0.38); p < .0016.38(95% CI for the rate, 4.9–26.3)
PBO482.5 (1.87–3.70)1.40 (1.20–1.83)13.04(95% CI for the rate, 0.0–7.4)
Li 2016 [78]Apatinib1466.5 (4.8–7.6)HR, 0.709 (0.537–0.937); p = .01492.6 (2.0–2.9)HR, 0.444 (0.331–0.595); p < .0012.84 (1.70)p = .1695 (p = .5532)
PBO784.7 (3.6–5.4)1.8 (1.4–1.9)0
ATTRACTION-2
Kang 2017 [75]ay
Nivolumab3305.26 (4.60–6.37)HR, 0.63 (0.51–0.78); p < .00011.61 (1.54–2.30)HR, 0.60 (0.49–0.75); p < .000111.2NR
PBO1634.14 (3.42–4.86)1.45 (1.45–1.54)0
JAVELIN Gastric 300
Bang 2018 [82]ay
Avelumab + BSC1854.6 (3.6–5.7)HR, 1.1 (0.9–1.4); p = .811.4 (1.4–1.5)HR, 1.73 (1.4–2.2); p > .992.2NR
Chemo + BSC1855.0 (4.5–6.3)2.7 (1.8–2.8)4.3
TAGS
Shitara 2018b [76]
Trifluridine/tipiracil + BSC3375.7 (4.8–6.2)HR, 0.69 (0.56–0.85); p = .000292.0 (1.9–2.3)HR, 0.57 (0.47–0.70); p < .00014p = .28
PBO + BSC1703.6 (3.1–4.1)1.8 (1.7–1.9)2
CheckMate-032
Janjigian 2018 [80]ay
Nivolumab 3 mg/kg596.2 (3.4–12.4)NR1.4 (1.2–1.5)NR7NR
Nivolumab 1 mg/kg + ipilimumab 3 mg/kg496.9 (3.7–11.5)1.4 (1.2–3.8)20NR
Nivolumab 3 mg/kg + ipilimumab 1 mg/kg524.8 (3.0–8.4)1.6 (1.4–2.6)4NR
ATTRACTION-2 subgroup analysis (Japanese patients)
Kato 2019 [79]ay
Nivolumab1525.4 (4.6–7.4)HR, 0.58 (0.42–0.78); p = .00021.7 (1.6–2.8)HR, 0.53 (0.39–0.72); p < .000114.0p = .0023
Placebo743.6 (2.8–5.0)1.5 (1.5–1.6)0
ATTRACTION-2 subgroup analysis (prior Tras use)
Satoh 2020 [119]ay
History of Tras: nivolumab598.3 (5.3–12.9)HR, 0.38 (0.22–0.66); p = .00061.6 (1.5–4.0)HR, 0.49 (0.29–0.85); p = .011116.9NR
History of Tras: PBO223.1 (1.9–5.3)1.5 (1.3–2.9)0
No history of Tras: nivolumab2714.8 (4.1–6.0)HR, 0.71 (0.57–0.88); p = .00221.6 (1.5–2.4)HR, 0.64 (0.51–0.80);p = .00017.7NR
No history of Tras: PBO1414.2 (3.6–4.9)1.5 (1.5–1.5)0
TrialTreatmentPatients, nOverall survivalacProgression-free survivalacORRac
Median (95% CI), moEffect size(95% CI)Median (95% CI), moEffect size (95% CI)%Effect size
First-line studies
Kang 2009 [24]d,eCis + Cap13910.4 (9.1–11.0)HR, 0.85 (0.65–1.11); p = .005 vs. noninferiority margin of 1.255.6 (4.8–6.9)HR, 0.80 (0.63–1.03); p < .001 vs. noninferiority margin of 1.25
Cis + FU1378.9 (7.3–10.2)5.0 (3.9–5.7)
FLAGS
Ajani 2010 [26]f,g
S-1 + Cis5278.6HR, 0.92 (0.80–1.05); p = .19834.8 (4.0–5.5)HR, 0.99 (0.86–1.14); p = .915829.1p = .40
FU + Cis5267.95.5 (4.4–5.8)31.9
ToGA
Bang 2010 [17]h,i
Tras + Chemoj29413.8HR, 0.74 (0.60–0.91); p = .00466.7HR, 0.71 (0.59–0.85); p = .000247kp = .0017
Chemo29011.15.535k
AVAGAST
Ohtsu 2011 [25]d,l
BEV + Cis + Cap/FUm25212.1 (11.1–13.8)HR, 0.87 (0.73–1.03); p = .10026.7 (5.9–7.1)HR, 0.80 (0.68–0.98); p = .0037
PBO + Cis + Cap/FUm26510.1 (9.0–11.3)5.3 (4.4–5.6)
REAL3
Waddell 2013 [18]d
Epir + Ox + Cap27511.3 (9.6–13.0)HR, 1.37 (1.07–1.76); p = .0136.0 (5.5–6.5)HR, 1.22 (0.98–1.52); p = .068NRNR
Epir + Ox + Cap + PAN2788.8 (7.7–9.8)7.4 (6.3–8.5)NR
EXPAND
Lordick 2013 [114]d,n
CTX + Cap + Cis4559.4 (8.3–10.6)HR, 1.00 (0.87–1.17); p = .954.4 (4.2–5.5)HR, 1.09 (0.92–1.29); p = .32
Cap + Cis44910.7 (9.4–11.3)5.6 (5.1–5.7)
Van Cutsem 2015 [37]iDoc + Ox798.97 (7.79–10.87)NR4.5 (3.7–5.3)NRNRNR
Doc + Ox + FU/FOL8914.59 (11.70–21.78)7.7 (7.0–9.4)
Doc + Ox + Cap8611.30 (8.08–14.03)5.6 (4.3–6.4)NR
Guimbaud 2014 [22]d,oEpir + Cis + Cap2099.49HR, 1.01 (0.82–1.24); p = .955.3HR, 0.99 (0.81–1.21); p = .9690.4NS
FOLFIRI2079.725.895.7
Kim 2014 [112]d,pSIM + Cap + Cis12011.6 (9.2–13.9)HR, 0.966 (0.722–1.293); p = .8185.2 (4.3–6.1)HR, 0.930 (0.684–1.264); p = .664
PBO + Cap + Cis12411.5 (9.9–13.1)4.6 (3.5–5.7)
SOS
Ryu 2015 [109]h,q
S-1 (D1–14) + Cis (D1)30614.1 (11.4–15.8)HR, 0.99 (0.81–1.21); p = .915.5 (4.7–6.6)HR, 0.82 (0.68–0.99); p = .041860p = .065
S-1 (D1–21) + Cis (D1 or 8)30913.9 (11.6–15.9)4.9 (4.2–5.5)50
AVATAR
Shen 2015 [110]d
PBO + Cap + Cis10211.4 (8.6–16.0)HR, 1.11 (0.79–1.56); p = .566.0 (4.9–7.4)HR, 0.89 (0.66–1.21); p = .47NRNR
BEV + Cap + Cis10010.5 (8.9–14.1)6.3 (5.7–7.4)NRNR
JapicCTI-101021
Yamada 2015 [20]d,r
S-1 + Ox31814.1 (13.0–15.8)HR, 0.958 (0.803–1.142) (S-1 + Ox noninferior to S-1 + Cis)5.5 (4.4–5.7)HR, 1.004 (0.840–1.199); p = .0044 (noninferiority)55.7NR
S-1 + Cis32413.1 (12.1–15.1)5.4 (4.2–5.7)52.2
Li 2015 [113]eS-1 + Cis12010.0 (8.59–14.52)p = .82
FU + Cis11610.46 (8.92–13.84)
Wang 2016 [19]d,sDoc + Cis + FU12110.2 (8.6–11.9)HR, 0.71 (0.52–0.97); p = .03197.2 (5.5–8.8)HR, 0.58 (0.42–0.80); p = .0008 (log-rank test)48.7p = .0244
Cis + FU1228.5 (7.1–9.5)4.9 (4.5–6.0)33.9
G-SOX
Bando 2016 [21]d
S-1 + Ox (≥70 yr)11617.5≥70 yr: HR, 0.857 (0.629–1.167); p = .3255.7HR, 0.805 (0.588–1.102); p = .174NRNR
S-1 + Cis (≥70 yr)10413.55.5NR
S-1 + Ox (<70 yr)22713.3<70 yr: HR, 0.984 (0.800–1.209); p = .8774.4HR, 1.019 (0.827–1.256); p = .862NR
S-1 + Cis (<70 yr)23813.15.3NR
TRIO-013/LOGiC
Hecht 2016 [23]d,n
LAP + Cap + Ox24912.2HR, 0.91 (0.73–1.12); p = .34926.0HR, 0.82 (0.68–1.00); p = .0381NRNR
PBO + Cap + Ox23810.55.4NR
DIGEST
Ajani 2017 [27]
S-1 + Cis2397.5 (6.7–9.3)Unstratified: HR, 0.99 (0.76–1.28); p = .9312
Stratified: HR, 0.90 (0.68–1.19); p = .4631
4.4HR, 0.86 (0.65–1.14); p = .303934.7p = .0122
FU + Cis1226.6 (5.7–8.1)3.919.8
RILOMET-1
Catenacci 2017 [28]
Rilotumumab + Epir + Cis + Cap2628.8 (7.7–10.2)HR, 1.34 (1.10–1.63); p = .0035.6 (5.3–5.9)HR, 1.26 (1.04–1.51); p = .01629.8p = .0005
PBO + Epir + Cis + Cap26710.7 (9.6–12.4)6.0 (5.7–7.2)44.6
METGastric
Shah 2017 [39]
Onartuzumab + mFOLFOX621711.0HR, 0.82 (0.59–1.15); p = .246.7HR, 0.90 (0.71–1.16); p = .4346.1p = .25
PBO + mFOLFOX620711.36.840.6
MET 2+/3+ subgroup:
onartuzumab + mFOLFOX6
7811.0HR, 0.64 (0.40–1.03); p = .066.9HR, 0.79 (0.54–1.15); p = .2253.8p = .23
MET 2+/3+ subgroup:
PBO + mFOLFOX6
929.75.744.6
Shu 2017 [32]Ox + tegafur16413.4 (12.2–15.1)HR, 0.96 (0.80–1.39)5.5 (4.6–6.2)HR, 1.02 (0.82–1.31)44.5p = .702
Ox + Cis16814.2 (13.1–16.0)6.1 (5.6–6.4)48.8
HELOISE
Shah 2017 [115]
SoC Tras + Cap + Cis12412.5HR, 1.24 (0.86–1.78); p = .24015.7HR, 1.04 (0.76–1.40); p = .822258.9p = .76
Higher-dose Tras + Cap + Cis12410.65.656.9
Lu 2018 [29]Pac + Cap16012.5 (11.5–14.5)HR, 0.878 (0.685–1.125); p = .304.994 (4.304–6.275)HR, 0.906 (0.706–1.164); p = .4443.1p = .01
Cis + Cap16011.8 (10.0–13.7)5.257 (4.665–5.815)28.8
Lu 2019 [35]S-1 + Doc150405 daysp = .5127180 daysp > .05NRNR
S-1 + Cis150378 days171 daysNR
RAINFALL
Fuchs 2019 [40]t,u
RAM + Cis + Cap/FUv32611.2 (9.9–11.9)HR, 0.962 (0.801–1.156); p = .68Investigator-assessed: 5.7 (5.5–6.5)
Independent review: 5.5 (4.2–5.8)
Investigator-assessed:
HR, 0.753 (0.607–0.935); p = .011
Independent review:
HR, 0.961 (0.78–1.203); p = .74
41.1p = .17
PBO + Cis + Cap/FUo31910.7 (9.5–11.9)Investigator-assessed: 5.4 (4.5–5.7)
Independent review: 5.4 (4.4–5.7)
36.4
JACOB
Tabernero 2018 [38]
Pertuzumab + Tras/Chemo38817.5 (16.2–19.3)HR,0.84 (0.71–1.00); p = .0578.5 (8.2–9.7)HR, 0.73 (0.62–0.86); p = .000156.7p = .026
PBO + Tras/Chemo39214.2 (12.9–15.5)7.0 (6.4–8.2)48.3
JCOG1013
Yamada 2019 [36]
Doc + Cis/S-137014.2 (12.9–15.9)HR, 0.99 (0.85–1.16); p = .477.4 (6.7–7.8)HR, 0.99 (0.86–1.15); p = .9259p = .50
Cis/S-137115.3 (14.2–16.2)6.5 (5.9–7.4)56
Second-line studies
Thuss-Patience 2011 [47]wy,asIRI + BSC214.0 (3.6–7.5)All patients died:
HR, 0.48 (0.25–0.92); p = .012
ITT population: 2.5 (1.6–3.9)
Per-protocol population: 2.6 (1.7–4.3)
NR (results provided for IRI arm only)0zNR
BSC192.4 (1.7–4.9)NR (results provided for IRI arm only)NR
Yi 2012 [43]x,asDoc + sunitinib568.0 (5.4–10.6)HR, 0.94 (0.60–1.49); p = .802NRNR41.1p = .002
Doc496.6 (3.6–9.7)NR14.3
Sym 2013 [55]x,aa,asIRI295.8 (3.0–8.7)HR, 1.21 (0.69–2.11); p = .5142.2 (0.2–4.3)HR, 1.20 (0.72–2.02); p = .48117.2p = .525
FOLFIRI306.7 (5.3–8.2)3.0 (2.0–3.7)20.0
WJOG 4007
Hironaka 2013 [53]aa,ab,as
Pac1089.5 (8.4–10.7)HR, 1.13 (0.86–1.49); p = .383.6 (3.3–3.8)HR, 1.14 (0.88–1.49); p = .3320.9p = .24
IRI1118.4 (7.6–9.8)2.3 (2.2–3.1)13.6
Roy 2013 [54]x,aa,asPEP02ac447.3 (3.84–9.17)NR2.7 (1.54–3.65)NR13.6NR
IRI447.8 (4.90–9.20)2.6 (1.48–4.34)6.8
Doc447.7 (5.32–12.32)2.7 (1.41–5.45)15.9
TyTAN
Satoh 2014 [46]x,aa,as
LAP + Pac13211.0adHR, 0.84 (0.64–1.11); p = .10445.5adHR, 0.85 (0.63–1.13); p = .2441ad27adEstimated OR, 3.85 (1.80–8.87); p < .001
Pac1298.9ad4.4ad9ad
COUGAR-02
Ford 2014 [49]at
Doc845.2 (4.1–5.9)HR, 0.67 (0.49–0.92); p = .01NRNRNRNR
Active symptom control843.6 (3.3–4.4)NRNR
TCOG GI-0801/BIRIP
Higuchi 2014 [44]x,aa,as
IRI + Cis6410.7HR, 1.00 (0.69–1.44); p = .98233.8HR, 0.68 (0.47–0.98); p = .039822p = .4975
IRI6310.12.816
REGARD
Fuchs 2014 [42]x,aa,av
RAM2385.2 (IQR, 2.3–9.9)HR, 0.776 (0.603–0.998); p = .0472.1 (IQR, 1.3–4.2)HR, 0.483 (0.376–0.620); p < .00013p = .76
PBO1173.8 (IQR, 1.7–7.1)1.3 (IQR, 1.1–2.1)3
RAINBOW
Wilke 2014 [41]ae,af,av
RAM + Pac3309.6 (8.5–10.8)HR, 0.807 (0.678–0.962); p = .0174.4 (4.2–5.3)HR, 0.635 (0.536–0.752); p < .000128p = .0001
PBO + Pac3357.4 (6.3–8.4)2.9 (2.8–3.0)16
JACCRO GC-05
Tanabe 2015 [56]aa,ag,as
S-1 + IRI1458.8 (IQR, 5.6–15.7)p = .92
HR for death, 0.99 (0.78–1.25)
3.8 (IQR, 1.9–6.6)HR for disease progression or death, 0.85 (0.67–1.07); p = .167.6NS
IRI1489.5 (IQR, 5.6–14.1)3.4 (IQR, 1.6–5.3)7.4
JapicCTI-090849
Satoh 2015 [62]as
Nimotuzumab + IRI40250.5 daysah (171.0–306.0)HR, 0.994 (0.618–1.599)ah; p = .977873.0 days (55.0–112.0)HR, 0.860 (0.516–1.435); p = .566818.4p = .3060
IRI42232.0 daysah (148.0–319.0)85.0 days (37.0–93.0)10.3
Study 39
Bang 2015 [48]ae,ah,au,ax
Olaparib + Pac6213.1HR, 0.56 (0.35–0.87); p = .0103.9HR, 0.80 (80% CI, 0.62–1.03); p = .13126.4NS
PBO + Pac628.33.619.1
TRICS
Nishikawa 2015 [57]ai,as
IRI + Cis8413.9 (10.8–17.6)HR, 0.834 (0.596–1.167); p = .2884.6 (3.4–5.9)HR, 0.860 (0.610–1.203); p = .376NRNR
IRI8412.7 (10.3–17.2)4.1 (3.3–4.9)NR
Kim 2015 [45]x,aeDoc277.2 (6.0–8.4)p = .3532.0 (1.2–2.9)p = .00214.8p = .40
Doc + Ox258.1 (7.6–8.6)4.9 (3.6–6.6)24.0
RAINBOW subgroup analysis (East Asia)
Muro 2016 [63]ae,aj,as
RAM + Pac10912.1HR, 0.986 (0.727–1.337); p = .9295.5HR, 0.628 (0.473–0.834)34OR, 2.24 (1.18–4.24); p = .0134
PBO + Pac11410.52.820
RAINBOW subgroup analysis (Japan)
Shitara 2016 [64]ae,ak,aw
Japanese: RAM + Pac6811.4HR, 0.880 (0.603–1.284); p = .51135.6HR, 0.503 (0.348–0.728); p = .000241.2p = .0035
Japanese: PBO + Pac7211.52.819.4
Western: RAM + Pac1988.6HR, 0.7326 (0.580–0.909); p = .0054.2HR, 0.631 (0.506–0.786); p < .000126.8p = .0004
Western: PBO + Pac2005.92.813.0
CCOG0701
Nakanishi 2016 [58]aa,al,as
Pac4010.0 (0.4–74.1)HR, 0.834 (0.511–1.359)4.6 (0.4–74.1)HR, 0.862 (0.543–1.367); difference NSb27p = .767
Pac + S-14910.0 (1.3–72.0)4.6 (0.4–59.6)22
JCOG0407
Nishina 2016 [50]w,as
Best available FU497.7 (6.7–9.0)HR, 0.89 (0.57–1.38); p = .2982.4 (1.7–3.6)HR, 0.58 (0.38–0.88); p = .005NRNR
Pac517.7 (6.0–9.7)3.7 (2.6–3.7)NR
Bang 2017b [70]w,aeIpilimumab5712.7 (10.5–18.9)NRirPFS: 2.92 (1.61–5.16)
PFS (mWHO criteria): 2.73 (1.45–2.96)
irPFS: HR, 1.44 80% CI, 1.09–1.91); p = .097
mWHO criteria: HR, 1.59 (80% CI, 1.20–2.10); p = .034 (“consistent with irPFS” [improvement was not observed])
1.8NR
BSC5712.1 (9.3–NE)irPFS: 4.90 (3.45–6.54)
PFS (mWHO criteria): 4.90 (3.45–6.08)
7.0NR
GOLD
Bang 2017a [69]x
Olaparib + Pac2638.8 (7.4–9.6)HR, 0.79 (97.5% CI, 0.63–1.00); p = .026 (NS)an3.7 (3.7–4.2)HR, 0.84 (97.5% CI, 0.67–1.04); p = .06517 (adjusted, 24)aeOR, 1.69 (97.5% CI, 0.92–3.17); p = .055
PBO + Pac2626.9 (6.3–7.9)3.2 (2.2–3.5)11 (adjusted, 16)ae
ATM-negative tumors subgroup: Olaparib + Pac4812.0 (7.8–18.1)HR, 0.73 (97.5% CI, 0.40–1.34); p = .255.3 (3.5–9.0)HR, 0.74 (97.5% CI, 0.42–1.29); p = .2225 (adjusted, 38)aeOR, 4.24 (0.95–23.23); p = .031
ATM-negative tumors subgroup: PBO + Pac4610.0 (6.4–13.3)3.7 (1.9–5.3)11 (adjusted, 16)ae
GATSBY
Thuss-Patience 2017 [66]x,ae
Taxane1178.6 (7.1–11.2)HR, 1.15 (0.87–1.51); p = .862.9 mo (2.8–4.0)HR, 1.13 (0.89–1.43); p = .3119.6p = .8406
Tras emtansine2287.9 (6.7–9.5)2.7 (1.6–2.7)20.6
SHINE
Van Cutsem 2017 [72]x,ae
AZD4547415.5 (95% CI, NR)HR, 1.31 (80% CI, 0.89–1.95); p = .81561.8HR, 1.57 (80% CI, 1.12–2.21); p = .95812.6OR, 0.09 (80% CI, 0.02–0.35); p = .9970
Pac306.6 (95% CI, NR)3.523.3
Lee 2017 [116]aaDoc2310.0 (7.8–12.2)All vs. Doc + S-1: p = .023
Doc + S-1 vs. Doc: p = .421
Doc + Cis vs. Doc: p = .035
1.3 (1.0–1.5)All vs. Doc + S-1: p = .072
Doc + Cis vs. Doc: p = .804
Doc + S-1 vs. Doc: p = .072
4.3p > .990 vs. Doc
Doc + Cis235.6 (4.4–6.7)1.8 (0.8–2.9)4.3
Doc + S-1236.9 (2.1–11.7)2.7 (1.0–4.4)8.7
ABSOLUTE
Shitara 2017 [52]
Nab-Pac Q3W24310.3 (8.7–11.4)Nab-Pac Q3W vs. Pac: HR, 1.06 (0.87–1.31); p = .0623.8 (3.5–4.4)Nab-Pac Q3W vs. Pac: HR, 1.03 (0.85–1.24); p = .77825(18.6–33.1); p = .897 vs. Pac
Nab-Pac QW24011.1 (9.9–13.0)Nab-Pac QW vs. Pac: HR, 0.97 (97.5% CI, 0.76–1.23); p = .00855.3 (4.0–5.6)Nab-Pac Q3W vs. Pac: HR, 0.88 (0.73–1.06); p = .17633(25.2–40.8); p = .106 vs. Pac
Pac24310.9 (9.4–11.8)3.8 (3.7–3.9)24(18.0–31.4)
DREAM
Kang 2018 [51]x,ae,ai
DHP107 (oral Pac)1189.7 (7.1–11.5)HR, 1.04 (0.76–1.41); p = .8243.0 (1.7–4.0)HR, 0.85 (0.64–1.13)NRNR
IV Pac1188.9 (7.1–12.2)2.6 (1.8–2.8)NR
KEYNOTE-061
Shitara 2018a [71]ae,ay
Pembrolizumab296NRNRNRNR12.2NR
Pac296NRNRNRNR15.2NR
PD-L1 CPS ≥1 subgroup: Pembrolizumab1969.1 (6.2–10.7)HR, 0.82 (0.66–1.03); p = .0421 (NS)ao1.5 (1.4–2.0)HR, 1.27 (1.03–1.57)16NR
PD-L1 CPS ≥1 subgroup: Pac1998.3 (7.6–9.0)4.1 (3.1–4.2)14NR
KCSG ST10-01
Lee 2019 [117]x,ae,am
Pac548.57 (7.1–10.0)HR, 1.39 (0.91–2.11); p = .1263.47 (2.2–4.7)HR, 1.27 (0.86–1.88); p = .23415.8p = .355
IRI587.03 (5.6–8.4)2.10 (1.4–2.8)13.6
Combined second- and third-line populations
Kang 2012 [61]asDoc or IRI + BSC1335.3 (4.1–6.5)HR, 0.657 (0.485–0.891); p = .007NRNRNRNR
BSC693.8 (3.1–4.5)NRNR
GRANITE-1
Ohtsu 2013 [73]aa,ap,as
Everolimus + BSC4395.4 (4.8–6.0)HR, 0.90 (0.75–1.08); p = .1241.7 (1.5–1.9)HR, 0.66 (0.56–0.78); p < .0014.5(95% CI for the rate, 2.6–7.1)
PBO + BSC2174.3 (3.8–5.5)1.4 (1.4–1.5)2.1(95% CI for the rate, 0.6–5.3)
Shitara 2014 [59]ae,aq,asDose-escalated Pac4411.8 (7.6–16.3)HR, 0.75 (0.45–1.22); p = .124.3 (3.0–5.7)HR, 0.55 (0.34–0.90); p = .01730.3(95% CI for the rate, 15.6–48.7)
Pac459.6 (7.4–11.7)2.5 (1.8–3.7)17.1(95% CI for the rate, 6.6–33.7)
Fushida 2016 [60]ae,asPac339.8HR, 1.19 (0.702–2.026); p = .514.5HR, 1.29 (0.753–2.211); p = .35NRNR
Pac + valproic acid318.73.0NR
Moehler 2016 [118]x,asNa-FOLFIRI + sunitinib4510.4 (4.5–10.9)HR, 0.82 (0.50–1.34); p = .423.5 (1.4–5.6)HR, 1.11 (0.70–1.74); p = .6620NR
Na-FOLFIRI + placebo458.9 (5.9–11.8)3.3 (1.5–5.2)29NR
INTEGRATE
Pavlakis 2016 [74]ae,ar,as
Regorafenib + BSC975.8 (4.4–6.8)HR, 0.74 (0.51–1.08); p = .1472.6 (1.8–3.1)HR, 0.40 (0.28–0.59); p < .0013.0(95% CI for the rate, 1–9)
Placebo + BSC504.5 (3.4–5.2)0.9 (0.9–0.9)2(95% CI for the rate, 0–11)
Third-line and beyond studies
Li 2013 [77]Apatinib 850 mg once daily474.83 (4.03–5.97)HR, 0.37 (0.22–0.62); p < .0013.67 (2.17–6.80)HR, 0.18 (0.10–0.34); p < .001)0(95% CI for the rate, 1.3–17.5)
Apatinib 425 mg twice daily464.27 (3.83–4.77)HR, 0.41 (0.24–0.72); p = .00173.20 (2.37–4.53)HR, 0.21 (0.11–0.38); p < .0016.38(95% CI for the rate, 4.9–26.3)
PBO482.5 (1.87–3.70)1.40 (1.20–1.83)13.04(95% CI for the rate, 0.0–7.4)
Li 2016 [78]Apatinib1466.5 (4.8–7.6)HR, 0.709 (0.537–0.937); p = .01492.6 (2.0–2.9)HR, 0.444 (0.331–0.595); p < .0012.84 (1.70)p = .1695 (p = .5532)
PBO784.7 (3.6–5.4)1.8 (1.4–1.9)0
ATTRACTION-2
Kang 2017 [75]ay
Nivolumab3305.26 (4.60–6.37)HR, 0.63 (0.51–0.78); p < .00011.61 (1.54–2.30)HR, 0.60 (0.49–0.75); p < .000111.2NR
PBO1634.14 (3.42–4.86)1.45 (1.45–1.54)0
JAVELIN Gastric 300
Bang 2018 [82]ay
Avelumab + BSC1854.6 (3.6–5.7)HR, 1.1 (0.9–1.4); p = .811.4 (1.4–1.5)HR, 1.73 (1.4–2.2); p > .992.2NR
Chemo + BSC1855.0 (4.5–6.3)2.7 (1.8–2.8)4.3
TAGS
Shitara 2018b [76]
Trifluridine/tipiracil + BSC3375.7 (4.8–6.2)HR, 0.69 (0.56–0.85); p = .000292.0 (1.9–2.3)HR, 0.57 (0.47–0.70); p < .00014p = .28
PBO + BSC1703.6 (3.1–4.1)1.8 (1.7–1.9)2
CheckMate-032
Janjigian 2018 [80]ay
Nivolumab 3 mg/kg596.2 (3.4–12.4)NR1.4 (1.2–1.5)NR7NR
Nivolumab 1 mg/kg + ipilimumab 3 mg/kg496.9 (3.7–11.5)1.4 (1.2–3.8)20NR
Nivolumab 3 mg/kg + ipilimumab 1 mg/kg524.8 (3.0–8.4)1.6 (1.4–2.6)4NR
ATTRACTION-2 subgroup analysis (Japanese patients)
Kato 2019 [79]ay
Nivolumab1525.4 (4.6–7.4)HR, 0.58 (0.42–0.78); p = .00021.7 (1.6–2.8)HR, 0.53 (0.39–0.72); p < .000114.0p = .0023
Placebo743.6 (2.8–5.0)1.5 (1.5–1.6)0
ATTRACTION-2 subgroup analysis (prior Tras use)
Satoh 2020 [119]ay
History of Tras: nivolumab598.3 (5.3–12.9)HR, 0.38 (0.22–0.66); p = .00061.6 (1.5–4.0)HR, 0.49 (0.29–0.85); p = .011116.9NR
History of Tras: PBO223.1 (1.9–5.3)1.5 (1.3–2.9)0
No history of Tras: nivolumab2714.8 (4.1–6.0)HR, 0.71 (0.57–0.88); p = .00221.6 (1.5–2.4)HR, 0.64 (0.51–0.80);p = .00017.7NR
No history of Tras: PBO1414.2 (3.6–4.9)1.5 (1.5–1.5)0

aIn Ajani et al. (2010) [26], Ryu et al. (2015) [109], Waddell et al. (2013) [18], Wang et al. (2016) [19], Bando et al. (2016) [21], Bang et al. (2010) [17], Guimbaud et al. (2014) [22], Hecht et al. (2016) [23], Li et al. (2015) [113], and Lordick et al. (2013) [114], data were not reported as being either adjusted or unadjusted.

bIn Shen et al. (2015) [110], Van Cutsem et al. (2015) [37], Yamada et al. (2015) [20], and Kang et al. (2009) [24], unadjusted data are presented for OS and PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

cIn Kim et al. (2014) [112] and Ohtsu et al. (2011) [25], unadjusted data are presented for OS, PFS, and response; for other endpoints, data were not reported as being either adjusted or unadjusted.

dIn Shen et al. (2015) [110], Waddell et al. (2013) [18], Wang et al. (2016) [19], Yamada et al. (2015) [20], Bando et al. (2016) [21], Guimbaud et al. (2014) [22], Hecht et al. (2016) [23], Kang et al. (2009) [24], Kim et al. (2014) [112], Lordick et al. (2013) [114], and Ohtsu et al. (2011) [25], RECIST version 1.0 criteria were used.

eIn Kang et al. (2009) [24] and Li et al. (2015) [113], only patients with measurable disease were included.

fIn Ajani et al. (2010) [26], RECIST criteria were used, but no version number was provided.

gIn Ajani et al. (2010) [26], patients were stratified based on whether disease was measurable (95.6% of patients had measurable disease).

hIn Bang et al. (2010) [17] and Van Cutsem et al. (2015) [37], there was no indication that RECIST criteria were used.

iIn Bang et al. (2010) [17], patients were stratified based on whether disease was measurable (~90% of patients had measurable disease).

jCap plus Cis or FU plus Cis, chosen at the investigator's discretion.

k“Overall tumor response rate” (complete response plus partial response).

lIn Ohtsu et al. (2011) [25], the measurable disease population was used to evaluate response rate (~79% of patients had measurable disease).

mPatients unable to take oral medications received FU. Switching from Cap to FU during the study was not permitted.

nIn Hecht et al. (2016) [23] and Lordick et al. (2013) [114], no information was provided regarding the handling of data from patients with measurable versus nonmeasurable disease.

oIn Guimbaud et al. (2014) [22], patients were stratified based on whether disease was measurable (the proportion of patients with measurable disease was not provided).

pIn Kim et al. (2014) [112], patients were stratified based on whether disease was measurable (~64% of patients had measurable disease).

qIn Ryu et al. (2015) [109], response rate was calculated only for patients with measurable disease (~61% of patients).

rIn Yamada et al. (2015) [20], those with no measurable disease were excluded from the per-protocol population (~2.5% of patients).

sIn Wang et al. (2016) [19], those with no measurable disease were excluded from the efficacy population (~1.3% of patients).

tIn Ryu et al. (2015) [109] and Fuchs et al. (2019) [40], RECIST version 1.1 criteria were used.

uIn Fuchs et al. (2019) [40], adjusted data were presented for PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

vFU IV infusion was permitted in patients unable to take oral capecitabine.

wIn Thuss-Patience et al. (2011) [47], Nishina et al. (2016) [50], and Bang et al. (2017b) [70], there was no indication that RECIST criteria were used.

xIn Satoh et al. (2015) [62], Satoh et al. (2014) [46], Sym et al. (2013) [55], Thuss-Patience et al. (2011) [47], Yi et al. (2012) [43], Higuchi et al. (2014) [44], Fuchs et al. (2014) [42], Kim et al. (2015) [45], Roy et al. (2013) [54], Bang et al. (2017a) [69], Kang et al. (2018) [51], Thuss-Patience et al. (2017) [66], Van Cutsem et al. (2017) [72], Lee et al. (2019) [117], and Moehler et al. (2016) [118], no information was provided regarding the handling of data from patients with measurable versus nonmeasurable disease.

yIn Thuss-Patience et al. (2011) [47], there was no indication that RECIST criteria were used.

zNo objective remission according to World Health Organization criteria.

aaIn Satoh et al. (2015) [62], Satoh et al. (2014) [46], Sym et al. (2013) [55], Tanabe et al. (2015) [56], Higuchi et al. (2014) [44], Fuchs et al. (2014) [42], Hironaka et al. (2013) [53], Nakanishi et al. (2016) [58], Roy et al. (2013) [54], Lee et al. (2017) [116], and Ohtsu et al. (2013) [73], RECIST version 1.0 criteria were used.

abIn Hironaka et al. (2013) [53], response rate was assessed in patients with measurable disease at baseline (~80% of patients).

acPEP02 is a highly stable liposomal nanocarrier formulation of irinotecan.

adITT population. Similar results were obtained for the modified ITT population (i.e., randomly assigned patients confirmed FISH positive by central laboratory).

aeIn Bang et al. (2015) [48], Wilke et al. (2014) [41], Muro et al. (2016) [63], Shitara et al. (2016) [64], Bang et al. (2017a) [69], Kang et al. (2018) [51], Kim et al. (2015) [45], Shitara et al. (2018b) [76], Thuss-Patience et al. (2017) [66], Van Cutsem et al. (2017) [72], Lee et al. (2019) [117], Fushida et al. (2016) [60], Pavlakis et al. (2016) [74], and Shitara et al. (2014) [59], RECIST version 1.1 criteria were used.

afIn Wilke et al. (2014) [41], patients were stratified based on whether disease was measurable (~81% of patients had measurable disease).

agIn Tanabe et al. (2015) [56], response rate was calculated only for patients with measurable disease (~82% of patients had measurable disease).

ahEighteen-month OS.

aiIn Nishikawa et al. (2015) [57] and Kang et al. (2012) [61], RECIST criteria were used, but no version number was provided.

ajIn Muro et al. (2016) [63], patients were stratified based on whether disease was measurable (~72% of East Asian patients and ~ 81% of non–East Asian patients had measurable disease).

akIn Shitara et al. (2016) [64], patients were stratified based on whether disease was measurable (72.1% of Japanese patients and 83.4% of Western patients had measurable disease).

alIn Nakanishi et al. (2016) [58], measurable disease was an adjustment factor during randomization (~42% of patients had measurable disease).

amIn Lee et al. (2019) [117], response evaluation was conducted on patients with at least one measurable lesion (38/54 in the paclitaxel group and 44/58 in the irinotecan group).

anIn the GOLD study (Bang et al. [2017a] [69]), statistical significance was set at p < .025.

aoIn the KEYNOTE-061 study (Shitara et al. [2018a] [71]), statistical significance was set at p < .0215.

apIn Ohtsu et al. (2013) [73], objective response of complete response or partial response, only assessed in the patients with measurable disease at baseline (approximately 86% in the everolimus plus BSC group and 88% in the placebo plus BSC group).

aqIn Shitara et al. (2014) [59], objective response of complete response or partial response, only assessed in the patients with measurable disease at baseline (78% in the weekly paclitaxel group and 75% in the dose-escalated weekly paclitaxel group).

arIn Pavlakis et al. (2016) [74], only patients with measurable disease were included in the study.

asIn Satoh et al. (2015) [62], Satoh et al. (2014) [46], Sym et al. (2013) [55], Tanabe et al. (2015) [56], Thuss-Patience et al. (2011) [47], Yi et al. (2012) [43], Higuchi et al. (2014) [44], Muro et al. (2016) [63], Hironaka et al. (2013) [53], Kim et al. (2015) [45], Nakanishi et al. (2016) [58], Nishina et al. (2016) [50], Roy et al. (2013) [54], Nishikawa et al. (2015) [50], Fushida et al. (2016) [60], Kang et al. (2012) [61], Moehler et al. (2016) [28], Ohtsu et al. (2013) [73], Pavlakis et al. (2016) [74], and Shitara et al. (2014) [59], data were not reported as being either adjusted or unadjusted.

atIn Ford et al. (2014) [49], adjusted data are presented for OS; for other endpoints, data were not reported as being either adjusted or unadjusted.

auIn Bang et al. (2015) [48], adjusted data are presented for OS and PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

avIn Fuchs et al. (2014) [42] and Wilke et al. (2014) [41], unadjusted data are presented for OS and PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

awIn Shitara et al. (2016) [64], adjusted data are presented for ORR and disease control rate; for other endpoints, data were not reported as being either adjusted or unadjusted.

axIn Bang et al. (2015) [48], response rate was calculated only for patients with measurable disease (~81% of patients).

ayIn Bang et al. (2018) [82], Kang et al. (2017) [75], Kato et al. (2019) [79], Shitara et al. (2018a) [71], and Janjigian et al. (2018) [80], data were not reported as being either adjusted or unadjusted. In Satoh et al. (2020) [119], adjusted data are presented for ORR and disease control rate; for other endpoints, data were not reported as being either adjusted or unadjusted.

Abbreviations: ATM, ataxia telangiectasia mutated; BEV, bevacizumab; BSC, best supportive care; Cap, capecitabine; Chemo, chemotherapy; CI, confidence interval; Cis, cisplatin; CPS, combined positive score; CTX, cetuximab; D, day; Doc, docetaxel; Epir, epirubicin; FOL, folinic acid FOLFIRI, irinotecan plus 5-fluorouracil plus folinic acid (also known as leucovorin); FU, fluorouracil; HR, hazard ratio; IQR, interquartile range; IRI, irinotecan; irPFS, immune-related progression-free survival; ITT, intent-to-treat; IV, intravenous; LAP, lapatinib; MET, mesenchymal-epithelial transition; mWHO, modified World Health Organization; nab-Pac, nab-paclitaxel; NR, not reported; NS, not significant; OR, odds ratio; ORR, overall response rate; Ox, oxaliplatin; Pac, paclitaxel; PAN, panitumumab; PBO, placebo; PD-L1, programmed death-ligand 1; PFS, progression-free survival; QW, once weekly; Q3W, once every 3 weeks; RAM, ramucirumab; SIM, simvastatin; SoC, standard of care; Tras, trastuzumab.

Table 2

Overall survival, progression-free survival, and overall response rate of included first-, second-, or third-line and beyond randomized controlled trials ordered by study publication year

TrialTreatmentPatients, nOverall survivalacProgression-free survivalacORRac
Median (95% CI), moEffect size(95% CI)Median (95% CI), moEffect size (95% CI)%Effect size
First-line studies
Kang 2009 [24]d,eCis + Cap13910.4 (9.1–11.0)HR, 0.85 (0.65–1.11); p = .005 vs. noninferiority margin of 1.255.6 (4.8–6.9)HR, 0.80 (0.63–1.03); p < .001 vs. noninferiority margin of 1.25
Cis + FU1378.9 (7.3–10.2)5.0 (3.9–5.7)
FLAGS
Ajani 2010 [26]f,g
S-1 + Cis5278.6HR, 0.92 (0.80–1.05); p = .19834.8 (4.0–5.5)HR, 0.99 (0.86–1.14); p = .915829.1p = .40
FU + Cis5267.95.5 (4.4–5.8)31.9
ToGA
Bang 2010 [17]h,i
Tras + Chemoj29413.8HR, 0.74 (0.60–0.91); p = .00466.7HR, 0.71 (0.59–0.85); p = .000247kp = .0017
Chemo29011.15.535k
AVAGAST
Ohtsu 2011 [25]d,l
BEV + Cis + Cap/FUm25212.1 (11.1–13.8)HR, 0.87 (0.73–1.03); p = .10026.7 (5.9–7.1)HR, 0.80 (0.68–0.98); p = .0037
PBO + Cis + Cap/FUm26510.1 (9.0–11.3)5.3 (4.4–5.6)
REAL3
Waddell 2013 [18]d
Epir + Ox + Cap27511.3 (9.6–13.0)HR, 1.37 (1.07–1.76); p = .0136.0 (5.5–6.5)HR, 1.22 (0.98–1.52); p = .068NRNR
Epir + Ox + Cap + PAN2788.8 (7.7–9.8)7.4 (6.3–8.5)NR
EXPAND
Lordick 2013 [114]d,n
CTX + Cap + Cis4559.4 (8.3–10.6)HR, 1.00 (0.87–1.17); p = .954.4 (4.2–5.5)HR, 1.09 (0.92–1.29); p = .32
Cap + Cis44910.7 (9.4–11.3)5.6 (5.1–5.7)
Van Cutsem 2015 [37]iDoc + Ox798.97 (7.79–10.87)NR4.5 (3.7–5.3)NRNRNR
Doc + Ox + FU/FOL8914.59 (11.70–21.78)7.7 (7.0–9.4)
Doc + Ox + Cap8611.30 (8.08–14.03)5.6 (4.3–6.4)NR
Guimbaud 2014 [22]d,oEpir + Cis + Cap2099.49HR, 1.01 (0.82–1.24); p = .955.3HR, 0.99 (0.81–1.21); p = .9690.4NS
FOLFIRI2079.725.895.7
Kim 2014 [112]d,pSIM + Cap + Cis12011.6 (9.2–13.9)HR, 0.966 (0.722–1.293); p = .8185.2 (4.3–6.1)HR, 0.930 (0.684–1.264); p = .664
PBO + Cap + Cis12411.5 (9.9–13.1)4.6 (3.5–5.7)
SOS
Ryu 2015 [109]h,q
S-1 (D1–14) + Cis (D1)30614.1 (11.4–15.8)HR, 0.99 (0.81–1.21); p = .915.5 (4.7–6.6)HR, 0.82 (0.68–0.99); p = .041860p = .065
S-1 (D1–21) + Cis (D1 or 8)30913.9 (11.6–15.9)4.9 (4.2–5.5)50
AVATAR
Shen 2015 [110]d
PBO + Cap + Cis10211.4 (8.6–16.0)HR, 1.11 (0.79–1.56); p = .566.0 (4.9–7.4)HR, 0.89 (0.66–1.21); p = .47NRNR
BEV + Cap + Cis10010.5 (8.9–14.1)6.3 (5.7–7.4)NRNR
JapicCTI-101021
Yamada 2015 [20]d,r
S-1 + Ox31814.1 (13.0–15.8)HR, 0.958 (0.803–1.142) (S-1 + Ox noninferior to S-1 + Cis)5.5 (4.4–5.7)HR, 1.004 (0.840–1.199); p = .0044 (noninferiority)55.7NR
S-1 + Cis32413.1 (12.1–15.1)5.4 (4.2–5.7)52.2
Li 2015 [113]eS-1 + Cis12010.0 (8.59–14.52)p = .82
FU + Cis11610.46 (8.92–13.84)
Wang 2016 [19]d,sDoc + Cis + FU12110.2 (8.6–11.9)HR, 0.71 (0.52–0.97); p = .03197.2 (5.5–8.8)HR, 0.58 (0.42–0.80); p = .0008 (log-rank test)48.7p = .0244
Cis + FU1228.5 (7.1–9.5)4.9 (4.5–6.0)33.9
G-SOX
Bando 2016 [21]d
S-1 + Ox (≥70 yr)11617.5≥70 yr: HR, 0.857 (0.629–1.167); p = .3255.7HR, 0.805 (0.588–1.102); p = .174NRNR
S-1 + Cis (≥70 yr)10413.55.5NR
S-1 + Ox (<70 yr)22713.3<70 yr: HR, 0.984 (0.800–1.209); p = .8774.4HR, 1.019 (0.827–1.256); p = .862NR
S-1 + Cis (<70 yr)23813.15.3NR
TRIO-013/LOGiC
Hecht 2016 [23]d,n
LAP + Cap + Ox24912.2HR, 0.91 (0.73–1.12); p = .34926.0HR, 0.82 (0.68–1.00); p = .0381NRNR
PBO + Cap + Ox23810.55.4NR
DIGEST
Ajani 2017 [27]
S-1 + Cis2397.5 (6.7–9.3)Unstratified: HR, 0.99 (0.76–1.28); p = .9312
Stratified: HR, 0.90 (0.68–1.19); p = .4631
4.4HR, 0.86 (0.65–1.14); p = .303934.7p = .0122
FU + Cis1226.6 (5.7–8.1)3.919.8
RILOMET-1
Catenacci 2017 [28]
Rilotumumab + Epir + Cis + Cap2628.8 (7.7–10.2)HR, 1.34 (1.10–1.63); p = .0035.6 (5.3–5.9)HR, 1.26 (1.04–1.51); p = .01629.8p = .0005
PBO + Epir + Cis + Cap26710.7 (9.6–12.4)6.0 (5.7–7.2)44.6
METGastric
Shah 2017 [39]
Onartuzumab + mFOLFOX621711.0HR, 0.82 (0.59–1.15); p = .246.7HR, 0.90 (0.71–1.16); p = .4346.1p = .25
PBO + mFOLFOX620711.36.840.6
MET 2+/3+ subgroup:
onartuzumab + mFOLFOX6
7811.0HR, 0.64 (0.40–1.03); p = .066.9HR, 0.79 (0.54–1.15); p = .2253.8p = .23
MET 2+/3+ subgroup:
PBO + mFOLFOX6
929.75.744.6
Shu 2017 [32]Ox + tegafur16413.4 (12.2–15.1)HR, 0.96 (0.80–1.39)5.5 (4.6–6.2)HR, 1.02 (0.82–1.31)44.5p = .702
Ox + Cis16814.2 (13.1–16.0)6.1 (5.6–6.4)48.8
HELOISE
Shah 2017 [115]
SoC Tras + Cap + Cis12412.5HR, 1.24 (0.86–1.78); p = .24015.7HR, 1.04 (0.76–1.40); p = .822258.9p = .76
Higher-dose Tras + Cap + Cis12410.65.656.9
Lu 2018 [29]Pac + Cap16012.5 (11.5–14.5)HR, 0.878 (0.685–1.125); p = .304.994 (4.304–6.275)HR, 0.906 (0.706–1.164); p = .4443.1p = .01
Cis + Cap16011.8 (10.0–13.7)5.257 (4.665–5.815)28.8
Lu 2019 [35]S-1 + Doc150405 daysp = .5127180 daysp > .05NRNR
S-1 + Cis150378 days171 daysNR
RAINFALL
Fuchs 2019 [40]t,u
RAM + Cis + Cap/FUv32611.2 (9.9–11.9)HR, 0.962 (0.801–1.156); p = .68Investigator-assessed: 5.7 (5.5–6.5)
Independent review: 5.5 (4.2–5.8)
Investigator-assessed:
HR, 0.753 (0.607–0.935); p = .011
Independent review:
HR, 0.961 (0.78–1.203); p = .74
41.1p = .17
PBO + Cis + Cap/FUo31910.7 (9.5–11.9)Investigator-assessed: 5.4 (4.5–5.7)
Independent review: 5.4 (4.4–5.7)
36.4
JACOB
Tabernero 2018 [38]
Pertuzumab + Tras/Chemo38817.5 (16.2–19.3)HR,0.84 (0.71–1.00); p = .0578.5 (8.2–9.7)HR, 0.73 (0.62–0.86); p = .000156.7p = .026
PBO + Tras/Chemo39214.2 (12.9–15.5)7.0 (6.4–8.2)48.3
JCOG1013
Yamada 2019 [36]
Doc + Cis/S-137014.2 (12.9–15.9)HR, 0.99 (0.85–1.16); p = .477.4 (6.7–7.8)HR, 0.99 (0.86–1.15); p = .9259p = .50
Cis/S-137115.3 (14.2–16.2)6.5 (5.9–7.4)56
Second-line studies
Thuss-Patience 2011 [47]wy,asIRI + BSC214.0 (3.6–7.5)All patients died:
HR, 0.48 (0.25–0.92); p = .012
ITT population: 2.5 (1.6–3.9)
Per-protocol population: 2.6 (1.7–4.3)
NR (results provided for IRI arm only)0zNR
BSC192.4 (1.7–4.9)NR (results provided for IRI arm only)NR
Yi 2012 [43]x,asDoc + sunitinib568.0 (5.4–10.6)HR, 0.94 (0.60–1.49); p = .802NRNR41.1p = .002
Doc496.6 (3.6–9.7)NR14.3
Sym 2013 [55]x,aa,asIRI295.8 (3.0–8.7)HR, 1.21 (0.69–2.11); p = .5142.2 (0.2–4.3)HR, 1.20 (0.72–2.02); p = .48117.2p = .525
FOLFIRI306.7 (5.3–8.2)3.0 (2.0–3.7)20.0
WJOG 4007
Hironaka 2013 [53]aa,ab,as
Pac1089.5 (8.4–10.7)HR, 1.13 (0.86–1.49); p = .383.6 (3.3–3.8)HR, 1.14 (0.88–1.49); p = .3320.9p = .24
IRI1118.4 (7.6–9.8)2.3 (2.2–3.1)13.6
Roy 2013 [54]x,aa,asPEP02ac447.3 (3.84–9.17)NR2.7 (1.54–3.65)NR13.6NR
IRI447.8 (4.90–9.20)2.6 (1.48–4.34)6.8
Doc447.7 (5.32–12.32)2.7 (1.41–5.45)15.9
TyTAN
Satoh 2014 [46]x,aa,as
LAP + Pac13211.0adHR, 0.84 (0.64–1.11); p = .10445.5adHR, 0.85 (0.63–1.13); p = .2441ad27adEstimated OR, 3.85 (1.80–8.87); p < .001
Pac1298.9ad4.4ad9ad
COUGAR-02
Ford 2014 [49]at
Doc845.2 (4.1–5.9)HR, 0.67 (0.49–0.92); p = .01NRNRNRNR
Active symptom control843.6 (3.3–4.4)NRNR
TCOG GI-0801/BIRIP
Higuchi 2014 [44]x,aa,as
IRI + Cis6410.7HR, 1.00 (0.69–1.44); p = .98233.8HR, 0.68 (0.47–0.98); p = .039822p = .4975
IRI6310.12.816
REGARD
Fuchs 2014 [42]x,aa,av
RAM2385.2 (IQR, 2.3–9.9)HR, 0.776 (0.603–0.998); p = .0472.1 (IQR, 1.3–4.2)HR, 0.483 (0.376–0.620); p < .00013p = .76
PBO1173.8 (IQR, 1.7–7.1)1.3 (IQR, 1.1–2.1)3
RAINBOW
Wilke 2014 [41]ae,af,av
RAM + Pac3309.6 (8.5–10.8)HR, 0.807 (0.678–0.962); p = .0174.4 (4.2–5.3)HR, 0.635 (0.536–0.752); p < .000128p = .0001
PBO + Pac3357.4 (6.3–8.4)2.9 (2.8–3.0)16
JACCRO GC-05
Tanabe 2015 [56]aa,ag,as
S-1 + IRI1458.8 (IQR, 5.6–15.7)p = .92
HR for death, 0.99 (0.78–1.25)
3.8 (IQR, 1.9–6.6)HR for disease progression or death, 0.85 (0.67–1.07); p = .167.6NS
IRI1489.5 (IQR, 5.6–14.1)3.4 (IQR, 1.6–5.3)7.4
JapicCTI-090849
Satoh 2015 [62]as
Nimotuzumab + IRI40250.5 daysah (171.0–306.0)HR, 0.994 (0.618–1.599)ah; p = .977873.0 days (55.0–112.0)HR, 0.860 (0.516–1.435); p = .566818.4p = .3060
IRI42232.0 daysah (148.0–319.0)85.0 days (37.0–93.0)10.3
Study 39
Bang 2015 [48]ae,ah,au,ax
Olaparib + Pac6213.1HR, 0.56 (0.35–0.87); p = .0103.9HR, 0.80 (80% CI, 0.62–1.03); p = .13126.4NS
PBO + Pac628.33.619.1
TRICS
Nishikawa 2015 [57]ai,as
IRI + Cis8413.9 (10.8–17.6)HR, 0.834 (0.596–1.167); p = .2884.6 (3.4–5.9)HR, 0.860 (0.610–1.203); p = .376NRNR
IRI8412.7 (10.3–17.2)4.1 (3.3–4.9)NR
Kim 2015 [45]x,aeDoc277.2 (6.0–8.4)p = .3532.0 (1.2–2.9)p = .00214.8p = .40
Doc + Ox258.1 (7.6–8.6)4.9 (3.6–6.6)24.0
RAINBOW subgroup analysis (East Asia)
Muro 2016 [63]ae,aj,as
RAM + Pac10912.1HR, 0.986 (0.727–1.337); p = .9295.5HR, 0.628 (0.473–0.834)34OR, 2.24 (1.18–4.24); p = .0134
PBO + Pac11410.52.820
RAINBOW subgroup analysis (Japan)
Shitara 2016 [64]ae,ak,aw
Japanese: RAM + Pac6811.4HR, 0.880 (0.603–1.284); p = .51135.6HR, 0.503 (0.348–0.728); p = .000241.2p = .0035
Japanese: PBO + Pac7211.52.819.4
Western: RAM + Pac1988.6HR, 0.7326 (0.580–0.909); p = .0054.2HR, 0.631 (0.506–0.786); p < .000126.8p = .0004
Western: PBO + Pac2005.92.813.0
CCOG0701
Nakanishi 2016 [58]aa,al,as
Pac4010.0 (0.4–74.1)HR, 0.834 (0.511–1.359)4.6 (0.4–74.1)HR, 0.862 (0.543–1.367); difference NSb27p = .767
Pac + S-14910.0 (1.3–72.0)4.6 (0.4–59.6)22
JCOG0407
Nishina 2016 [50]w,as
Best available FU497.7 (6.7–9.0)HR, 0.89 (0.57–1.38); p = .2982.4 (1.7–3.6)HR, 0.58 (0.38–0.88); p = .005NRNR
Pac517.7 (6.0–9.7)3.7 (2.6–3.7)NR
Bang 2017b [70]w,aeIpilimumab5712.7 (10.5–18.9)NRirPFS: 2.92 (1.61–5.16)
PFS (mWHO criteria): 2.73 (1.45–2.96)
irPFS: HR, 1.44 80% CI, 1.09–1.91); p = .097
mWHO criteria: HR, 1.59 (80% CI, 1.20–2.10); p = .034 (“consistent with irPFS” [improvement was not observed])
1.8NR
BSC5712.1 (9.3–NE)irPFS: 4.90 (3.45–6.54)
PFS (mWHO criteria): 4.90 (3.45–6.08)
7.0NR
GOLD
Bang 2017a [69]x
Olaparib + Pac2638.8 (7.4–9.6)HR, 0.79 (97.5% CI, 0.63–1.00); p = .026 (NS)an3.7 (3.7–4.2)HR, 0.84 (97.5% CI, 0.67–1.04); p = .06517 (adjusted, 24)aeOR, 1.69 (97.5% CI, 0.92–3.17); p = .055
PBO + Pac2626.9 (6.3–7.9)3.2 (2.2–3.5)11 (adjusted, 16)ae
ATM-negative tumors subgroup: Olaparib + Pac4812.0 (7.8–18.1)HR, 0.73 (97.5% CI, 0.40–1.34); p = .255.3 (3.5–9.0)HR, 0.74 (97.5% CI, 0.42–1.29); p = .2225 (adjusted, 38)aeOR, 4.24 (0.95–23.23); p = .031
ATM-negative tumors subgroup: PBO + Pac4610.0 (6.4–13.3)3.7 (1.9–5.3)11 (adjusted, 16)ae
GATSBY
Thuss-Patience 2017 [66]x,ae
Taxane1178.6 (7.1–11.2)HR, 1.15 (0.87–1.51); p = .862.9 mo (2.8–4.0)HR, 1.13 (0.89–1.43); p = .3119.6p = .8406
Tras emtansine2287.9 (6.7–9.5)2.7 (1.6–2.7)20.6
SHINE
Van Cutsem 2017 [72]x,ae
AZD4547415.5 (95% CI, NR)HR, 1.31 (80% CI, 0.89–1.95); p = .81561.8HR, 1.57 (80% CI, 1.12–2.21); p = .95812.6OR, 0.09 (80% CI, 0.02–0.35); p = .9970
Pac306.6 (95% CI, NR)3.523.3
Lee 2017 [116]aaDoc2310.0 (7.8–12.2)All vs. Doc + S-1: p = .023
Doc + S-1 vs. Doc: p = .421
Doc + Cis vs. Doc: p = .035
1.3 (1.0–1.5)All vs. Doc + S-1: p = .072
Doc + Cis vs. Doc: p = .804
Doc + S-1 vs. Doc: p = .072
4.3p > .990 vs. Doc
Doc + Cis235.6 (4.4–6.7)1.8 (0.8–2.9)4.3
Doc + S-1236.9 (2.1–11.7)2.7 (1.0–4.4)8.7
ABSOLUTE
Shitara 2017 [52]
Nab-Pac Q3W24310.3 (8.7–11.4)Nab-Pac Q3W vs. Pac: HR, 1.06 (0.87–1.31); p = .0623.8 (3.5–4.4)Nab-Pac Q3W vs. Pac: HR, 1.03 (0.85–1.24); p = .77825(18.6–33.1); p = .897 vs. Pac
Nab-Pac QW24011.1 (9.9–13.0)Nab-Pac QW vs. Pac: HR, 0.97 (97.5% CI, 0.76–1.23); p = .00855.3 (4.0–5.6)Nab-Pac Q3W vs. Pac: HR, 0.88 (0.73–1.06); p = .17633(25.2–40.8); p = .106 vs. Pac
Pac24310.9 (9.4–11.8)3.8 (3.7–3.9)24(18.0–31.4)
DREAM
Kang 2018 [51]x,ae,ai
DHP107 (oral Pac)1189.7 (7.1–11.5)HR, 1.04 (0.76–1.41); p = .8243.0 (1.7–4.0)HR, 0.85 (0.64–1.13)NRNR
IV Pac1188.9 (7.1–12.2)2.6 (1.8–2.8)NR
KEYNOTE-061
Shitara 2018a [71]ae,ay
Pembrolizumab296NRNRNRNR12.2NR
Pac296NRNRNRNR15.2NR
PD-L1 CPS ≥1 subgroup: Pembrolizumab1969.1 (6.2–10.7)HR, 0.82 (0.66–1.03); p = .0421 (NS)ao1.5 (1.4–2.0)HR, 1.27 (1.03–1.57)16NR
PD-L1 CPS ≥1 subgroup: Pac1998.3 (7.6–9.0)4.1 (3.1–4.2)14NR
KCSG ST10-01
Lee 2019 [117]x,ae,am
Pac548.57 (7.1–10.0)HR, 1.39 (0.91–2.11); p = .1263.47 (2.2–4.7)HR, 1.27 (0.86–1.88); p = .23415.8p = .355
IRI587.03 (5.6–8.4)2.10 (1.4–2.8)13.6
Combined second- and third-line populations
Kang 2012 [61]asDoc or IRI + BSC1335.3 (4.1–6.5)HR, 0.657 (0.485–0.891); p = .007NRNRNRNR
BSC693.8 (3.1–4.5)NRNR
GRANITE-1
Ohtsu 2013 [73]aa,ap,as
Everolimus + BSC4395.4 (4.8–6.0)HR, 0.90 (0.75–1.08); p = .1241.7 (1.5–1.9)HR, 0.66 (0.56–0.78); p < .0014.5(95% CI for the rate, 2.6–7.1)
PBO + BSC2174.3 (3.8–5.5)1.4 (1.4–1.5)2.1(95% CI for the rate, 0.6–5.3)
Shitara 2014 [59]ae,aq,asDose-escalated Pac4411.8 (7.6–16.3)HR, 0.75 (0.45–1.22); p = .124.3 (3.0–5.7)HR, 0.55 (0.34–0.90); p = .01730.3(95% CI for the rate, 15.6–48.7)
Pac459.6 (7.4–11.7)2.5 (1.8–3.7)17.1(95% CI for the rate, 6.6–33.7)
Fushida 2016 [60]ae,asPac339.8HR, 1.19 (0.702–2.026); p = .514.5HR, 1.29 (0.753–2.211); p = .35NRNR
Pac + valproic acid318.73.0NR
Moehler 2016 [118]x,asNa-FOLFIRI + sunitinib4510.4 (4.5–10.9)HR, 0.82 (0.50–1.34); p = .423.5 (1.4–5.6)HR, 1.11 (0.70–1.74); p = .6620NR
Na-FOLFIRI + placebo458.9 (5.9–11.8)3.3 (1.5–5.2)29NR
INTEGRATE
Pavlakis 2016 [74]ae,ar,as
Regorafenib + BSC975.8 (4.4–6.8)HR, 0.74 (0.51–1.08); p = .1472.6 (1.8–3.1)HR, 0.40 (0.28–0.59); p < .0013.0(95% CI for the rate, 1–9)
Placebo + BSC504.5 (3.4–5.2)0.9 (0.9–0.9)2(95% CI for the rate, 0–11)
Third-line and beyond studies
Li 2013 [77]Apatinib 850 mg once daily474.83 (4.03–5.97)HR, 0.37 (0.22–0.62); p < .0013.67 (2.17–6.80)HR, 0.18 (0.10–0.34); p < .001)0(95% CI for the rate, 1.3–17.5)
Apatinib 425 mg twice daily464.27 (3.83–4.77)HR, 0.41 (0.24–0.72); p = .00173.20 (2.37–4.53)HR, 0.21 (0.11–0.38); p < .0016.38(95% CI for the rate, 4.9–26.3)
PBO482.5 (1.87–3.70)1.40 (1.20–1.83)13.04(95% CI for the rate, 0.0–7.4)
Li 2016 [78]Apatinib1466.5 (4.8–7.6)HR, 0.709 (0.537–0.937); p = .01492.6 (2.0–2.9)HR, 0.444 (0.331–0.595); p < .0012.84 (1.70)p = .1695 (p = .5532)
PBO784.7 (3.6–5.4)1.8 (1.4–1.9)0
ATTRACTION-2
Kang 2017 [75]ay
Nivolumab3305.26 (4.60–6.37)HR, 0.63 (0.51–0.78); p < .00011.61 (1.54–2.30)HR, 0.60 (0.49–0.75); p < .000111.2NR
PBO1634.14 (3.42–4.86)1.45 (1.45–1.54)0
JAVELIN Gastric 300
Bang 2018 [82]ay
Avelumab + BSC1854.6 (3.6–5.7)HR, 1.1 (0.9–1.4); p = .811.4 (1.4–1.5)HR, 1.73 (1.4–2.2); p > .992.2NR
Chemo + BSC1855.0 (4.5–6.3)2.7 (1.8–2.8)4.3
TAGS
Shitara 2018b [76]
Trifluridine/tipiracil + BSC3375.7 (4.8–6.2)HR, 0.69 (0.56–0.85); p = .000292.0 (1.9–2.3)HR, 0.57 (0.47–0.70); p < .00014p = .28
PBO + BSC1703.6 (3.1–4.1)1.8 (1.7–1.9)2
CheckMate-032
Janjigian 2018 [80]ay
Nivolumab 3 mg/kg596.2 (3.4–12.4)NR1.4 (1.2–1.5)NR7NR
Nivolumab 1 mg/kg + ipilimumab 3 mg/kg496.9 (3.7–11.5)1.4 (1.2–3.8)20NR
Nivolumab 3 mg/kg + ipilimumab 1 mg/kg524.8 (3.0–8.4)1.6 (1.4–2.6)4NR
ATTRACTION-2 subgroup analysis (Japanese patients)
Kato 2019 [79]ay
Nivolumab1525.4 (4.6–7.4)HR, 0.58 (0.42–0.78); p = .00021.7 (1.6–2.8)HR, 0.53 (0.39–0.72); p < .000114.0p = .0023
Placebo743.6 (2.8–5.0)1.5 (1.5–1.6)0
ATTRACTION-2 subgroup analysis (prior Tras use)
Satoh 2020 [119]ay
History of Tras: nivolumab598.3 (5.3–12.9)HR, 0.38 (0.22–0.66); p = .00061.6 (1.5–4.0)HR, 0.49 (0.29–0.85); p = .011116.9NR
History of Tras: PBO223.1 (1.9–5.3)1.5 (1.3–2.9)0
No history of Tras: nivolumab2714.8 (4.1–6.0)HR, 0.71 (0.57–0.88); p = .00221.6 (1.5–2.4)HR, 0.64 (0.51–0.80);p = .00017.7NR
No history of Tras: PBO1414.2 (3.6–4.9)1.5 (1.5–1.5)0
TrialTreatmentPatients, nOverall survivalacProgression-free survivalacORRac
Median (95% CI), moEffect size(95% CI)Median (95% CI), moEffect size (95% CI)%Effect size
First-line studies
Kang 2009 [24]d,eCis + Cap13910.4 (9.1–11.0)HR, 0.85 (0.65–1.11); p = .005 vs. noninferiority margin of 1.255.6 (4.8–6.9)HR, 0.80 (0.63–1.03); p < .001 vs. noninferiority margin of 1.25
Cis + FU1378.9 (7.3–10.2)5.0 (3.9–5.7)
FLAGS
Ajani 2010 [26]f,g
S-1 + Cis5278.6HR, 0.92 (0.80–1.05); p = .19834.8 (4.0–5.5)HR, 0.99 (0.86–1.14); p = .915829.1p = .40
FU + Cis5267.95.5 (4.4–5.8)31.9
ToGA
Bang 2010 [17]h,i
Tras + Chemoj29413.8HR, 0.74 (0.60–0.91); p = .00466.7HR, 0.71 (0.59–0.85); p = .000247kp = .0017
Chemo29011.15.535k
AVAGAST
Ohtsu 2011 [25]d,l
BEV + Cis + Cap/FUm25212.1 (11.1–13.8)HR, 0.87 (0.73–1.03); p = .10026.7 (5.9–7.1)HR, 0.80 (0.68–0.98); p = .0037
PBO + Cis + Cap/FUm26510.1 (9.0–11.3)5.3 (4.4–5.6)
REAL3
Waddell 2013 [18]d
Epir + Ox + Cap27511.3 (9.6–13.0)HR, 1.37 (1.07–1.76); p = .0136.0 (5.5–6.5)HR, 1.22 (0.98–1.52); p = .068NRNR
Epir + Ox + Cap + PAN2788.8 (7.7–9.8)7.4 (6.3–8.5)NR
EXPAND
Lordick 2013 [114]d,n
CTX + Cap + Cis4559.4 (8.3–10.6)HR, 1.00 (0.87–1.17); p = .954.4 (4.2–5.5)HR, 1.09 (0.92–1.29); p = .32
Cap + Cis44910.7 (9.4–11.3)5.6 (5.1–5.7)
Van Cutsem 2015 [37]iDoc + Ox798.97 (7.79–10.87)NR4.5 (3.7–5.3)NRNRNR
Doc + Ox + FU/FOL8914.59 (11.70–21.78)7.7 (7.0–9.4)
Doc + Ox + Cap8611.30 (8.08–14.03)5.6 (4.3–6.4)NR
Guimbaud 2014 [22]d,oEpir + Cis + Cap2099.49HR, 1.01 (0.82–1.24); p = .955.3HR, 0.99 (0.81–1.21); p = .9690.4NS
FOLFIRI2079.725.895.7
Kim 2014 [112]d,pSIM + Cap + Cis12011.6 (9.2–13.9)HR, 0.966 (0.722–1.293); p = .8185.2 (4.3–6.1)HR, 0.930 (0.684–1.264); p = .664
PBO + Cap + Cis12411.5 (9.9–13.1)4.6 (3.5–5.7)
SOS
Ryu 2015 [109]h,q
S-1 (D1–14) + Cis (D1)30614.1 (11.4–15.8)HR, 0.99 (0.81–1.21); p = .915.5 (4.7–6.6)HR, 0.82 (0.68–0.99); p = .041860p = .065
S-1 (D1–21) + Cis (D1 or 8)30913.9 (11.6–15.9)4.9 (4.2–5.5)50
AVATAR
Shen 2015 [110]d
PBO + Cap + Cis10211.4 (8.6–16.0)HR, 1.11 (0.79–1.56); p = .566.0 (4.9–7.4)HR, 0.89 (0.66–1.21); p = .47NRNR
BEV + Cap + Cis10010.5 (8.9–14.1)6.3 (5.7–7.4)NRNR
JapicCTI-101021
Yamada 2015 [20]d,r
S-1 + Ox31814.1 (13.0–15.8)HR, 0.958 (0.803–1.142) (S-1 + Ox noninferior to S-1 + Cis)5.5 (4.4–5.7)HR, 1.004 (0.840–1.199); p = .0044 (noninferiority)55.7NR
S-1 + Cis32413.1 (12.1–15.1)5.4 (4.2–5.7)52.2
Li 2015 [113]eS-1 + Cis12010.0 (8.59–14.52)p = .82
FU + Cis11610.46 (8.92–13.84)
Wang 2016 [19]d,sDoc + Cis + FU12110.2 (8.6–11.9)HR, 0.71 (0.52–0.97); p = .03197.2 (5.5–8.8)HR, 0.58 (0.42–0.80); p = .0008 (log-rank test)48.7p = .0244
Cis + FU1228.5 (7.1–9.5)4.9 (4.5–6.0)33.9
G-SOX
Bando 2016 [21]d
S-1 + Ox (≥70 yr)11617.5≥70 yr: HR, 0.857 (0.629–1.167); p = .3255.7HR, 0.805 (0.588–1.102); p = .174NRNR
S-1 + Cis (≥70 yr)10413.55.5NR
S-1 + Ox (<70 yr)22713.3<70 yr: HR, 0.984 (0.800–1.209); p = .8774.4HR, 1.019 (0.827–1.256); p = .862NR
S-1 + Cis (<70 yr)23813.15.3NR
TRIO-013/LOGiC
Hecht 2016 [23]d,n
LAP + Cap + Ox24912.2HR, 0.91 (0.73–1.12); p = .34926.0HR, 0.82 (0.68–1.00); p = .0381NRNR
PBO + Cap + Ox23810.55.4NR
DIGEST
Ajani 2017 [27]
S-1 + Cis2397.5 (6.7–9.3)Unstratified: HR, 0.99 (0.76–1.28); p = .9312
Stratified: HR, 0.90 (0.68–1.19); p = .4631
4.4HR, 0.86 (0.65–1.14); p = .303934.7p = .0122
FU + Cis1226.6 (5.7–8.1)3.919.8
RILOMET-1
Catenacci 2017 [28]
Rilotumumab + Epir + Cis + Cap2628.8 (7.7–10.2)HR, 1.34 (1.10–1.63); p = .0035.6 (5.3–5.9)HR, 1.26 (1.04–1.51); p = .01629.8p = .0005
PBO + Epir + Cis + Cap26710.7 (9.6–12.4)6.0 (5.7–7.2)44.6
METGastric
Shah 2017 [39]
Onartuzumab + mFOLFOX621711.0HR, 0.82 (0.59–1.15); p = .246.7HR, 0.90 (0.71–1.16); p = .4346.1p = .25
PBO + mFOLFOX620711.36.840.6
MET 2+/3+ subgroup:
onartuzumab + mFOLFOX6
7811.0HR, 0.64 (0.40–1.03); p = .066.9HR, 0.79 (0.54–1.15); p = .2253.8p = .23
MET 2+/3+ subgroup:
PBO + mFOLFOX6
929.75.744.6
Shu 2017 [32]Ox + tegafur16413.4 (12.2–15.1)HR, 0.96 (0.80–1.39)5.5 (4.6–6.2)HR, 1.02 (0.82–1.31)44.5p = .702
Ox + Cis16814.2 (13.1–16.0)6.1 (5.6–6.4)48.8
HELOISE
Shah 2017 [115]
SoC Tras + Cap + Cis12412.5HR, 1.24 (0.86–1.78); p = .24015.7HR, 1.04 (0.76–1.40); p = .822258.9p = .76
Higher-dose Tras + Cap + Cis12410.65.656.9
Lu 2018 [29]Pac + Cap16012.5 (11.5–14.5)HR, 0.878 (0.685–1.125); p = .304.994 (4.304–6.275)HR, 0.906 (0.706–1.164); p = .4443.1p = .01
Cis + Cap16011.8 (10.0–13.7)5.257 (4.665–5.815)28.8
Lu 2019 [35]S-1 + Doc150405 daysp = .5127180 daysp > .05NRNR
S-1 + Cis150378 days171 daysNR
RAINFALL
Fuchs 2019 [40]t,u
RAM + Cis + Cap/FUv32611.2 (9.9–11.9)HR, 0.962 (0.801–1.156); p = .68Investigator-assessed: 5.7 (5.5–6.5)
Independent review: 5.5 (4.2–5.8)
Investigator-assessed:
HR, 0.753 (0.607–0.935); p = .011
Independent review:
HR, 0.961 (0.78–1.203); p = .74
41.1p = .17
PBO + Cis + Cap/FUo31910.7 (9.5–11.9)Investigator-assessed: 5.4 (4.5–5.7)
Independent review: 5.4 (4.4–5.7)
36.4
JACOB
Tabernero 2018 [38]
Pertuzumab + Tras/Chemo38817.5 (16.2–19.3)HR,0.84 (0.71–1.00); p = .0578.5 (8.2–9.7)HR, 0.73 (0.62–0.86); p = .000156.7p = .026
PBO + Tras/Chemo39214.2 (12.9–15.5)7.0 (6.4–8.2)48.3
JCOG1013
Yamada 2019 [36]
Doc + Cis/S-137014.2 (12.9–15.9)HR, 0.99 (0.85–1.16); p = .477.4 (6.7–7.8)HR, 0.99 (0.86–1.15); p = .9259p = .50
Cis/S-137115.3 (14.2–16.2)6.5 (5.9–7.4)56
Second-line studies
Thuss-Patience 2011 [47]wy,asIRI + BSC214.0 (3.6–7.5)All patients died:
HR, 0.48 (0.25–0.92); p = .012
ITT population: 2.5 (1.6–3.9)
Per-protocol population: 2.6 (1.7–4.3)
NR (results provided for IRI arm only)0zNR
BSC192.4 (1.7–4.9)NR (results provided for IRI arm only)NR
Yi 2012 [43]x,asDoc + sunitinib568.0 (5.4–10.6)HR, 0.94 (0.60–1.49); p = .802NRNR41.1p = .002
Doc496.6 (3.6–9.7)NR14.3
Sym 2013 [55]x,aa,asIRI295.8 (3.0–8.7)HR, 1.21 (0.69–2.11); p = .5142.2 (0.2–4.3)HR, 1.20 (0.72–2.02); p = .48117.2p = .525
FOLFIRI306.7 (5.3–8.2)3.0 (2.0–3.7)20.0
WJOG 4007
Hironaka 2013 [53]aa,ab,as
Pac1089.5 (8.4–10.7)HR, 1.13 (0.86–1.49); p = .383.6 (3.3–3.8)HR, 1.14 (0.88–1.49); p = .3320.9p = .24
IRI1118.4 (7.6–9.8)2.3 (2.2–3.1)13.6
Roy 2013 [54]x,aa,asPEP02ac447.3 (3.84–9.17)NR2.7 (1.54–3.65)NR13.6NR
IRI447.8 (4.90–9.20)2.6 (1.48–4.34)6.8
Doc447.7 (5.32–12.32)2.7 (1.41–5.45)15.9
TyTAN
Satoh 2014 [46]x,aa,as
LAP + Pac13211.0adHR, 0.84 (0.64–1.11); p = .10445.5adHR, 0.85 (0.63–1.13); p = .2441ad27adEstimated OR, 3.85 (1.80–8.87); p < .001
Pac1298.9ad4.4ad9ad
COUGAR-02
Ford 2014 [49]at
Doc845.2 (4.1–5.9)HR, 0.67 (0.49–0.92); p = .01NRNRNRNR
Active symptom control843.6 (3.3–4.4)NRNR
TCOG GI-0801/BIRIP
Higuchi 2014 [44]x,aa,as
IRI + Cis6410.7HR, 1.00 (0.69–1.44); p = .98233.8HR, 0.68 (0.47–0.98); p = .039822p = .4975
IRI6310.12.816
REGARD
Fuchs 2014 [42]x,aa,av
RAM2385.2 (IQR, 2.3–9.9)HR, 0.776 (0.603–0.998); p = .0472.1 (IQR, 1.3–4.2)HR, 0.483 (0.376–0.620); p < .00013p = .76
PBO1173.8 (IQR, 1.7–7.1)1.3 (IQR, 1.1–2.1)3
RAINBOW
Wilke 2014 [41]ae,af,av
RAM + Pac3309.6 (8.5–10.8)HR, 0.807 (0.678–0.962); p = .0174.4 (4.2–5.3)HR, 0.635 (0.536–0.752); p < .000128p = .0001
PBO + Pac3357.4 (6.3–8.4)2.9 (2.8–3.0)16
JACCRO GC-05
Tanabe 2015 [56]aa,ag,as
S-1 + IRI1458.8 (IQR, 5.6–15.7)p = .92
HR for death, 0.99 (0.78–1.25)
3.8 (IQR, 1.9–6.6)HR for disease progression or death, 0.85 (0.67–1.07); p = .167.6NS
IRI1489.5 (IQR, 5.6–14.1)3.4 (IQR, 1.6–5.3)7.4
JapicCTI-090849
Satoh 2015 [62]as
Nimotuzumab + IRI40250.5 daysah (171.0–306.0)HR, 0.994 (0.618–1.599)ah; p = .977873.0 days (55.0–112.0)HR, 0.860 (0.516–1.435); p = .566818.4p = .3060
IRI42232.0 daysah (148.0–319.0)85.0 days (37.0–93.0)10.3
Study 39
Bang 2015 [48]ae,ah,au,ax
Olaparib + Pac6213.1HR, 0.56 (0.35–0.87); p = .0103.9HR, 0.80 (80% CI, 0.62–1.03); p = .13126.4NS
PBO + Pac628.33.619.1
TRICS
Nishikawa 2015 [57]ai,as
IRI + Cis8413.9 (10.8–17.6)HR, 0.834 (0.596–1.167); p = .2884.6 (3.4–5.9)HR, 0.860 (0.610–1.203); p = .376NRNR
IRI8412.7 (10.3–17.2)4.1 (3.3–4.9)NR
Kim 2015 [45]x,aeDoc277.2 (6.0–8.4)p = .3532.0 (1.2–2.9)p = .00214.8p = .40
Doc + Ox258.1 (7.6–8.6)4.9 (3.6–6.6)24.0
RAINBOW subgroup analysis (East Asia)
Muro 2016 [63]ae,aj,as
RAM + Pac10912.1HR, 0.986 (0.727–1.337); p = .9295.5HR, 0.628 (0.473–0.834)34OR, 2.24 (1.18–4.24); p = .0134
PBO + Pac11410.52.820
RAINBOW subgroup analysis (Japan)
Shitara 2016 [64]ae,ak,aw
Japanese: RAM + Pac6811.4HR, 0.880 (0.603–1.284); p = .51135.6HR, 0.503 (0.348–0.728); p = .000241.2p = .0035
Japanese: PBO + Pac7211.52.819.4
Western: RAM + Pac1988.6HR, 0.7326 (0.580–0.909); p = .0054.2HR, 0.631 (0.506–0.786); p < .000126.8p = .0004
Western: PBO + Pac2005.92.813.0
CCOG0701
Nakanishi 2016 [58]aa,al,as
Pac4010.0 (0.4–74.1)HR, 0.834 (0.511–1.359)4.6 (0.4–74.1)HR, 0.862 (0.543–1.367); difference NSb27p = .767
Pac + S-14910.0 (1.3–72.0)4.6 (0.4–59.6)22
JCOG0407
Nishina 2016 [50]w,as
Best available FU497.7 (6.7–9.0)HR, 0.89 (0.57–1.38); p = .2982.4 (1.7–3.6)HR, 0.58 (0.38–0.88); p = .005NRNR
Pac517.7 (6.0–9.7)3.7 (2.6–3.7)NR
Bang 2017b [70]w,aeIpilimumab5712.7 (10.5–18.9)NRirPFS: 2.92 (1.61–5.16)
PFS (mWHO criteria): 2.73 (1.45–2.96)
irPFS: HR, 1.44 80% CI, 1.09–1.91); p = .097
mWHO criteria: HR, 1.59 (80% CI, 1.20–2.10); p = .034 (“consistent with irPFS” [improvement was not observed])
1.8NR
BSC5712.1 (9.3–NE)irPFS: 4.90 (3.45–6.54)
PFS (mWHO criteria): 4.90 (3.45–6.08)
7.0NR
GOLD
Bang 2017a [69]x
Olaparib + Pac2638.8 (7.4–9.6)HR, 0.79 (97.5% CI, 0.63–1.00); p = .026 (NS)an3.7 (3.7–4.2)HR, 0.84 (97.5% CI, 0.67–1.04); p = .06517 (adjusted, 24)aeOR, 1.69 (97.5% CI, 0.92–3.17); p = .055
PBO + Pac2626.9 (6.3–7.9)3.2 (2.2–3.5)11 (adjusted, 16)ae
ATM-negative tumors subgroup: Olaparib + Pac4812.0 (7.8–18.1)HR, 0.73 (97.5% CI, 0.40–1.34); p = .255.3 (3.5–9.0)HR, 0.74 (97.5% CI, 0.42–1.29); p = .2225 (adjusted, 38)aeOR, 4.24 (0.95–23.23); p = .031
ATM-negative tumors subgroup: PBO + Pac4610.0 (6.4–13.3)3.7 (1.9–5.3)11 (adjusted, 16)ae
GATSBY
Thuss-Patience 2017 [66]x,ae
Taxane1178.6 (7.1–11.2)HR, 1.15 (0.87–1.51); p = .862.9 mo (2.8–4.0)HR, 1.13 (0.89–1.43); p = .3119.6p = .8406
Tras emtansine2287.9 (6.7–9.5)2.7 (1.6–2.7)20.6
SHINE
Van Cutsem 2017 [72]x,ae
AZD4547415.5 (95% CI, NR)HR, 1.31 (80% CI, 0.89–1.95); p = .81561.8HR, 1.57 (80% CI, 1.12–2.21); p = .95812.6OR, 0.09 (80% CI, 0.02–0.35); p = .9970
Pac306.6 (95% CI, NR)3.523.3
Lee 2017 [116]aaDoc2310.0 (7.8–12.2)All vs. Doc + S-1: p = .023
Doc + S-1 vs. Doc: p = .421
Doc + Cis vs. Doc: p = .035
1.3 (1.0–1.5)All vs. Doc + S-1: p = .072
Doc + Cis vs. Doc: p = .804
Doc + S-1 vs. Doc: p = .072
4.3p > .990 vs. Doc
Doc + Cis235.6 (4.4–6.7)1.8 (0.8–2.9)4.3
Doc + S-1236.9 (2.1–11.7)2.7 (1.0–4.4)8.7
ABSOLUTE
Shitara 2017 [52]
Nab-Pac Q3W24310.3 (8.7–11.4)Nab-Pac Q3W vs. Pac: HR, 1.06 (0.87–1.31); p = .0623.8 (3.5–4.4)Nab-Pac Q3W vs. Pac: HR, 1.03 (0.85–1.24); p = .77825(18.6–33.1); p = .897 vs. Pac
Nab-Pac QW24011.1 (9.9–13.0)Nab-Pac QW vs. Pac: HR, 0.97 (97.5% CI, 0.76–1.23); p = .00855.3 (4.0–5.6)Nab-Pac Q3W vs. Pac: HR, 0.88 (0.73–1.06); p = .17633(25.2–40.8); p = .106 vs. Pac
Pac24310.9 (9.4–11.8)3.8 (3.7–3.9)24(18.0–31.4)
DREAM
Kang 2018 [51]x,ae,ai
DHP107 (oral Pac)1189.7 (7.1–11.5)HR, 1.04 (0.76–1.41); p = .8243.0 (1.7–4.0)HR, 0.85 (0.64–1.13)NRNR
IV Pac1188.9 (7.1–12.2)2.6 (1.8–2.8)NR
KEYNOTE-061
Shitara 2018a [71]ae,ay
Pembrolizumab296NRNRNRNR12.2NR
Pac296NRNRNRNR15.2NR
PD-L1 CPS ≥1 subgroup: Pembrolizumab1969.1 (6.2–10.7)HR, 0.82 (0.66–1.03); p = .0421 (NS)ao1.5 (1.4–2.0)HR, 1.27 (1.03–1.57)16NR
PD-L1 CPS ≥1 subgroup: Pac1998.3 (7.6–9.0)4.1 (3.1–4.2)14NR
KCSG ST10-01
Lee 2019 [117]x,ae,am
Pac548.57 (7.1–10.0)HR, 1.39 (0.91–2.11); p = .1263.47 (2.2–4.7)HR, 1.27 (0.86–1.88); p = .23415.8p = .355
IRI587.03 (5.6–8.4)2.10 (1.4–2.8)13.6
Combined second- and third-line populations
Kang 2012 [61]asDoc or IRI + BSC1335.3 (4.1–6.5)HR, 0.657 (0.485–0.891); p = .007NRNRNRNR
BSC693.8 (3.1–4.5)NRNR
GRANITE-1
Ohtsu 2013 [73]aa,ap,as
Everolimus + BSC4395.4 (4.8–6.0)HR, 0.90 (0.75–1.08); p = .1241.7 (1.5–1.9)HR, 0.66 (0.56–0.78); p < .0014.5(95% CI for the rate, 2.6–7.1)
PBO + BSC2174.3 (3.8–5.5)1.4 (1.4–1.5)2.1(95% CI for the rate, 0.6–5.3)
Shitara 2014 [59]ae,aq,asDose-escalated Pac4411.8 (7.6–16.3)HR, 0.75 (0.45–1.22); p = .124.3 (3.0–5.7)HR, 0.55 (0.34–0.90); p = .01730.3(95% CI for the rate, 15.6–48.7)
Pac459.6 (7.4–11.7)2.5 (1.8–3.7)17.1(95% CI for the rate, 6.6–33.7)
Fushida 2016 [60]ae,asPac339.8HR, 1.19 (0.702–2.026); p = .514.5HR, 1.29 (0.753–2.211); p = .35NRNR
Pac + valproic acid318.73.0NR
Moehler 2016 [118]x,asNa-FOLFIRI + sunitinib4510.4 (4.5–10.9)HR, 0.82 (0.50–1.34); p = .423.5 (1.4–5.6)HR, 1.11 (0.70–1.74); p = .6620NR
Na-FOLFIRI + placebo458.9 (5.9–11.8)3.3 (1.5–5.2)29NR
INTEGRATE
Pavlakis 2016 [74]ae,ar,as
Regorafenib + BSC975.8 (4.4–6.8)HR, 0.74 (0.51–1.08); p = .1472.6 (1.8–3.1)HR, 0.40 (0.28–0.59); p < .0013.0(95% CI for the rate, 1–9)
Placebo + BSC504.5 (3.4–5.2)0.9 (0.9–0.9)2(95% CI for the rate, 0–11)
Third-line and beyond studies
Li 2013 [77]Apatinib 850 mg once daily474.83 (4.03–5.97)HR, 0.37 (0.22–0.62); p < .0013.67 (2.17–6.80)HR, 0.18 (0.10–0.34); p < .001)0(95% CI for the rate, 1.3–17.5)
Apatinib 425 mg twice daily464.27 (3.83–4.77)HR, 0.41 (0.24–0.72); p = .00173.20 (2.37–4.53)HR, 0.21 (0.11–0.38); p < .0016.38(95% CI for the rate, 4.9–26.3)
PBO482.5 (1.87–3.70)1.40 (1.20–1.83)13.04(95% CI for the rate, 0.0–7.4)
Li 2016 [78]Apatinib1466.5 (4.8–7.6)HR, 0.709 (0.537–0.937); p = .01492.6 (2.0–2.9)HR, 0.444 (0.331–0.595); p < .0012.84 (1.70)p = .1695 (p = .5532)
PBO784.7 (3.6–5.4)1.8 (1.4–1.9)0
ATTRACTION-2
Kang 2017 [75]ay
Nivolumab3305.26 (4.60–6.37)HR, 0.63 (0.51–0.78); p < .00011.61 (1.54–2.30)HR, 0.60 (0.49–0.75); p < .000111.2NR
PBO1634.14 (3.42–4.86)1.45 (1.45–1.54)0
JAVELIN Gastric 300
Bang 2018 [82]ay
Avelumab + BSC1854.6 (3.6–5.7)HR, 1.1 (0.9–1.4); p = .811.4 (1.4–1.5)HR, 1.73 (1.4–2.2); p > .992.2NR
Chemo + BSC1855.0 (4.5–6.3)2.7 (1.8–2.8)4.3
TAGS
Shitara 2018b [76]
Trifluridine/tipiracil + BSC3375.7 (4.8–6.2)HR, 0.69 (0.56–0.85); p = .000292.0 (1.9–2.3)HR, 0.57 (0.47–0.70); p < .00014p = .28
PBO + BSC1703.6 (3.1–4.1)1.8 (1.7–1.9)2
CheckMate-032
Janjigian 2018 [80]ay
Nivolumab 3 mg/kg596.2 (3.4–12.4)NR1.4 (1.2–1.5)NR7NR
Nivolumab 1 mg/kg + ipilimumab 3 mg/kg496.9 (3.7–11.5)1.4 (1.2–3.8)20NR
Nivolumab 3 mg/kg + ipilimumab 1 mg/kg524.8 (3.0–8.4)1.6 (1.4–2.6)4NR
ATTRACTION-2 subgroup analysis (Japanese patients)
Kato 2019 [79]ay
Nivolumab1525.4 (4.6–7.4)HR, 0.58 (0.42–0.78); p = .00021.7 (1.6–2.8)HR, 0.53 (0.39–0.72); p < .000114.0p = .0023
Placebo743.6 (2.8–5.0)1.5 (1.5–1.6)0
ATTRACTION-2 subgroup analysis (prior Tras use)
Satoh 2020 [119]ay
History of Tras: nivolumab598.3 (5.3–12.9)HR, 0.38 (0.22–0.66); p = .00061.6 (1.5–4.0)HR, 0.49 (0.29–0.85); p = .011116.9NR
History of Tras: PBO223.1 (1.9–5.3)1.5 (1.3–2.9)0
No history of Tras: nivolumab2714.8 (4.1–6.0)HR, 0.71 (0.57–0.88); p = .00221.6 (1.5–2.4)HR, 0.64 (0.51–0.80);p = .00017.7NR
No history of Tras: PBO1414.2 (3.6–4.9)1.5 (1.5–1.5)0

aIn Ajani et al. (2010) [26], Ryu et al. (2015) [109], Waddell et al. (2013) [18], Wang et al. (2016) [19], Bando et al. (2016) [21], Bang et al. (2010) [17], Guimbaud et al. (2014) [22], Hecht et al. (2016) [23], Li et al. (2015) [113], and Lordick et al. (2013) [114], data were not reported as being either adjusted or unadjusted.

bIn Shen et al. (2015) [110], Van Cutsem et al. (2015) [37], Yamada et al. (2015) [20], and Kang et al. (2009) [24], unadjusted data are presented for OS and PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

cIn Kim et al. (2014) [112] and Ohtsu et al. (2011) [25], unadjusted data are presented for OS, PFS, and response; for other endpoints, data were not reported as being either adjusted or unadjusted.

dIn Shen et al. (2015) [110], Waddell et al. (2013) [18], Wang et al. (2016) [19], Yamada et al. (2015) [20], Bando et al. (2016) [21], Guimbaud et al. (2014) [22], Hecht et al. (2016) [23], Kang et al. (2009) [24], Kim et al. (2014) [112], Lordick et al. (2013) [114], and Ohtsu et al. (2011) [25], RECIST version 1.0 criteria were used.

eIn Kang et al. (2009) [24] and Li et al. (2015) [113], only patients with measurable disease were included.

fIn Ajani et al. (2010) [26], RECIST criteria were used, but no version number was provided.

gIn Ajani et al. (2010) [26], patients were stratified based on whether disease was measurable (95.6% of patients had measurable disease).

hIn Bang et al. (2010) [17] and Van Cutsem et al. (2015) [37], there was no indication that RECIST criteria were used.

iIn Bang et al. (2010) [17], patients were stratified based on whether disease was measurable (~90% of patients had measurable disease).

jCap plus Cis or FU plus Cis, chosen at the investigator's discretion.

k“Overall tumor response rate” (complete response plus partial response).

lIn Ohtsu et al. (2011) [25], the measurable disease population was used to evaluate response rate (~79% of patients had measurable disease).

mPatients unable to take oral medications received FU. Switching from Cap to FU during the study was not permitted.

nIn Hecht et al. (2016) [23] and Lordick et al. (2013) [114], no information was provided regarding the handling of data from patients with measurable versus nonmeasurable disease.

oIn Guimbaud et al. (2014) [22], patients were stratified based on whether disease was measurable (the proportion of patients with measurable disease was not provided).

pIn Kim et al. (2014) [112], patients were stratified based on whether disease was measurable (~64% of patients had measurable disease).

qIn Ryu et al. (2015) [109], response rate was calculated only for patients with measurable disease (~61% of patients).

rIn Yamada et al. (2015) [20], those with no measurable disease were excluded from the per-protocol population (~2.5% of patients).

sIn Wang et al. (2016) [19], those with no measurable disease were excluded from the efficacy population (~1.3% of patients).

tIn Ryu et al. (2015) [109] and Fuchs et al. (2019) [40], RECIST version 1.1 criteria were used.

uIn Fuchs et al. (2019) [40], adjusted data were presented for PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

vFU IV infusion was permitted in patients unable to take oral capecitabine.

wIn Thuss-Patience et al. (2011) [47], Nishina et al. (2016) [50], and Bang et al. (2017b) [70], there was no indication that RECIST criteria were used.

xIn Satoh et al. (2015) [62], Satoh et al. (2014) [46], Sym et al. (2013) [55], Thuss-Patience et al. (2011) [47], Yi et al. (2012) [43], Higuchi et al. (2014) [44], Fuchs et al. (2014) [42], Kim et al. (2015) [45], Roy et al. (2013) [54], Bang et al. (2017a) [69], Kang et al. (2018) [51], Thuss-Patience et al. (2017) [66], Van Cutsem et al. (2017) [72], Lee et al. (2019) [117], and Moehler et al. (2016) [118], no information was provided regarding the handling of data from patients with measurable versus nonmeasurable disease.

yIn Thuss-Patience et al. (2011) [47], there was no indication that RECIST criteria were used.

zNo objective remission according to World Health Organization criteria.

aaIn Satoh et al. (2015) [62], Satoh et al. (2014) [46], Sym et al. (2013) [55], Tanabe et al. (2015) [56], Higuchi et al. (2014) [44], Fuchs et al. (2014) [42], Hironaka et al. (2013) [53], Nakanishi et al. (2016) [58], Roy et al. (2013) [54], Lee et al. (2017) [116], and Ohtsu et al. (2013) [73], RECIST version 1.0 criteria were used.

abIn Hironaka et al. (2013) [53], response rate was assessed in patients with measurable disease at baseline (~80% of patients).

acPEP02 is a highly stable liposomal nanocarrier formulation of irinotecan.

adITT population. Similar results were obtained for the modified ITT population (i.e., randomly assigned patients confirmed FISH positive by central laboratory).

aeIn Bang et al. (2015) [48], Wilke et al. (2014) [41], Muro et al. (2016) [63], Shitara et al. (2016) [64], Bang et al. (2017a) [69], Kang et al. (2018) [51], Kim et al. (2015) [45], Shitara et al. (2018b) [76], Thuss-Patience et al. (2017) [66], Van Cutsem et al. (2017) [72], Lee et al. (2019) [117], Fushida et al. (2016) [60], Pavlakis et al. (2016) [74], and Shitara et al. (2014) [59], RECIST version 1.1 criteria were used.

afIn Wilke et al. (2014) [41], patients were stratified based on whether disease was measurable (~81% of patients had measurable disease).

agIn Tanabe et al. (2015) [56], response rate was calculated only for patients with measurable disease (~82% of patients had measurable disease).

ahEighteen-month OS.

aiIn Nishikawa et al. (2015) [57] and Kang et al. (2012) [61], RECIST criteria were used, but no version number was provided.

ajIn Muro et al. (2016) [63], patients were stratified based on whether disease was measurable (~72% of East Asian patients and ~ 81% of non–East Asian patients had measurable disease).

akIn Shitara et al. (2016) [64], patients were stratified based on whether disease was measurable (72.1% of Japanese patients and 83.4% of Western patients had measurable disease).

alIn Nakanishi et al. (2016) [58], measurable disease was an adjustment factor during randomization (~42% of patients had measurable disease).

amIn Lee et al. (2019) [117], response evaluation was conducted on patients with at least one measurable lesion (38/54 in the paclitaxel group and 44/58 in the irinotecan group).

anIn the GOLD study (Bang et al. [2017a] [69]), statistical significance was set at p < .025.

aoIn the KEYNOTE-061 study (Shitara et al. [2018a] [71]), statistical significance was set at p < .0215.

apIn Ohtsu et al. (2013) [73], objective response of complete response or partial response, only assessed in the patients with measurable disease at baseline (approximately 86% in the everolimus plus BSC group and 88% in the placebo plus BSC group).

aqIn Shitara et al. (2014) [59], objective response of complete response or partial response, only assessed in the patients with measurable disease at baseline (78% in the weekly paclitaxel group and 75% in the dose-escalated weekly paclitaxel group).

arIn Pavlakis et al. (2016) [74], only patients with measurable disease were included in the study.

asIn Satoh et al. (2015) [62], Satoh et al. (2014) [46], Sym et al. (2013) [55], Tanabe et al. (2015) [56], Thuss-Patience et al. (2011) [47], Yi et al. (2012) [43], Higuchi et al. (2014) [44], Muro et al. (2016) [63], Hironaka et al. (2013) [53], Kim et al. (2015) [45], Nakanishi et al. (2016) [58], Nishina et al. (2016) [50], Roy et al. (2013) [54], Nishikawa et al. (2015) [50], Fushida et al. (2016) [60], Kang et al. (2012) [61], Moehler et al. (2016) [28], Ohtsu et al. (2013) [73], Pavlakis et al. (2016) [74], and Shitara et al. (2014) [59], data were not reported as being either adjusted or unadjusted.

atIn Ford et al. (2014) [49], adjusted data are presented for OS; for other endpoints, data were not reported as being either adjusted or unadjusted.

auIn Bang et al. (2015) [48], adjusted data are presented for OS and PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

avIn Fuchs et al. (2014) [42] and Wilke et al. (2014) [41], unadjusted data are presented for OS and PFS; for other endpoints, data were not reported as being either adjusted or unadjusted.

awIn Shitara et al. (2016) [64], adjusted data are presented for ORR and disease control rate; for other endpoints, data were not reported as being either adjusted or unadjusted.

axIn Bang et al. (2015) [48], response rate was calculated only for patients with measurable disease (~81% of patients).

ayIn Bang et al. (2018) [82], Kang et al. (2017) [75], Kato et al. (2019) [79], Shitara et al. (2018a) [71], and Janjigian et al. (2018) [80], data were not reported as being either adjusted or unadjusted. In Satoh et al. (2020) [119], adjusted data are presented for ORR and disease control rate; for other endpoints, data were not reported as being either adjusted or unadjusted.

Abbreviations: ATM, ataxia telangiectasia mutated; BEV, bevacizumab; BSC, best supportive care; Cap, capecitabine; Chemo, chemotherapy; CI, confidence interval; Cis, cisplatin; CPS, combined positive score; CTX, cetuximab; D, day; Doc, docetaxel; Epir, epirubicin; FOL, folinic acid FOLFIRI, irinotecan plus 5-fluorouracil plus folinic acid (also known as leucovorin); FU, fluorouracil; HR, hazard ratio; IQR, interquartile range; IRI, irinotecan; irPFS, immune-related progression-free survival; ITT, intent-to-treat; IV, intravenous; LAP, lapatinib; MET, mesenchymal-epithelial transition; mWHO, modified World Health Organization; nab-Pac, nab-paclitaxel; NR, not reported; NS, not significant; OR, odds ratio; ORR, overall response rate; Ox, oxaliplatin; Pac, paclitaxel; PAN, panitumumab; PBO, placebo; PD-L1, programmed death-ligand 1; PFS, progression-free survival; QW, once weekly; Q3W, once every 3 weeks; RAM, ramucirumab; SIM, simvastatin; SoC, standard of care; Tras, trastuzumab.

Chemotherapeutic Agents

The majority of studies assessed combination chemotherapy in both arms. Only one study included a monotherapy arm.

In this SLR, studies that excluded patients with HER2-overexpressing (HER2+) tumors generally evaluated regimens without biomarker targets, focusing on new combinations to optimize the benefit-to-risk ratio. Eleven RCTs compared the efficacy and/or safety of different doublet regimens. Cisplatin plus capecitabine versus cisplatin plus 5-FU showed noninferior OS and PFS and higher ORR while not significantly affecting toxicity [24].

Two studies compared the effect of S-1 plus cisplatin versus 5-FU plus cisplatin [26,27]. The FLAGS study found that median TTF was longer and the AE profile was more favorable with S-1 plus cisplatin than with 5-FU plus cisplatin [26]. The DIGEST study found no significant difference in OS between S-1 plus cisplatin and 5-FU plus cisplatin [27]. Although outside the inclusion parameters used in this review, the SC-101 and START studies established the benefits of frontline S-1–based combination therapies in Asian populations [30,31]. SC-101 demonstrated superior benefits for S-1 plus cisplatin compared with S-1 monotherapy or 5-FU plus cisplatin in Chinese patients, and the START study demonstrated significant clinical benefits (OS, 12.5 vs. 10.8 months; p = .032; PFS, 5.3 vs. 4.2 months; p = .001) in Korean and Japanese patients treated with docetaxel plus S-1 compared with S-1 monotherapy.

Shu et al. found that oxaliplatin plus S-1 was noninferior to oxaliplatin plus tegafur in terms of PFS and OS [32]. The G-SOX study evaluated S-1 plus oxaliplatin or S-1 plus cisplatin and showed noninferiority that was statistically significant [21]. These results may have been mediated by the observed better tolerability with oxaliplatin versus cisplatin in the elderly. In G-SOX, discontinuation rates due to AEs and serious AEs were higher in the S-1 plus cisplatin group than in the S-1 plus oxaliplatin group. Although outside the inclusion parameters of this review, Al-Batran et al. compared fluorouracil, leucovorin, and oxaliplatin (FLO) with fluorouracil, leucovorin, and cisplatin (FLP) in patients with advanced gastric cancer [33]. No significant OS or PFS benefits were observed between FLO and FLP arms, although in older adults FLO was associated with increased efficacy. Importantly, FLO was associated with significantly lower frequency of AEs (e.g., any grade vomiting 31% [FLO] vs. 52% [FLP]) and treatment-related serious AEs (9% [FLO] vs. 19% [FLP]). Along these lines, although the randomized phase II CALGB 80403 study of cetuximab with one of three chemotherapy regimens (epirubicin, cisplatin, and continuous-infusion fluorouracil; irinotecan plus cisplatin; or folinic acid plus 5-FU plus oxaliplatin [FOLFOX]) did not meet the inclusion criteria of 200 or more patients in 1L, its results indicated that FOLFOX was better tolerated and was the recommended backbone for 1L [34]. The FOLFOX arm reported fewer treatment modifications and discontinuations due to treatment-related AEs or deaths [34]. These data suggest better tolerability for oxaliplatin-based regimens versus cisplatin, with comparable efficacy.

One study compared the effect of cisplatin and docetaxel when paired with S-1 [35]. OS was numerically longer with S-1 plus docetaxel than with S-1 plus cisplatin (405 days vs. 378 days; p = .5127), although the difference was not significant. One study compared the effect of paclitaxel plus capecitabine with cisplatin plus capecitabine [29]. Lu et al. found no significant difference in OS between the two regimens [29]. These data suggest that a taxane-based doublet may be a suitable alternative to a platinum-based doublet.

Despite statistically significant longer OS (10.2 vs. 8.5 months; hazard ratio [HR], 0.71; p = .0319) and PFS (7.2 vs. 4.9 months; HR, 0.58; p = .0008) and improved ORR in patients treated with a modified combination of docetaxel plus cisplatin/5-FU (mDCF) relative to cisplatin/5-FU, toxicity was greater [19]. Incidence of grade 3/4 AEs (e.g., neutropenia) was higher in the mDCF arm [19]. A Japanese study showed no OS benefit (14.2 vs. 15.3 months; HR, 0.99) but higher grade 3 or worse AEs (neutropenia, leukopenia, and anorexia) when docetaxel was added to cisplatin plus S-1 [36].

A single three-arm RCT compared doublet with two triplet chemotherapy regimens: docetaxel plus oxaliplatin versus this doublet combined with 5-FU (TEF) or capecitabine [37]. With better safety, median PFS (mPFS) of 7.7 months, median OS of 14.6 months, and ORR of 46.6% in TEF-treated patients, TEF was deemed to have a significantly better therapeutic index. These studies demonstrate that although efficacy is better with triplet regimens, toxicities are increased compared with doublets.

Guimbaud et al. were the first to prospectively address therapy sequencing (1L and 2L) ECX (epirubicin, cisplatin, and capecitabine) followed by FOLFIRI (folinic acid plus 5-FU plus irinotecan) versus FOLFIRI followed by ECX [22]. Although PFS, OS, and ORR were similar, FOLFIRI administered prior to ECX as 2L led to a statistically significant increase in the primary endpoint of TTF relative to ECX given first (median 5.1 vs. 4.2 months, respectively). First-line FOLFIRI was also better tolerated with lower rates of grade 3/4 toxicities and hematologic AEs but similar rates of nonhematologic AEs [22].

Targeted Therapies

Findings from the current SLR in patients with HER2+ tumors support those of a previous Cochrane review (2010), which recommended trastuzumab plus cisplatin plus 5-FU or capecitabine [9]. In ToGA, addition of trastuzumab to chemotherapy improved OS (13.8 vs. 11.1 months), PFS (6.7 vs. 5.5 months), ORR (47% vs. 35%), TTP, and duration of response [17]. Similarly, in the TRIO-013/LOGiC study, mPFS was longer and ORR was higher with the addition of lapatinib to a combination of capecitabine plus oxaliplatin; however, lapatinib increased toxicity and OS was not significantly improved [23]. In the JACOB trial, addition of pertuzumab to trastuzumab plus chemotherapy did not significantly improve OS [38].

Two studies (RILOMET-1 and METGastric) assessed the impact of adding targeted therapy (rilotumumab or onartuzumab) to chemotherapy in patients with advanced mesenchymal-epithelial transition (MET)–positive G/GEA, a population with a poor prognosis [28,39]. However, neither improved clinical outcomes when combined with chemotherapy.

RAINFALL assessed the impact of adding ramucirumab to chemotherapy (cisplatin plus capecitabine or 5-FU) in patients with HER2-negative tumors. Investigator-assessed PFS was significantly longer for ramucirumab plus chemotherapy versus placebo plus chemotherapy; however, the benefit was not confirmed by an independent, central review, and there was no difference in OS between groups [40]. In the AVAGAST study, the addition of bevacizumab to chemotherapy did not improve OS [25].

The remaining 1L targeted therapy studies included in this review reported either no significant differences in PFS or OS or worsened clinical efficacy in the investigational versus comparator arm [18]. Despite several attempts, targeted therapies in 1L have not yielded significant benefits except for patients with HER2+ tumors.

Efficacy and Safety of 2L Interventions

Of the included studies, singlets and doublets with or without a targeted agent were the most commonly assessed interventions. Fifteen of the 34 included RCTs reported statistically significant findings for OS, PFS, TTF, ORR, and/or disease control rate (DCR) [26,4150].

Chemotherapeutic Agents

Consistent with prior reviews [8,9], single-agent chemotherapy prolonged OS when compared with BSC or active symptom control measures in the post-1L setting [47,49]. RCTs that compared monotherapies included the JCOG0407 trial, where paclitaxel improved mPFS by 1.3 months compared with 5-FU [50]. This PFS benefit appeared to outweigh the toxicity profile. The DREAM study assessed the efficacy of DHP107, an oral paclitaxel, in patients with advanced gastric cancer after failure of first-line therapy [51]. DREAM demonstrated PFS noninferiority and a similar safety profile for DHP107. The ABSOLUTE study showed noninferior OS with weekly nab-paclitaxel compared with standard paclitaxel [52].

WJOG 4007 evaluated paclitaxel versus irinotecan and found similar OS and manageable toxicities for both [53]. Roy et al. showed the ORR of irinotecan was lower than that of either docetaxel or PEP02, a liposomal irinotecan (6.8% vs. 15.9% vs. 13.6%, respectively), although mPFS was similar [54].

Additional RCTs suggested that irinotecan combination regimens (e.g., FOLFIRI or irinotecan plus cisplatin) may be suitable post-1L chemotherapy. Sym et al. indicated the addition of 5-FU/leucovorin is as effective and tolerable as irinotecan monotherapy [55]. Thuss-Patience et al. found that OS (4.0 vs. 2.4 months, respectively) was longer when irinotecan was added to BSC [47]. In the TCOG GI-0801 study, irinotecan plus cisplatin improved PFS and DCR, but not OS or ORR, when compared with cisplatin alone [44]. JACCRO GC-05 [56] and TRICS [57] concluded that the addition of a second cytotoxic agent did not improve irinotecan efficacy. Taken together, these studies suggest the benefit-to-risk ratio for paclitaxel and irinotecan monotherapies in 2L is equivalent, whereas combination irinotecan-based chemotherapy, namely, modified FOLFIRI or irinotecan plus cisplatin, may be suitable although clinical benefit is debatable.

Taxane-containing doublets (docetaxel plus oxaliplatin) compared with taxane monotherapy (docetaxel) improved mPFS from 2 to 4.9 months in docetaxel alone, although OS and ORR were not different [45]. In contrast, the doublet of paclitaxel plus S-1 did not improve efficacy over paclitaxel alone [58]. Moreover, there were nearly twice as many discontinuations due to AEs in the combination, although grade 3/4 AE rates were similar between treatment arms. Lee et al. reported the addition of S-1, but not cisplatin, to docetaxel resulted in better PFS compared with docetaxel alone. These data indicate that careful consideration of efficacy and toxicities is necessary, especially of AEs observed in 1L, when planning taxane/platinum-based doublet therapies in 2L.

Several studies included in the SLR combined 2L and 3L. Shitara et al. reported that dose-escalated paclitaxel resulted in longer PFS compared with standard-dose paclitaxel [59]. Frequency of all grades of neutropenia was significantly higher with dose-escalated paclitaxel; however, no significant difference was observed in the proportion of patients experiencing grade 3 or higher AEs. Fushida et al. reported that the addition of paclitaxel to valproic acid did not significantly improve OS or PFS [60]. Kang et al. observed longer OS (5.3 vs. 3.8 months) and similar tolerability when docetaxel or irinotecan were added to BSC [61].

Targeted and Immunotherapies

Targeted therapies, either alone or in combination, were investigated in 13 2L studies [4143,46,48,6264]. Two trials examined ramucirumab as monotherapy (vs. BSC in REGARD) or combined with paclitaxel (RAINBOW) [41,42]. OS and PFS were significantly improved in the ramucirumab-containing arms in both studies. In REGARD, OS was 5.2 versus 3.8 months and PFS was 2.1 versus 1.3 months, respectively. In RAINBOW, OS was 9.6 versus 7.4 months and PFS was 4.4 versus 2.9 months, respectively. Although not powered to show significance, post hoc analyses supported clinical benefits for ramucirumab plus paclitaxel efficacy in both East Asian and non–East Asian patients [63,64]. Unlike PFS, significant OS benefits were not noted in either of these two subgroup analyses in Asian populations, and the authors suggested that post-discontinuation therapy may play a role in the observed modest OS differences [63,64]. Recently, the phase III RAINBOW-Asia study demonstrated significant PFS benefit for ramucirumab combined with paclitaxel compared with paclitaxel alone; however, no OS benefits were observed [65]. Taken together, these studies indicate that in Asian populations the OS benefit from a ramucirumab plus paclitaxel regimen may be limited. The pan-tyrosine kinase inhibitor, sunitinib, combined with docetaxel was compared with docetaxel alone for the primary endpoint of TTP in a phase II trial. Although TTP was not statistically different, higher ORR was observed and safety was reduced in the doublet combination arm [43].

In TyTAN, addition of lapatinib to paclitaxel failed to demonstrate significant survival benefits (PFS, OS) versus paclitaxel alone in patients with HER2+ tumors [46]. Of note, when compared with similar subgroups of patients treated with paclitaxel, patients treated with the doublet combination who had higher HER2 expression or who were mainland Chinese patients had improved OS (11.0 vs. 8.9) and PFS (5.5 vs. 4.4) [46]. Safety was not affected by the addition of lapatinib to paclitaxel. In the GATSBY study, trastuzumab emtansine was not superior to a taxane in improving OS in patients with HER2+ tumors [66]. The COG phase III study analyzed gefitinib (epidermal growth factor receptor [EGFR] inhibitor) versus placebo in esophageal cancer demonstrating no statistical OS or PFS benefit, although palliative benefits in subgroups were observed [67,68]. More recently, the JAPICTI RCT compared irinotecan alone with adding irinotecan to nimotuzumab, an anti-EGFR targeting antibody [62]. The primary endpoint, PFS, was similar between treatment arms, although patients with high EGFR levels by immunohistochemistry had improved OS, PFS, and ORR without adversely affecting safety [62]. Despite these results, the phase III study of nimotuzumab with irinotecan was terminated (NCT01813253).

Other studies of targeted 2L therapies included olaparib, ipilimumab, pembrolizumab, and trastuzumab emtansine. Bang et al. (Study 39, 2015) showed that the addition of PARP inhibitor olaparib to paclitaxel improved OS in patients with low ataxia telangiectasia mutated levels in the intent-to-treat population, although these results are discordant with the GOLD trial in which OS benefit was not observed [48,69]. Bang et al. (2017b) reported that ipilimumab monotherapy did not improve PFS or OS compared with BSC [70]. In KEYNOTE-061, pembrolizumab did not significantly improve OS compared with paclitaxel in patients with programmed death-ligand 1 (PD-L1) combined positive score (CPS) of 1 or higher [71]. In the study by Van Cutsem et al., AZD4547 (a selective fibroblast growth factor receptor [FGFR] 1–3 tyrosine kinase inhibitor) did not significantly improve PFS compared with paclitaxel in patients with FGFR2 polysomy or gene amplification [72]. These negative results indicate that further studies are necessary to support the possibility for improving outcomes in biomarker enriched subgroups.

The GRANITE study failed to show statistically significant benefit for OS (primary), unlike for PFS, comparing everolimus plus BSC with placebo plus BSC [73]. The phase II INTEGRATE study, evaluating both 2L and 3L therapy, found that addition of regorafenib to BSC significantly improved PFS; the phase III study is ongoing [74].

Overall, these studies indicate that, in a 2L setting, single-agent chemotherapy (or combination with targeted therapy) is more efficacious than BSC, highlighting the need for careful consideration of control arms in future study designs.

Efficacy and Safety of 3L+ Interventions

Eight articles were identified that assessed 3L+ treatments: six primary RCTs and two secondary reports. Four of the six RCTs reported significant findings for OS, PFS, DCR, and/or TTP [7578]. One secondary study reported significant findings for ORR [79].

ATTRACTION-2 showed statistically significantly longer OS (5.3 vs. 4.1 months) and PFS (1.61 vs. 1.45 months) and higher DCR with nivolumab (anti–PD-1 monoclonal antibody) than placebo in Asian patients with disease progression after at least two prior chemotherapies [75]. The safety profile was manageable, and survival benefit with nivolumab was sustained beyond 1 year, independent of PD-L1 expression (although this was evaluated with tumor positivity score (TPS), not combined positivity score (CPS)). Subgroup analyses of Japanese patients and patients with prior trastuzumab use from the ATTRACTION-2 study also demonstrated similar clinical and safety results.

Similarly, the phase I/II CheckMate-032 study demonstrated that nivolumab as monotherapy and combined with ipilimumab (dual PD-1/cytotoxic T-lymphocyte–associated antigen 4 blockade) produced some durable responses, long-term OS, and a manageable safety profile in Western patients who experienced disease progression following at least one prior chemotherapy regimen [80]. Nivolumab was approved for 3L treatment of metastatic gastric cancer in Japan, Taiwan, and Korea, supported by results from the ATTRACTION-2 study [80,81].

The JAVELIN Gastric 300 study found that avelumab did not statistically significantly improve OS, PFS, or ORR compared with chemotherapy, with a trend to worse OS [82]. The studies conducted in China by Li et al. found that apatinib significantly improved OS (6.5 vs. 4.7 months) and PFS (2.6 vs. 1.8 months) compared with placebo with an acceptable safety profile [77,78]; however, the global phase III ANGEL study, which included patients from Europe and North America in addition to Asia, failed to show significant OS benefit in the overall population (3L+) [83].

The TAGS study reported statistically significantly longer OS (5.7 vs. 3.6 months), PFS (2.0 vs. 1.8 months), and DFS with trifluridine/tipiracil (TAS-102) compared with placebo [76].

Discussion

Unlike previous SLRs, this SLR aimed to inform optimal treatment sequencing in advanced metastatic G/GEA. This study parallels earlier work by Wagner et al. that identified study types, disease, treatment, and population [9]. All 1L RCTs in the current study had a fluoropyrimidine/platinum combination in at least one treatment arm, and 1L, 2L, and 3L+ treatments were considered separately to address the treatment sequencing question. In previous reports, HER2 status was not considered, and comparisons of singlet or doublet regimens versus supportive care, and doublets compared with monotherapy, were a primary focus [9]. Despite our focus on larger RCTs in this population with advanced G/GEA, descriptive cross-trial comparisons that cannot account for confounding variables between differing study populations are limitations of this assessment. Treatment decisions are heavily reliant on clinician discernment of available evidence, and this report attempts to highlight important differences in the studies included within.

Despite considerable improvements in therapeutic options, the treatment of advanced G/GEA remains heterogeneous [3].

For those likely to tolerate chemotherapy, doublet regimens (i.e., platinum/fluoropyrimidine) are preferable over triplet chemotherapy. Doublets often exhibited lower toxicity rates, which may outweigh any incremental clinical benefits seen with triplet therapy. For example, the toxicity observed with addition of a third chemotherapy (docetaxel or epirubicin) to a platinum/fluoropyrimidine appears to outweigh a survival benefit, as was observed in the V325 study [13]. However, an mDCF regimen shows promise of extending survival with acceptable toxicity in two trials [19,84]. Controversy remains with taxane triplets. The phase III JCOG1013 trial (n = 741) was recently published comparing cisplatin plus S-1 (CS) versus CS plus docetaxel (DCS) in an exclusively Japanese patient population [36]; no significant difference was seen in OS between CS and DCS (median 15.3 vs. 14.2 months). In line with other trials examining taxane triplets, higher grade 3/4 neutropenia was seen with DCS (58.5%) versus CS (32.1%). Another emerging regimen is FOLFIRINOX (irinotecan plus platinum plus fluoropyrimidine), while not a randomized study, demonstrated similar clinical outcomes to platinum/fluropyrimidine/taxane but with better tolerabilty due to non-overlapping toxicity [85]. For 1L treatment of HER2+ advanced G/GEA, trastuzumab should be added to platinum/fluoropyrimidine, although recently, oxaliplatin-based regimens (capecitabine plus oxaliplatin [XELOX] or FOLFOX) have also been widely adopted instead of cisplatin/fluoropyrimidine for HER2+ tumors [86,87].

For patients with advanced HER2-negative G/GEA and a good performance status but who are not amenable to surgical resection, 1L recommended treatment options include FOLFOX or a combination of capecitabine plus oxaliplatin. The 2L RAINBOW study did not enroll patients with a prior docetaxel containing triplet therapy, and an exploratory analysis indicated increased toxicities with prior triplet compared with doublet therapies [88]. Given the improvements in OS in patients with favorable performance status using various 2L regimens, sequentially navigating patients to active 2L therapy as opposed to upfront triplets containing taxanes may provide survival benefits with less toxicity. Triplet 1L chemotherapy, however, may be a consideration for patients with heavy disease burden severe cancer-related symptoms at diagnosis but with minimal comorbidities.

With a greater emphasis of biologic, targeted agents in 1L trials, the lower toxicity of doublet versus triplet chemotherapy favors a backbone regimen such as FOLFOX. Indeed, the majority of recently published 1L clinical trial data with other targeted agents with or without a chemotherapy backbone has reported negative results. Theoretically, with taxane use increasing in 2L therapy, restricting taxanes in 1L could prevent drug resistance.

For G/GEA that progressed on a fluoropyrimidine/platinum 1L therapy (plus trastuzumab for HER2+ tumors), taxane-based therapy, or consideration of ramucirumab monotherapy if the patient is not a good candidate for cytotoxic chemotherapy, is indicated. Efficacy, safety, and treatment compliance are high-priority considerations when choosing a 2L therapy. Data also support use of irinotecan, either as monotherapy or in FOLFIRI. The addition of ramucirumab to an irinotecan backbone is a possibility, particularly in patients with neuropathy including oxaliplatin-induced neuropathy from 1L therapy. Evidence supports ramucirumab plus FOLFIRI or ramucirumab plus irinotecan as an alternative in 2L patients ineligible for ramucirumab/paclitaxel [8991]. In a retrospective analysis by Klempner et al., patients receiving ramucirumab plus FOLFIRI (after 1L platinum plus fluoropyrimidine) had ORR of 23%, DCR of 79%, mPFS of 6.0, and median OS (mOS) of 13.4 months [89]. Lorenzen et al. reported that patients with prior taxane use receiving ramucirumab plus FOLFIRI had ORR of 24%, DCR of 67%, mPFS of 4.3, and mOS of 7.5 months [90], whereas Park et al. reported ORR of 25% for patients who advanced on 1L and were then treated with ramucirumab plus irinotecan [91]. The authors posit that a shorter time to initiation of 2L treatment following disease progression or development of unacceptable toxicity, but before patients experience performance status decline, is a key consideration. This in turn will benefit patients who are eligible to further receive 3L treatment options like TAS-102 that demonstrated statistically significant survival benefits (OS, PFS, DCR) in the TAGS study [76].

The Argument for Limiting Time on 1L/Maintenance 1L Therapy in Advanced G/GEA

Based on the success of maintenance therapy in colorectal cancer (OPTIMOX1 [92] and CAIRO3 [93]), many oncologists have adapted this approach to advanced G/GEA. Following a predetermined length of 1L therapy (typically 4–6 months), maintenance therapy may provide similar (or better) efficacy with less toxicity (particularly cumulative oxaliplatin-related neuropathy) compared with continuing 1L therapy until disease progression. Maintenance options include switch therapy or low-dose continuation of a 1L agent (i.e., 5-FU or capecitabine). In support of maintenance therapy, the 1L trial, ToGA, stopped chemotherapy after six cycles but continued trastuzumab [17]; AVAGAST and RAINFALL stopped cisplatin after six cycles but continued bevacizumab/placebo or ramucirumab/placebo with fluoropyrimidine, respectively [25,40]. OS rates were similar to other phase III studies without a maintenance approach, indicating that not all agents in 1L need to be continued indefinitely. The mPFS across major 1L trials ranged from 4.4 to 8.5 months. Park et al. compared continuous versus stop-and-go chemotherapy after disease stabilization with 1L induction chemotherapy [94]. After receiving six cycles of S-1 plus oxaliplatin (SOX), patients were randomized to receive continuous SOX until progression (continuous arm) or to have a chemotherapy-free interval followed by SOX reintroduction at progression (stop-and-go arm). Continued chemotherapy improved PFS but not duration of disease control or OS, had a negative impact on quality of life, and increased frequencies of adverse events, suggesting that the stop-and-go strategy may be an appropriate option compared with continuous 1L therapy. Indeed, for the use of oxaliplatin in 1L treatment regimens, the International Duration Evaluation of Adjuvant Therapy in therapy for colorectal cancer demonstrated more than doubling of grade 2 or higher neurotoxicity rates, 16.6% versus 47.7%, with 3 versus 6 months of FOLFOX exposure, respectively [95].

Potential for Integrating Immunotherapeutics

Beyond this review, we include a discussion of immunotherapeutics in the context of treatment sequencing in metastatic G/GEA. Immunotherapy has received significant attention in recent years, advancing therapy options in many tumor types. Recent large, phase III, randomized studies in the 2L and 3L settings of G/GEA compared monotherapy immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 signaling axis with standard monotherapy cytotoxic therapy (paclitaxel or irinotecan); however, reported results failed to meet primary endpoints for KEYNOTE-061 and JAVELIN Gastric 300, even for PD-L1–positive patients [71,82]. Pembrolizumab in the 3L setting was considered an option based on results from a single-arm phase II study (KEYNOTE-059) of PD-L1–positive patients, the incidence of which is ~50%–60% of G/GEA when using a CPS cutoff of ≥1 (CPS of both PD-L1–expressing tumor and immune cells) [96], however the conditional approval has since been withdrawn. Nivolumab is also a 3L+ option in Asian patients based on improved OS versus placebo in the phase III ATTRACTION-2 study [75]. In the 2L setting or later, pembrolizumab was shown to be efficacious in tumors with high microsatellite instability (MSI) or mismatch repair deficiency, the incidence of which is ~3% in metastatic G/GEA [97], as did a combined analysis of KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 [105]. Recently, pembrolizumab received tumor-agnostic U.S. Food and Drug Administration (FDA) approval for high tumor mutational burden (TMB) (≥10 mutations per megabase) based on the KEYNOTE-158 study [98]. It is important to note that KEYNOTE-158 did not include patients with G/GEA, although an exploratory analysis from the 2L KEYNOTE-061 study reported positive association with clinical outcomes in patients with TMB-high gastric cancer treated with pembrolizumab [99].

A Korean phase II trial of pembrolizumab also identified Epstein-Barr virus–positive tumors as a small molecular subset exhibiting a high proportion of durable responses [100]. Key 1L studies with ICIs have also been reported [101104]. JAVELIN Gastric 100 failed to demonstrate avelumab switch maintenance therapy as superior to continuation of 1L FOLFOX/CAPOX (capecitabine plus oxaliplatin) chemotherapy [101]. A post hoc analysis using the CPS assay, as opposed to the trial's predefined analysis of tumor cell enumeration only (TPS), to determine PD-L1 expression demonstrated OS benefit of avelumab therapy, highlighting challenges to assay heterogeneity. KEYNOTE-062 failed to demonstrate significant benefit of 1L pembrolizumab monotherapy to chemotherapy, in patients preselected for PD-L1 CPS ≥1 [102]. ATTRACTION-4 analyzed the benefit of 1L nivolumab plus chemotherapy versus chemotherapy (SOX/CAPOX) in a non–PD-L1 selected Asian population; statistical PFS benefit was observed for ICI plus chemotherapy, whereas OS failed to demonstrate such benefits [103]; PD-L1 data were not reported to date to determine differential benefit in outcome as would be expected based on all studies to date. Meanwhile CheckMate-649, investigating 1L nivolumab plus FOLFOX/XELOX against FOLFOX/XELOX, demonstrated significant benefits for all endpoints of ICI plus chemotherapy in a global population with the analysis restricted to patients with PD-L1 CPS ≥5 [104] and recently received FDA approval in all comers as a 1L regimen while NCCN guidelines have provided a tiered recommendation based on PD-L1 score with category 1 for CPS ≥5, category 2B for CPS 1-4, and no recommendation for CPS 0. Overall, these recent studies demonstrate a combination regimen (ICI plus chemotherapy) to be efficacious compared with ICI monotherapy in 1L, particularly at higher PD-L1 cutoffs. Irrespective, the ICI plus chemotherapy regimen from CheckMate-649 is expected to become 1L therapy of choice for PD-L1 CPS ≥5, whereas 2L options are expected to remain unchanged. It is also important to highlight the role of significant benefits seen in patients with MSI-high tumors treated with ICIs, including within CheckMate-649 where overall survival was most pronounced in this group, with the median overall survival of 8.8 months versus not reached in the 1L chemotherapy versus 1L chemotherapy plus nivolumab arms, respectively (HR 0.33, 95% C.I. 0.12-0.87). Pembrolizumab is FDA approved for patients with MSI-high or mismatch repair–deficient tumors in 2L and beyond, and data from recent trials continue to demonstrate benefit in this patient subgroup [105]. Although outside the parameters of this review, it is important to note additional recent FDA approvals. KEYNOTE-590 analyzed pembrolizumab in combination with cisplatinum and fluoropyrimidine-based chemotherapy in 1L and demonstrated a statistically significant improvement in OS and PFS for patients receiving ICI plus chemotherapy irrespective of PD-L1 status, but again with improvements notably in tumors with PD-L1 CPS ≥10. This FDA approval provides another ICI regimen for patients with esophageal and gastroesophageal junction Siewert type I carcinoma, and similar to the tiered recommendation of the NCCN guidlines for nivolumab, a tiered recommendation for pembrolizumab includes category 1 for CPS ≥10, category 2B for CPS 1-9, and no recommendation for CPS 0. More recently, based on the KEYNOTE-811 study, the FDA granted accelerated approval for 1L pembrolizumab plus trastuzumab in combination with fluoropyrimidine- and platinum-containing chemotherapy for patients with locally advanced unresectable/metastatic HER2+ gastric or gastroesophageal adenocarcinoma based on interim analysis response rates. Coming shortly after the CheckMate-649 and KEYNOTE-590 approvals, the KEYNOTE-811 approval expands the frontline ICI availability to HER2+ patients, and the outcomes of the phase 3 study are awaited, as are the assessments to determine whether or not their is differential benefit by PD-L1 status in HER2+ tumors as their has repeatedly been shown in HER2- patients.

Overview of Studies Published After the Review Inclusion Period and Trials in Progress

Several large RCTs were either presented in abstract form or published in peer-reviewed journals after this literature search was performed or did not meet the inclusion criteria. Some are currently considered by oncologists when selecting regimens. For example, in the U.S., there is notable off-label use of trastuzumab continuation into 2L, despite the phase II randomized T-ACT trial (WJOG7112G) demonstrating that trastuzumab continued, with or without paclitaxel, does not provide additional benefit for patients with HER2+ advanced G/GEA refractory to 1L trastuzumab plus platinum/fluoropyrimidine [106]. However, the notion of loss of HER2 amplification in resistant disease in a large proportion of patients in that and other studies leads to the possibility of continued anti-HER2 therapy in those patients not having this conversion take place [108]. Recently, the DESTINY-Gastric01 study reported significant benefit of trastuzumab deruxtecan (T-DXd) versus paclitaxel or irinotecan in 3L and was approved in Japan and by the FDA. Importantly, patients had received a 1L trastuzumab-containing regimen, thereby making T-DXd a novel option for HER2+, trastuzumab-resistant, G/GEA tumors. Significant benefits favoring T-DXd were observed (mOS, 12.5 vs. 8.4 months; HR, 0.59; p = .01; ORR, 51.3% vs. 14.3%; p < .0001) with interstitial lung disease being a notable AE from T-DXd. [107]. Additional studies with T-DXd in 2L (NCT04014075, NCT04704934) and 1L (NCT03329690) are ongoing.

With similar conclusions to those of this report, the PANGEA phase 2 study highlights the importance of optimally sequenced therapies and endorses a combined personalized treatment strategy, starting from diagnosis and across all treatment lines, to enhance benefits compared with standard treatment approaches [108].

Considering all the evidence discussed in this SLR, we propose a treatment sequencing algorithm (Fig. 4). The regimen chosen at each line of therapy should balance the patient's performance status and comorbidities with the potential for serious AEs. It is important to consider that the proposed sequence or algorithm was not tested in clinical trial settings but was based on discussions of trial evidence in this review. In 1L, considering a doublet is recommended based on manageable toxicities compared with triplets; in addition, the recent approvals of CheckMate-649, KEYNOTE-590, and KEYNOTE-811 should be considered as 1L ICI plus chemotherapy options. In 2L (and 3L), T-DXd should be considered for HER2+ patients, although challenges to rebiopsy exist, and hence liquid biopsy to determine HER2 status should be considered where feasible. Beside a FOLFIRI/irinotecan-based regimen, combinations with biologics like ramucirumab plus taxane/irinotecan options should be considered for eligible patients, especially in patients who are ineligible to receive a taxane due to neuropathies in 1L. Furthermore, patients with MSI-high and TMB-high status should be considered for ICI-based treatment (pembrolizumab). In 3L, TAS-102 is a chemotherapy option along with chemotherapy-free options with the ICIs pembrolizumab (CPS >1) and nivolumab, which should be considered. Overall, screening patients for signs of progression across all lines of therapy is recommended so that eligible patients can be administered subsequent treatment options in a timely manner.

Potential treatment sequencing algorithm. The proposed sequential algorithm is based on the current analysis of randomized controlled trials as described in this systematic literature review. Recent approvals and key trial readouts are highlighted under “Key Considerations” and discussed in this article (see Discussion section). Checkmate-649, KEYNOTE-590, and KEYNOTE-811 are noted under 1L options that should be considered in treatment planning; HER2- tumors will be assessed and dichotimzed into PD-L1 CPS ≥5 or ≥10 and eligible for anti-PD1 therapy, or HER2-/PD-L1- and receive chemotherapy alone. These sequences were not tested in a clinical trial setting. 3L options also include irinotecan or taxane, whichever not yet used previously. Pembrolizumab in 3L had its approval recently voluntarily withdrawn; nivolumab approved for 3L only in Asia. Abbreviations: 1L, first line; 2L, second line; CAPOX, capecitabine plus oxaliplatin; CPS, combined positive score; DCF, docetaxel plus cisplatin/5-fluorouracil; FOLFOX, folinic acid plus 5-FU plus oxaliplatin; FOLFIRI, folinic acid plus 5-fluorouracil plus irinotecan; HER2+, HER2 overexpressing; HER2−, HER2 negative; ICI, immune checkpoint inhibitor; MSI-h, high microsatellite instability; SOX, S-1 plus oxaliplatin; TAS-102, trifluridine/tipiracil; T-DXd, trastuzumab deruxtecan; TMB, tumor mutational burden.
Figure 4

Potential treatment sequencing algorithm. The proposed sequential algorithm is based on the current analysis of randomized controlled trials as described in this systematic literature review. Recent approvals and key trial readouts are highlighted under “Key Considerations” and discussed in this article (see Discussion section). Checkmate-649, KEYNOTE-590, and KEYNOTE-811 are noted under 1L options that should be considered in treatment planning; HER2- tumors will be assessed and dichotimzed into PD-L1 CPS ≥5 or ≥10 and eligible for anti-PD1 therapy, or HER2-/PD-L1- and receive chemotherapy alone. These sequences were not tested in a clinical trial setting. 3L options also include irinotecan or taxane, whichever not yet used previously. Pembrolizumab in 3L had its approval recently voluntarily withdrawn; nivolumab approved for 3L only in Asia. Abbreviations: 1L, first line; 2L, second line; CAPOX, capecitabine plus oxaliplatin; CPS, combined positive score; DCF, docetaxel plus cisplatin/5-fluorouracil; FOLFOX, folinic acid plus 5-FU plus oxaliplatin; FOLFIRI, folinic acid plus 5-fluorouracil plus irinotecan; HER2+, HER2 overexpressing; HER2−, HER2 negative; ICI, immune checkpoint inhibitor; MSI-h, high microsatellite instability; SOX, S-1 plus oxaliplatin; TAS-102, trifluridine/tipiracil; T-DXd, trastuzumab deruxtecan; TMB, tumor mutational burden.

Conclusion

To our knowledge, this is the first systematic review that begins to address treatment sequencing in unresectable, advanced G/GEA, including recent evidence from larger RCTs. It builds upon currently available guidelines and provides a framework for planning effective disease management, with the potential for further improvement in outcomes for patients and select patient subgroups.

See http://www.TheOncologist.com for supplemental material available online.

Acknowledgments

Eli Lilly & Co. provided funding to RTI Health Solutions for the SLR and also provided medical writing and editing support, in accordance with Good Publication Practice (GPP3) guidelines (http://www.ismpp.org/gpp3). Hannah M. Messersmith of Eli Lilly & Co. provided writing support for this manuscript. Eli Lilly & Co. contracted with Syneos Health for editorial support provided by Antonia Baldo.

Author Contributions

Conception/design: Daniel V. Catenacci, Joseph Chao, Astra M. Liepa, Holly Knoderer, Anindya Chatterjee, Eric Van Cutsem

Collection and/or assembly of data: Kei Muro, Salah Eddin Al-Batran, Zev A. Wainberg

Data analysis and interpretation: Daniel V. Catenacci, Joseph Chao, Salah Eddin Al-Batran, Samuel J. Klempner, Zev A. Wainberg, Manish A. Shah, Sun Young Rha, Atsushi Ohtsu, Astra M. Liepa, Holly Knoderer, Anindya Chatterjee, Eric Van Cutsem

Manuscript writing: Daniel V. Catenacci, Joseph Chao, Kei Muro, Salah Eddin Al-Batran, Samuel J. Klempner, Zev A. Wainberg, Manish A. Shah, Sun Young Rha, Atsushi Ohtsu, Astra M. Liepa, Holly Knoderer, Anindya Chatterjee, Eric Van Cutsem

Final approval of manuscript: Daniel V. Catenacci, Joseph Chao, Kei Muro, Salah Eddin Al-Batran, Samuel J. Klempner, Zev A. Wainberg, Manish A. Shah, Sun Young Rha, Atsushi Ohtsu, Astra M. Liepa, Holly Knoderer, Anindya Chatterjee, Eric Van Cutsem

Disclosures

Daniel V. Catenacci: Amgen, Astellas Pharma, Bristol-Myers Squibb, Five Prime Therapeutics, Foundation Medicine, Genentech/Roche, Gritstone Oncology, Guardant Health, Eli Lilly & Co., Merck, Seattle Genetics, Taiho Pharmaceutical, Tempus (H); Joseph Chao: Eli Lilly & Co., Merck, Amgen, Macrogenics, Ono Pharmaceuticals, Daiichi-Sankyo, Foundation Medicine, Bristol-Myers Squibb, AstraZeneca, Astellas (C/A), Merck (RF, H); Samuel J. Klempner: Eli Lilly & Co., Bristol-Myers Squibb, Merck, Astellas, Daiichi-Sankyo, Pieris, Natera Inc. (C/A), Turning Point Therapeutics (OI); Zev A. Wainberg: Amgen, AstraZeneca, Bayer, Bristol-Myers Squibb, Merck, Five Prime, Eli Lilly & Co., Macrogenics, Gilead, Seagen, Novartis, Array (C/A), Novartis, Plexxikon (RF); Astra M. Liepa: Eli Lilly & Co. (E, OI); Anindya Chatterjee: Eli Lilly & Co. (E, OI); Eric Van Cutsem: Array, Astellas, AstraZeneca, Bayer, Beigene, Biocartis, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Daiichi, Halozyme, GlaxoSmithKline, Incyte, Ipsen, Eli Lilly & Co., Merck Sharp & Dohme, Merck KGaA, Novartis, Pierre Fabre, Roche, Servier, Sirtex, Taiho (C/A), Amgen, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Ipsen, Eli Lilly & Co., Merck Sharp & Dohme, Merck KGaA, Novartis, Roche, Servier (RF—institution). The other authors indicated no financial relationships.

(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board

References

1

Bray
 
F
,
Ferlay
 
J
,
Soerjomataram
 
I
et al.
Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
 
2018
;
68
:
394
424
.

2

Pellino
 
A
,
Riello
 
E
,
Nappo
 
F
et al.
Targeted therapies in metastatic gastric cancer: Current knowledge and future perspectives
.
World J Gastroenterol
 
2019
;
25
:
5773
5788
.

3

Gambardella
 
V
,
Fleitas
 
T
,
Tarazona
 
N
et al.
Precision medicine to treat advanced gastroesophageal adenocarcinoma: A work in progress
.
J Clin Med
 
2020
;
9
:
3049
.

4

Sastre
 
J
,
Garcia-Saenz
 
JA
,
Diaz-Rubio
 
E
.
Chemotherapy for gastric cancer
.
World J Gastroenterol
 
2006
;
12
:
204
213
.

5

Power
 
DG
,
Kelsen
 
DP
,
Shah
 
MA
.
Advanced gastric cancer–slow but steady progress
.
Cancer Treat Rev
 
2010
;
36
:
384
392
.

6

Abrams
 
T
,
Hess
 
LM
,
Zhu
 
YE
et al.
Predictors of heterogeneity in the first-line treatment of patients with advanced/metastatic gastric cancer in the U.S
.
Gastric Cancer
 
2018
;
21
:
738
744
.

7

Barzi
 
A
,
Thara
 
E
.
Angiogenesis in esophageal and gastric cancer: A paradigm shift in treatment
.
Expert Opin Biol Ther
 
2014
;
14
:
1319
1332
.

8

Wagner
 
AD
,
Syn
 
NL
,
Moehler
 
M
et al.
Chemotherapy for advanced gastric cancer
.
Cochrane Database Syst Rev
 
2017
;(
8
):CD004064.

9

Wagner
 
AD
,
Unverzagt
 
S
,
Grothe
 
W
et al.
Chemotherapy for advanced gastric cancer
.
Cochrane Database Syst Rev
 
2010
;(
3
):CD004064.

10

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
:
475
477
.

11

Van Cutsem
 
E
,
Sagaert
 
X
,
Topal
 
B
et al.
Gastric cancer
.
Lancet
 
2016
;
388
:
2654
2664
.

12

Smyth
 
EC
,
Verheij
 
M
,
Allum
 
W
et al.
Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
 
2016
;
27
(
suppl 5
):
v38
v49
.

13

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
:
4991
4997
.

14

Harada
 
K
,
Lopez
 
A
,
Shanbhag
 
N
et al.
Recent advances in the management of gastric adenocarcinoma patients
.
F1000Res
 
2018
;
7
:
F1000
Faculty Rev-1365.

15

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
:
1286
1312
.

16

Moher
 
D
,
Liberati
 
A
,
Tetzlaff
 
J
et al.
Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement
.
PLoS Med
 
2009
;
6
:e1000097.

17

Bang
 
YJ
,
Van Cutsem
 
E
,
Feyereislova
 
A
et al.
Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial
.
Lancet
 
2010
;
376
:
687
697
.

18

Waddell
 
T
,
Chau
 
I
,
Cunningham
 
D
et al.
Epirubicin, oxaliplatin, and capecitabine with or without panitumumab for patients with previously untreated advanced oesophagogastric cancer (REAL3): A randomised, open-label phase 3 trial
.
Lancet Oncol
 
2013
;
14
:
481
489
.

19

Wang
 
J
,
Xu
 
R
,
Li
 
J
et al.
Randomized multicenter phase III study of a modified docetaxel and cisplatin plus fluorouracil regimen compared with cisplatin and fluorouracil as first-line therapy for advanced or locally recurrent gastric cancer
.
Gastric Cancer
 
2016
;
19
:
234
244
.

20

Yamada
 
Y
,
Higuchi
 
K
,
Nishikawa
 
K
et al.
Phase III study comparing oxaliplatin plus S-1 with cisplatin plus S-1 in chemotherapy-naive patients with advanced gastric cancer
.
Ann Oncol
 
2015
;
26
:
141
148
.

21

Bando
 
H
,
Yamada
 
Y
,
Tanabe
 
S
et al.
Efficacy and safety of S-1 and oxaliplatin combination therapy in elderly patients with advanced gastric cancer
.
Gastric Cancer
 
2016
;
19
:
919
926
.

22

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
:
3520
3526
.

23

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
:
443
451
.

24

Kang
 
YK
,
Kang
 
WK
,
Shin
 
DB
et al.
Capecitabine/cisplatin versus 5-fluorouracil/cisplatin as first-line therapy in patients with advanced gastric cancer: A randomised phase III noninferiority trial
.
Ann Oncol
 
2009
;
20
:
666
673
.

25

Ohtsu
 
A
,
Shah
 
MA
,
Van Cutsem
 
E
et al.
Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: A randomized, double-blind, placebo-controlled phase III study
.
J Clin Oncol
 
2011
;
29
:
3968
3976
.

26

Ajani
 
JA
,
Rodriguez
 
W
,
Bodoky
 
G
et al.
Multicenter phase III comparison of cisplatin/S-1 with cisplatin/infusional fluorouracil in advanced gastric or gastroesophageal adenocarcinoma study: The FLAGS trial
.
J Oncol
 
2010
;
28
:
1547
1553
.

27

Ajani
 
JA
,
Abramov
 
M
,
Bondarenko
 
I
et al.
A phase III trial comparing oral S-1/cisplatin and intravenous 5-fluorouracil/cisplatin in patients with untreated diffuse gastric cancer
.
Ann Oncol
 
2017
;
28
:
2142
2148
.

28

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
:
1467
1482
.

29

Lu
 
Z
,
Zhang
 
X
,
Liu
 
W
et al.
A multicenter, randomized trial comparing efficacy and safety of paclitaxel/capecitabine and cisplatin/capecitabine in advanced gastric cancer
.
Gastric Cancer
 
2018
;
21
:
782
791
.

30

Jin
 
M
,
Lu
 
H
,
Li
 
J
et al.
Randomized 3-armed phase III study of S-1 monotherapy versus S-1/CDDP (SP) versus 5-FU/CDDP (FP) in patients (pts) with advanced gastric cancer (AGC): SC-101 study
.
J J Clin Oncol.
 
2008
;
26
(
suppl 15
):
4533a
.

31

Koizumi
 
W
,
Kim
 
YH
,
Fujii
 
M
et al.
Addition of docetaxel to S-1 without platinum prolongs survival of patients with advanced gastric cancer: A randomized study (START)
.
J Cancer Res Clin Oncol
 
2014
;
140
:
319
328
.

32

Shu
 
Z
,
Ding
 
D
,
Li
 
Y
.
Adjuvant chemotherapy for chemotherapy-naive advanced gastric cancer patients with oxaliplatin plus tegafur versus oxaliplatin plus S-1
.
Int J Clin Exp Med
 
2017
;
10
:
9322
9329
.

33

Al-Batran
 
SE
,
Hartmann
 
JT
,
Probst
 
S
et al.
Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: A study of the Arbeitsgemeinschaft Internistische Onkologie
.
J Clin Oncol
 
2008
;
26
:
1435
1442
.

34

Enzinger
 
PC
,
Burtness
 
BA
,
Niedzwiecki
 
D
et al.
CALGB 80403 (Alliance)/E1206: A randomized phase II study of three chemotherapy regimens plus cetuximab in metastatic esophageal and gastroesophageal junction cancers
.
J Clin Oncol
 
2016
;
34
:
2736
2742
.

35

Lu
 
J
,
Zhou
 
Z
,
Wu
 
C
et al.
S-1 combined with docetaxel (SD) versus S-1 combined with cisplatin (SP) in the treatment of advanced gastric cancer: Randomized, controlled, multicenter and clinical study
.
Int J Clin Exp Med
 
2019
;
12
:
9359
9365
.

36

Yamada
 
Y
,
Boku
 
N
,
Mizusawa
 
J
et al.
Docetaxel plus cisplatin and S-1 versus cisplatin and S-1 in patients with advanced gastric cancer (JCOG1013): An open-label, phase 3, randomised controlled trial
.
Lancet Gastroenterol Hepatol
 
2019
;
4
:
501
510
.

37

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
:
149
156
.

38

Tabernero
 
J
,
Hoff
 
PM
,
Shen
 
L
et al.
Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): Final analysis of a double-blind, randomised, placebo-controlled phase 3 study
.
Lancet Oncol
 
2018
;
19
:
1372
1384
.

39

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
:
620
627
.

40

Fuchs
 
CS
,
Shitara
 
K
,
Di Bartolomeo
 
M
et al.
Ramucirumab with cisplatin and fluoropyrimidine as first-line therapy in patients with metastatic gastric or junctional adenocarcinoma (RAINFALL): A double-blind, randomised, placebo-controlled, phase 3 trial
.
Lancet Oncol
 
2019
;
20
:
420
435
.

41

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
:
1224
1235
.

42

Fuchs
 
CS
,
Tomasek
 
J
,
Yong
 
CJ
et al.
Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): An international, randomised, multicentre, placebo-controlled, phase 3 trial
.
Lancet
 
2014
;
383
:
31
39
.

43

Yi
 
JH
,
Lee
 
J
,
Lee
 
J
et al.
Randomised phase II trial of docetaxel and sunitinib in patients with metastatic gastric cancer who were previously treated with fluoropyrimidine and platinum
.
Br J Cancer
 
2012
;
106
:
1469
1474
.

44

Higuchi
 
K
,
Tanabe
 
S
,
Shimada
 
K
et al.
Biweekly irinotecan plus cisplatin versus irinotecan alone as second-line treatment for advanced gastric cancer: A randomised phase III trial (TCOG GI-0801/BIRIP trial)
.
Eur J Cancer
 
2014
;
50
:
1437
1445
.

45

Kim
 
JY
,
Ryoo
 
HM
,
Bae
 
SH
et al.
Multi-center randomized phase II study of weekly docetaxel versus weekly docetaxel-plus-oxaliplatin as a second-line chemotherapy for patients with advanced gastric cancer
.
Anticancer Res
 
2015
;
35
:
3531
3536
.

46

Satoh
 
T
,
Xu
 
RH
,
Chung
 
HC
et al.
Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN–a randomized, phase III study
.
J Clin Oncol
 
2014
;
32
:
2039
2049
.

47

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
:
2306
2314
.

48

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
:
3858
3865
.

49

Ford
 
HE
,
Marshall
 
A
,
Bridgewater
 
JA
et al.
Docetaxel versus active symptom control for refractory oesophagogastric adenocarcinoma (COUGAR-02): An open-label, phase 3 randomised controlled trial
.
Lancet Oncol
 
2014
;
15
:
78
86
.

50

Nishina
 
T
,
Boku
 
N
,
Gotoh
 
M
et al.
Randomized phase II study of second-line chemotherapy with the best available 5-fluorouracil regimen versus weekly administration of paclitaxel in far advanced gastric cancer with severe peritoneal metastases refractory to 5-fluorouracil-containing regimens (JCOG0407)
.
Gastric Cancer
 
2016
;
19
:
902
910
.

51

Kang
 
YK
,
Ryu
 
MH
,
Park
 
SH
et al.
Efficacy and safety findings from DREAM: A phase III study of DHP107 (oral paclitaxel) versus i.v. paclitaxel in patients with advanced gastric cancer after failure of first-line chemotherapy
.
Ann Oncol
 
2018
;
29
:
1220
1226
.

52

Shitara
 
K
,
Takashima
 
A
,
Fujitani
 
K
et al.
Nab-paclitaxel versus solvent-based paclitaxel in patients with previously treated advanced gastric cancer (ABSOLUTE): An open-label, randomised, non-inferiority, phase 3 trial
.
Lancet Gastroenterol Hepatol
 
2017
;
2
:
277
287
.

53

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
:
4438
4444
.

54

Roy
 
AC
,
Park
 
SR
,
Cunningham
 
D
et al.
A randomized phase II study of PEP02 (MM-398), irinotecan or docetaxel as a second-line therapy in patients with locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma
.
Ann Oncol
 
2013
;
24
:
1567
1573
.

55

Sym
 
SJ
,
Hong
 
J
,
Park
 
J
et al.
A randomized phase II study of biweekly irinotecan monotherapy or a combination of irinotecan plus 5-fluorouracil/leucovorin (mFOLFIRI) in patients with metastatic gastric adenocarcinoma refractory to or progressive after first-line chemotherapy
.
Cancer Chemother Pharmacol
 
2013
;
71
:
481
488
.

56

Tanabe
 
K
,
Fujii
 
M
,
Nishikawa
 
K
et al.
Phase II/III study of second-line chemotherapy comparing irinotecan-alone with S-1 plus irinotecan in advanced gastric cancer refractory to first-line treatment with S-1 (JACCRO GC-05)
.
Ann Oncol
 
2015
;
26
:
1916
1922
.

57

Nishikawa
 
K
,
Fujitani
 
K
,
Inagaki
 
H
et al.
Randomised phase III trial of second-line irinotecan plus cisplatin versus irinotecan alone in patients with advanced gastric cancer refractory to S-1 monotherapy: TRICS trial
.
Eur J Cancer
 
2015
;
51
:
808
816
.

58

Nakanishi
 
K
,
Kobayashi
 
D
,
Mochizuki
 
Y
et al.
Phase II multi-institutional prospective randomized trial comparing S-1 plus paclitaxel with paclitaxel alone as second-line chemotherapy in S-1 pretreated gastric cancer (CCOG0701)
.
Int J Clin Oncol
 
2016
;
21
:
557
565
.

59

Shitara
 
K
,
Yuki
 
S
,
Tahahari
 
D
et al.
Randomised phase II study comparing dose-escalated weekly paclitaxel vs standard-dose weekly paclitaxel for patients with previously treated advanced gastric cancer
.
Br J Cancer
 
2014
;
110
:
271
277
.

60

Fushida
 
S
,
Kinoshita
 
J
,
Kaji
 
M
et al.
Paclitaxel plus valproic acid versus paclitaxel alone as second-or third-line therapy for advanced gastric cancer: A randomized phase II trial
.
Drug Des Devel Ther
 
2016
;
10
:
2353
2358
.

61

Kang
 
JH
,
Lee
 
SI
, Lim Det 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
:
1513
1518
.

62

Satoh
 
T
,
Lee
 
KH
,
Rha
 
SY
et al.
Randomized phase II trial of nimotuzumab plus irinotecan versus irinotecan alone as second-line therapy for patients with advanced gastric cancer
.
Gastric Cancer
 
2015
;
18
:
824
832
.

63

Muro
 
K
,
Oh
 
SC
,
Shimada
 
Y
et al.
Subgroup analysis of East Asians in RAINBOW: A phase 3 trial of ramucirumab plus paclitaxel for advanced gastric cancer
.
J Gastroenterol Hepatol
 
2016
;
31
:
581
589
.

64

Shitara
 
K
,
Muro
 
K
,
Shimada
 
Y
et al.
Subgroup analyses of the safety and efficacy of ramucirumab in Japanese and Western patients in RAINBOW: A randomized clinical trial in second-line treatment of gastric cancer
.
Gastric Cancer
 
2016
;
19
:
927
938
.

65

Xu
 
RH
,
Zhang
 
YQ
,
Pan
 
H
et al.
RAINBOW-Asia: A randomized, multicenter, double-blind, phase III study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma following disease progression on first-line chemotherapy with platinum and fluoropyrimidine
.
J Clin Oncol
 
2021
;
39
(
suppl 3
):
199a
.

66

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
:
640
653
.

67

Ferry
 
DR
,
Dutton
 
SJ
,
Mansoor
 
W
et al.
Phase III multi-centre, randomised, double-blind, placebo-controlled trial of gefitinib versus placebo in esophageal cancer progressing after chemotherapy, COG (cancer oesophagus gefitinib)
.
Ann Oncol
 
2012
;
23
(
suppl 9
):
IXE12
.

68

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
:
894
904
.

69

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
:
1637
1651
.

70

Bang
 
YJ
,
Cho
 
JY
,
Kim
 
YH
et al.
Efficacy of sequential ipilimumab monotherapy versus best supportive care for unresectable locally advanced/metastatic gastric or gastroesophageal junction cancer
.
Clin Cancer Res
 
2017
;
23
:
5671
5678
.

71

Shitara
 
K
,
Ozguroglu
 
M
,
Bang
 
YJ
et al.
Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): A randomised, open-label, controlled, phase 3 trial
.
Lancet
 
2018
;
392
:
123
133
.

72

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
:
1316
1324
.

73

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
:
3935
.

74

Pavlakis
 
N
,
Sjoquist
 
KM
,
Martin
 
AJ
et al.
Regorafenib for the treatment of advanced gastric cancer (INTEGRATE): A multinational placebo-controlled phase II trial
.
J Clin Oncol
 
2016
;
34
:
2728
2735
.

75

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
:
2461
2471
.

76

Shitara
 
K
,
Doi
 
T
,
Dvorkin
 
M
et al.
Trifluridine/tipiracil versus placebo in patients with heavily pretreated metastatic gastric cancer (TAGS): A randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet Oncol
 
2018
;
19
:
1437
1448
.

77

Li
 
J
,
Qin
 
S
,
Xu
 
J
et al.
Apatinib for chemotherapy-refractory advanced metastatic gastric cancer: Results from a randomized, placebo-controlled, parallel-arm, phase II trial
.
J Clin Oncol
 
2013
;
31
:
3219
3225
.

78

Li
 
J
,
Qin
 
S
,
Xu
 
J
et al.
Randomized, double-blind, placebo-controlled phase III trial of apatinib in patients with chemotherapy-refractory advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction
.
J Clin Oncol
 
2016
;
34
:
1448
1454
.

79

Kato
 
K
,
Satoh
 
T
,
Muro
 
K
et al.
A subanalysis of Japanese patients in a randomized, double-blind, placebo-controlled, phase 3 trial of nivolumab for patients with advanced gastric or gastro-esophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2)
.
Gastric Cancer
 
2019
;
22
:
344
354
.

80

Janjigian
 
YY
,
Bendell
 
J
,
Calvo
 
E
et al.
CheckMate-032 study: Efficacy and safety of nivolumab and nivolumab plus ipilimumab in patients with metastatic esophagogastric cancer
.
J Clin Oncol
 
2018
;
36
:
2836
2844
.

81

Smyth
 
EC
,
Moehler
 
M
.
Late-line treatment in metastatic gastric cancer: Today and tomorrow
.
Ther Adv Med Oncol
 
2019
;
11
:1758835919867522.

82

Bang
 
YJ
,
Ruiz
 
EY
,
Van Cutsem
 
E
et al.
Phase III, randomised trial of avelumab versus physician's choice of chemotherapy as third-line treatment of patients with advanced gastric or gastro-oesophageal junction cancer: Primary analysis of JAVELIN Gastric 300
.
Ann Oncol
 
2018
;
29
:
2052
2060
.

83

Kang
 
YK
,
Kang
 
WK
,
Di Bartolomeo
 
M
et al.
Randomized phase III ANGEL study of rivoceranib (apatinib) 1 best supportive care (BSC) vs placebo 1 BSC in patients with advanced/metastatic gastric cancer who failed 2 prior chemotherapy regimens
.
Ann Oncol
 
2019
;
30
(
suppl 5
):
V877
V878
.

84

Shah
 
MA
,
Janjigian
 
YY
,
Stoller
 
R
et al.
Randomized multicenter phase II study of modified docetaxel, fisplatin, 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
:
3874
3879
.

85

Park
 
H
,
Jin
 
RU
,
Wang-Gillam
 
A
et al.
Folfirinox for the treatment of advanced gastroesophageal cancers
.
JAMA Oncol
 
2020
;
6
:
1231
.

86

Soularue
 
E
,
Cohen
 
R
,
Tournigand
 
C
et al.
Efficacy and safety of trastuzumab in combination with oxaliplatin and fluorouracil-based chemotherapy for patients with HER2-positive metastatic gastric and gastro-oesophageal junction adenocarcinoma patients: A retrospective study
.
Bull Cancer
 
2015
;
102
:
324
331
.

87

Ryu
 
MH
,
Yoo
 
C
,
Kim
 
JG
et al.
Multicenter phase II study of trastuzumab in combination with capecitabine and oxaliplatin for advanced gastric cancer
.
Eur J Cancer
 
2015
;
51
:
482
488
.

88

Klempner
 
SJ
,
Wainberg
 
ZA
,
Muro
 
K
et al.
Impact of frontline doublet versus triplet therapy on clinical outcomes: Exploratory analysis from the RAINBOW study
.
J Clin Oncol
.
2020
;
38
(
suppl 15
):
4543a
.

89

Klempner
 
SJ
,
Maron
 
SB
,
Chase
 
L
et al.
Initial report of second-line FOLFIRI in combination with ramucirumab in advanced gastroesophageal adenocarcinomas: A multi-institutional retrospective analysis
.
The Oncologist
 
2019
;
24
:
475
482
.

90

Lorenzen
 
S
,
Thuss-Patience
 
PC
,
Pauligk
 
C
et al.
FOLFIRI plus ramucirumab versus paclitaxel plus ramucirumab as second-line therapy for patients with advanced or metastatic gastroesophageal adenocarcinoma with or without prior docetaxel: Results from the phase II RAMIRIS study of the AIO
.
J Clin Oncol
 
2020
;
38
(
suppl 15
):
4514a
.

91

Park
 
H
,
Sanjeevaiah
 
A
,
Suresh
 
R
et al.
Ramucirumab and irinotecan in patients with previously treated gastroesophageal adenocarcinoma: Interim analysis of a phase II trial
.
Ann Oncol
 
2020
;
31
(
suppl 3
):
S132
.

92

Tournigand
 
C
,
Cervantes
 
A
,
Figer
 
A
et al.
OPTIMOX1: A randomized study of FOLFOX4 or FOLFOX7 with oxaliplatin in a stop-and-go fashion in advanced colorectal cancer–a GERCOR study
.
J Clin Oncol
 
2006
;
24
:
394
400
.

93

Simkens
 
LH
,
van Tinteren
 
H
,
May
 
A
et al.
Maintenance treatment with capecitabine and bevacizumab in metastatic colorectal cancer (CAIRO3): A phase 3 randomised controlled trial of the Dutch Colorectal Cancer Group
.
Lancet
 
2015
;
385
:
1843
1852
.

94

Park
 
SR
,
Kim
 
MJ
,
Nam
 
BH
et al.
A randomised phase II study of continuous versus stop-and-go S-1 plus oxaliplatin following disease stabilisation in first-line chemotherapy in patients with metastatic gastric cancer
.
Eur J Cancer
 
2017
;
83
:
32
42
.

95

Shi
 
Q
,
Paul
 
J
,
Grothey
 
A
.
Duration of adjuvant chemotherapy for stage III colon cancer
.
N Engl J Med
 
2018
;
379
:
396
397
.

96

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
:e180013.

97

Marabelle
 
A
,
Le
 
DT
,
Ascierto
 
PA
et al.
Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair–deficient cancer: Results from the phase II KEYNOTE-158 study
.
J Clin Oncol
 
2020
;
38
:
1
10
.

98

Marabelle
 
A
,
Fakih
 
M
,
Lopez
 
J
et al.
Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study
.
Lancet Oncol
 
2020
;
21
:
1353
1365
.

99

Fuchs
 
CS
,
Özgüroğlu
 
M
,
Bang
 
YJ
et al.
The association of molecular biomarkers with efficacy of pembrolizumab versus paclitaxel in patients with gastric cancer (GC) from KEYNOTE-061
.
J Clin Oncol
 
2020
;
38
(
suppl 15
):
4512a
.

100

Kim
 
ST
,
Cristescu
 
R
,
Bass
 
AJ
et al.
Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer
.
Nat Med
 
2018
;
24
:
1449
1458
.

101

Moehler
 
M
,
Dvorkin
 
M
,
Boku
 
N
et al.
Phase III trial of avelumab maintenance after first-line induction chemotherapy versus continuation of chemotherapy in patients with gastric cancers: Results from JAVELIN Gastric 100
.
J Clin Oncol
 
2021
;
39
:
966
977
.

102

Shitara
 
K
,
Van Cutsem
 
E
,
Bang
 
YJ
et al.
Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: The KEYNOTE-062 phase 3 randomized clinical trial
.
JAMA Oncol
 
2020
;
6
:
1571
1580
.

103

Boku
 
N
,
Ryu
 
MH
,
Oh
 
DY
et al.
LBA7_PR Nivolumab plus chemotherapy versus chemotherapy alone in patients with previously untreated advanced or recurrent gastric/gastroesophageal junction (G/GEJ) cancer: ATTRACTION-4 (ONO-4538-37) study
.
Ann Oncol
 
2020
;
31
(
suppl 4
):
S1192a
.

104

Janjigian
 
YY
,
Shitara
 
K
,
Moehler
 
M
et al.
First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): A randomised, open-label, phase 3 trial
.
Lancet
 
2021
;
398
:
27
40
.

105

Chao
 
J
,
Fuchs
 
CS
,
Shitara
 
K
et al.
Assessment of pembrolizumab therapy for the treatment of microsatellite instability-high gastric or gastroesophageal junction cancer among patients in the KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 clinical trials
.
JAMA Oncol
 
2021
;
7
:
895
902
.

106

Makiyama
 
A
,
Sukawa
 
Y
,
Kashiwada
 
T
et al.
Randomized, phase II study of trastuzumab beyond progression in patients with HER2-positive advanced gastric or gastroesophageal junction cancer: WJOG7112G (T-ACT Study)
.
J Clin Oncol
 
2020
;
38
:
1919
1927
.

107

Shitara
 
K
,
Bang
 
YJ
,
Iwasa
 
S
et al.
Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer
.
N Engl J Med
 
2020
;
382
:
2419
2430
.

108

Catenacci
 
DVT
,
Moya
 
S
,
Lomnicki
 
S
et al.
Personalized antibodies for gastroesophageal adenocarcinoma (PANGEA): A phase 2 study evaluating an individualized treatment strategy for metastatic disease
.
Cancer Discov
 
2021
;
11
:
308
325
.

109

Ryu
 
MH
,
Baba
 
E
,
Lee
 
KH
et al.
Comparison of two different S-1 plus cisplatin dosing schedules as first-line chemotherapy for metastatic and/or recurrent gastric cancer: A multicenter, randomized phase III trial (SOS)
.
Ann Oncol
 
2015
;
26
:
2097
2101
.

110

Shen
 
L
,
Li
 
J
,
Xu
 
J
et al.
Bevacizumab plus capecitabine and cisplatin in Chinese patients with inoperable locally advanced or metastatic gastric or gastroesophageal junction cancer: Randomized, double-blind, phase III study (AVATAR study)
.
Gastric Cancer
 
2015
;
18
:
168
176
.

111

Curran
 
D
,
Pozzo
 
C
,
Zaluski
 
J
et al.
Quality of life of palliative chemotherapy naive patients with advanced adenocarcinoma of the stomach or esophagogastric junction treated with irinotecan combined with 5-fluorouracil and folinic acid: Results of a randomised phase III trial
.
Qual Life Res
 
2009
;
18
:
853
861
.

112

Kim
 
ST
,
Kang
 
JH
,
Lee
 
J
et al.
Simvastatin plus capecitabine-cisplatin versus placebo plus capecitabine-cisplatin in patients with previously untreated advanced gastric cancer: A double-blind randomised phase 3 study
.
Eur J Cancer
 
2014
;
50
:
2822
2830
.

113

Li
 
YH
,
Qiu
 
MZ
,
Xu
 
JM
et al.
S-1 plus cisplatin versus fluorouracil plus cisplatin in advanced gastric or gastro-esophageal junction adenocarcinoma patients: A pilot study
.
Oncotarget
 
2015
;
6
:
35107
35115
.

114

Lordick
 
F
,
Kang
 
YK
,
Chung
 
HC
et al.
Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): A randomised, open-label phase 3 trial
.
Lancet Oncol
 
2013
;
14
:
490
499
.

115

Shah
 
MA
,
Xu
 
RH
,
Bang
 
YJ
et al.
HELOISE: Phase IIIb randomized multicenter study comparing standard-of-care and higher-dose trastuzumab regimens combined with chemotherapy as first-line therapy in patients with human epidermal growth factor receptor 2-positive metastatic gastric or gastroesophageal junction adenocarcinoma
.
J Clin Oncol
 
2017
;
35
:
2558
2567
.

116

Lee
 
KW
,
Kim
 
BJ
,
Kim
 
MJ
et al.
A multicenter randomized phase II study of docetaxel vs. docetaxel plus cisplatin vs. docetaxel plus S-1 as second-line chemotherapy in metastatic gastric cancer patients who had progressed after cisplatin plus either S-1 or capecitabine
.
Cancer Res Treat
 
2017
;
49
:
706
716
.

117

Lee
 
KW
,
Maeng
 
CH
,
Kim
 
TY
et al.
A phase III study to compare the efficacy and safety of paclitaxel versus irinotecan in patients with metastatic or recurrent gastric cancer who failed in first-line therapy (KCSG ST10-01)
.
The Oncologist
 
2019
;
24
:
18
e24
.

118

Moehler
 
M
,
Gepfner-Tuma
 
I
,
Maderer
 
A
et al.
Sunitinib added to FOLFIRI versus FOLFIRI in patients with chemorefractory advanced adenocarcinoma of the stomach or lower esophagus: A randomized, placebo-controlled phase II AIO trial with serum biomarker program
.
BMC Cancer
 
2016
;
16
:
699
.

119

Satoh
 
T
,
Kang
 
YK
,
Chao
 
Y
et al.
Exploratory subgroup analysis of patients with prior trastuzumab use in the ATTRACTION-2 trial: A randomized phase III clinical trial investigating the efficacy and safety of nivolumab in patients with advanced gastric/gastroesophageal junction cancer
.
Gastric Cancer
 
2020
;
23
:
143
153
.

Author notes

Contributed equally.

Disclosures of potential conflicts of interest may be found at the end of this article.

This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.