Brave new world—new and emerging treatments for gastric cancer
Review Article

Brave new world—new and emerging treatments for gastric cancer

Emer Lynch1, Austin G. Duffy1, Ronan J. Kelly2

1Department of Medical Oncology, The Mater Hospital, Dublin, Ireland; 2The Charles A. Sammons Cancer Center at Baylor University Medical Center, Dallas, TX, USA

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Dr. Ronan J. Kelly. Director of the Charles A. Sammons Cancer Center at Baylor University Medical Center, 3420 Worth Street, Suite 550, Dallas, TX 75246, USA. Email: Ronan.Kelly@bswhealth.org.

Abstract: Despite considerable international heterogeneity in the incidence and histological subtypes of gastric cancer (GC), in addition to more recent epidemiological trends, chemotherapy has long represented the main systemic therapeutic option in its treatment. For the roughly 20% of GC with human epidermal growth factor receptor 2 (HER2) overexpression, there is a more recently established role for the addition of HER2+ based therapy in the form of trastuzumab. However, while immune checkpoint inhibitors (ICIs) have revolutionised the treatment of other malignancies including melanoma and renal cell carcinoma over the past decade, they have only gained a foothold in GC in more recent years. This article reviews the existing evidence for ICIs in GC as a novel therapeutic option. It also looks to ongoing trials of immune checkpoint inhibition both in the perioperative and advanced setting, and in combination with other therapeutic targets including HER2+. Other investigational immune based therapies including chimeric antigen receptor T-cell (CAR-T) therapy and anti-T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (anti-TIGIT) therapy are considered, in addition to reviewing the building evidence for alternative therapeutic targets currently under investigation in GC, including fibroblast growth factor receptor 2b (FGFR2b) and claudin 18.2 amongst others. These novel and evolving targets represent a brave new world in therapeutic intervention in GC, with the potential to transform outcomes for patients internationally.

Keywords: Immunotherapy; gastric adenocarcinoma; immune checkpoint inhibitors (ICIs); gastroesophageal junction adenocarcinoma (GEJ adenocarcinoma)


Submitted Feb 20, 2023. Accepted for publication Aug 17, 2023. Published online Aug 28, 2023.

doi: 10.21037/cco-23-13


Background

Gastric cancer (GC) is one of the most common cancers worldwide, ranking 5th in global incidence (1). There are regional variations with regard to incidence and anatomical location. Within United States (US) and Western populations, there has been an increase in proximal and junctional adenocarcinoma with a concurrent reduction in non-cardia/distal GC. This shift has underlying aetiological drivers with proximal ‘western’ GC being associated with obesity and reflux while, for distal GC, the major driver is helicobacter pylori. As we shall discuss, these global differences impact on biology and behaviour and must be taken into account when interpreting clinical trial data. The initial foothold for chemotherapy in GC was established, in the metastatic setting, by studies which proved a (modest) benefit compared to supportive care alone and, in the perioperative setting, by the MAGIC study (2). Since then, efforts to build on chemotherapy did not have a major impact on survival times and—with the exception of human epidermal growth factor receptor 2 (HER2)-directed treatments with trastuzumab (3) or more recently trastuzumab deruxtecan (T-DXd) (4)—clinical trials evaluating so-called targeted therapies in GC were on the whole disappointing. There is now however a strong sense within the field that this has changed. New targets have emerged most notably claudin 18.2 and fibroblast growth factor receptor 2b (FGFR2b), with zolbetuximab and bemarituzumab (5) likely to be approved in the near future. The immune checkpoint inhibitor (ICI) class of drugs have had an enormous impact on the entire field of oncology and 2021 saw the first landmark studies in GC which have seen ICIs become part of the standard of care. In this review, we will discuss the impact to date of the ICI class in this disease with a focus on the advanced disease setting, and also discuss new and emerging targets and treatment strategies in GC.


Immunogenicity of GC

The immune system plays a significant role in cancer development and evolution (6,7). The Cancer Genomic Atlas (TCGA) project grouped GC into four distinct subtypes, two of which—microsatellite instable (MSI) and Epstein-Barr virus (EBV)-related—could be characterized as ‘immunogenic’ based on expression of immune-relevant genes (8). Validation of this has been seen in the striking efficacy of ICI drugs in both these subtypes with—in the MSI-high group—pathologic complete response (pCR) rates of 59% and—in the EBV group—objective radiologic responses have been seen in high numbers but all EBV positive tumors also have high programmed death-ligand 1 (PD-L1) combined positive score (CPS). Due to a lack of data at the present time, routine testing of all GCs for EBV is not recommended due to the overlap with PD-L1 positive tumors (9,10). More broadly, the extent to which the immune system reacts in response to a cancer can be inferred indirectly by pathologic variables such as tumor-infiltrating lymphocytes (TILs) and expression of PD-L1. A meta-analysis of over twenty studies found a strong correlation between presence of TILs—specifically non-regulatory T (Treg) TILs—and survival which points to an active immune role even when no immune drug is being employed (11). Treatment can of course impact on this and this is hugely important to consider in the non-MSI-high/EBV scenarios where we try to combine the ICI class with standard chemotherapy. Certain chemotherapeutic agents (e.g., oxaliplatin) appear to be more immunogenic than others (12). It is perhaps one of the disappointing aspects of the huge number of ongoing studies interrogating the combination of immune drugs with chemotherapy (or radiation) that the chemotherapy backbone, in terms of dose and schedule, is the same as that which negotiated the dose-limiting toxicity constraints of traditional trial design.


Immune approaches

First-line, ICI chemotherapy combinations

In 2021, KEYNOTE-590 became the first randomized phase 3 study in gastroesophageal cancer to demonstrate a meaningful improvement in overall survival (OS) with the addition of an ICI—pembrolizumab—to chemotherapy [5-fluorouracil (5-FU) and cisplatin] in comparison with chemotherapy alone (13). The study met its primary endpoint overall and this was consistent across the pre-specified subtypes including the 27% of patients who had adenocarcinoma and the 12% with a Siewert type 1 gastroesophageal junction (GEJ) tumor. For this subgroup, OS was longer with the addition of pembrolizumab to chemotherapy {median 11.6 [95% confidence interval (CI): 9.7–15.2] vs. 9.9 (95% CI: 7.8–12.3) months; hazard ratio (HR): 0.74, 95% CI: 0.54–1.02}. Subsequently, at the 2022 American Society of Clinical Oncology (ASCO) Gastrointestinal Symposium further updated results were presented. These demonstrated that median OS in the esophageal squamous cell carcinoma (ESCC) cohort was 12.6 months (HR: 0.73, 95% CI: 0.61–0.88) in comparison with 11.6 months in those with adenocarcinoma (HR: 0.73, 95% CI: 0.55–0.99). Of note, the updated data demonstrated better OS regardless of histopathology, which had not been seen in the earlier results (14). The US Food and Drug Administration (FDA) approval for this regimen however is limited to esophageal [both ESCC and esophageal adenocarcinoma (EAC)] and Siewert class 1 GEJ tumors. KEYNOTE-590 did not enrol any GCs and to date, pembrolizumab has not been licensed for this indication. However, this may imminently change with the recent presentation of the positive results from the phase III KEYNOTE-859 trial at the European Society for Medical Oncology (ESMO) virtual plenary session in February 2023, comparing pembrolizumab plus chemotherapy to chemotherapy alone as first-line therapy in gastric and GEJ adenocarcinoma (15).

Similarly, both CheckMate-649 and ATTRACTION-4 investigated the addition of nivolumab to chemotherapy in the first-line setting with a focus on adenocarcinoma (16,17). CheckMate-649 has three arms comparing nivolumab in combination with oxaliplatin-based chemotherapy, with ipilimumab and nivolumab and with chemotherapy alone. The primary endpoint assessable in patients whose tumors were PD-L1-postive (defined on amendment as a CPS of five or more). N=1,581 patients were randomized, and about 60% demonstrated PD-L1 CPS of ≥5. The majority of patients were non-Asian (76%) and—in contrast to the KEYNOTE-590 study mentioned above—most (70%) had GC. Nivolumab in combination with oxaliplatin based chemotherapy resulted in significant improvements in OS (HR: 0.71, 98.4% CI: 0.59–0.86, P<0.0001) and progression-free survival (PFS) (HR: 0.68, 98.4% CI: 0.56–0.81, P<0.0001) vs. chemotherapy alone in the enriched population (PD-L1 ≥5) although improvement was also seen with a PD-L1 CPS ≥1 who were enrolled. Updated results were presented at ESMO 2021, to include the ipilimumab and nivolumab arm. A secondary endpoint of OS in PD-L1 CPS ≥5 in the combined immunotherapy arm in comparison with chemotherapy was not met, and this calls into question the role—if any—of CTLA-4 inhibition as a strategy moving forwards in gastric or GEJ cancers (18). Although the US FDA approved the addition of nivolumab without limitation to CPS ≥5, the National Comprehensive Cancer Network (NCCN) has given category 1 evidence to CPS ≥5 but only category 2B evidence for those patients with CPS ≥1. In comparison, the European Medicines Agency (EMA) has only afforded approval where CPS ≥5. ATTRACTION-4, like CHECKMATE 649, combines nivolumab with oxaliplatin-based chemotherapy—but in an Asian population and the primary endpoints were applied regardless of PD-L1 expression albeit with four stratification blocks for CPS (19). Ninety percent of participants had GC. While the addition of nivolumab improved PFS [10.45 (95% CI: 8.44–14.75) vs. 8.34 (95% CI: 6.97–9.40) months] OS data was not different [17.45 (95% CI: 15.67–20.83) vs. 17.15 (95% CI: 15.18–19.65) months], a finding that many ascribe to the high numbers of patients who received an ICI as part of subsequent therapy.

Interim analysis results from ORIENT-16 were presented at ESMO 2021, comparing sintilimab, a programmed cell death protein 1 (PD-1) inhibitor, in combination with chemotherapy vs. chemotherapy alone (20). In total, 61.1% of patients had a CPS ≥5. Median OS was greater for sintilimab plus chemotherapy vs. chemotherapy alone both in those with CPS ≥5 (18.4 vs. 12.9 months; HR: 0.660, 95% CI: 0.505–0.864, P=0.0023) and for all patients (15.2 vs. 12.3 months; HR: 0.766, 95% CI: 0.626–0.936, P=0.0090). Greater median PFS was also observed for sintilimab plus chemotherapy vs. chemotherapy alone with CPS ≥5 (HR: 0.628, 95% CI: 0.489–0.805, P=0.0002) and again in all patients (HR: 0.636, 95% CI: 0.525–0.771, P<0.0001).

Similarly, positive interim analysis results from the PD-L1 positive population from the phase III Rationale 305 trial were presented at ASCO Gastrointestinal in January 2023 (21). A total of 274 patients with PD-L1 positive unresectable and previously untreated gastric or GEJ adenocarcinoma were randomized to either tislelizumab, an anti-PD1 monoclonal antibody, or placebo, in combination with chemotherapy. Chemotherapy was investigator chosen, either oxaliplatin plus capecitabine or cisplatin and 5-FU. The experimental arm demonstrated improved median OS vs. chemotherapy alone, at 17.2 vs. 12.6 months (HR: 0.74, 95% CI: 0.59–0.94, one-sided P=0.0056), in addition to superior median PFS (7.2 vs. 5.9 months; HR: 0.67, 95% CI: 0.55–0.83). Higher overall response rates (ORRs) were also seen in the tislelizumab arm with 50.4% vs. 43%.

Finally, results from KEYNOTE-859 were presented as the ESMO 2023 virtual plenary, cementing the role of up-front chemotherapy with immunotherapy in previously untreated HER2-negative gastric or GEJ adenocarcinoma. A total of 1,579 patients were randomized to either pembrolizumab or placebo 3 weekly for up to 35 cycles with chemotherapy [5-FU with cisplatin or capecitabine with oxaliplatin (CAPOX)]. At median study follow-up of 31 (range, 15.3–46.3) months, median OS was 12.9 months in the pembrolizumab arm in comparison with 11.5 months in the placebo arm (HR: 0.78, 95% CI: 0.70–0.87, P<0.0001). Median PFS was also greater in the immunotherapy arm at 6.9 vs. 5.6 months with placebo (HR: 0.76, 95% CI: 0.67–0.85, P<0.0001). Pembrolizumab with chemotherapy demonstrated an of ORR of 51.3% in comparison with 42.0% for those receiving placebo with chemotherapy (P=0.00009). Efficacy in the full intention-to-treat (ITT) cohort was presented with results largely consistent across the subgroups including PD-L1 CPS status, however specific subgroup analysis data for the PD-L1 subgroups will be presented at a later date (15).

ICI monotherapy

The entry point for the ICI class into the clinic has been either as a combination with chemotherapy in the front-line setting or as monotherapy in patients who had been previously treated (often with multiple lines of therapy). The initial data were for pembrolizumab in this latter setting. KEYNOTE-012 (22) was a phase 1b trial in Asia in PD-L1-positive recurrent or metastatic adenocarcinoma of the stomach or GEJ and had a response rate of 22%. This led to a larger, phase 2 study—KEYNOTE-059—which investigated third or later line pembrolizumab in previously treated metastatic GC (23). The ORR was 15.5% and 6.4% in those with PD-L1 positive and negative tumors respectively, with positivity being defined as greater or equal to one. As a result, pembrolizumab was approved by the US FDA in September 2017 for this indication, however approval was later withdrawn in 2021 following a recommendation by the Oncology Drugs Advisory Committee (ODAC). The ODAC recommendation was informed by the results of two pembrolizumab studies, KEYNOTE-062 (24)—a front-line study discussed below—and KEYNOTE-061 (25), which compared pembrolizumab with paclitaxel in patients with PD-L1-positive (defined as CPS ≥1) advanced gastric/GEJ cancer that had progressed on first-line chemotherapy. The median OS was 9.1 (95% CI: 6.2–10.7) and 8.3 (95% CI: 7.6–9.0) months for pembrolizumab and paclitaxel respectively with median PFS favoring paclitaxel 4.1 vs. 1.5 months.

Similarly, nivolumab also first demonstrated activity in previously treated gastric and GEJ malignancies in the ATTRACTION-2 study (26). This was a phase III study in an Asian population randomizing n=493 patients to nivolumab or placebo and demonstrated a modest improvement in median PFS (1.61 vs. 1.45 months; HR: 0.60, P<0.0001) and OS (5.26 vs. 4.14 months; HR: 0.63, P<0.0001). The landmark endpoints demonstrate a 12-month OS rate in favor of nivolumab (26.6% vs. 10.9%). ORR was 11.2% (vs. 0% in the placebo group), with a median duration of response (DOR) of 9.53 months. A retrospective multivariate analysis by Kim et al. showed that PD-L1 positivity and low neutrophil-lymphocyte ratio were predictors of benefit (27). Longer follow-up showed that the median OS of responders (n=32) was 26.7 months with a 3-year survival rate of 35.5% (28). However, although Japan afforded regulatory approval for nivolumab in this indication regardless of PD-L1 status in October 2017, these data were insufficient for EMA approval.

Encouraging pre-clinical data exists to support the investigation of combined immune checkpoint inhibition in GCs (29,30), and dual cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and PD-L1 inhibition is established in the first-line management of other malignancies including melanoma and renal cell carcinoma (31,32). CheckMate-032 evaluated the safety and efficacy of single agent nivolumab or combination nivolumab with ipilimumab at different dose and schedules in n=160 patients with advanced esophagogastric cancer (33). Results demonstrated promising effectiveness with durable responses and encouraging long-term OS results in addition to a tolerable toxicity profile in all three schedules evaluated. As would be expected, adding anti-CTLA-4 inhibition did increase toxicity. These encouraging results prompted more comprehensive investigation in the phase III CheckMate-649 study discussed below (34). Another anti-CTLA-4 combination of durvalumab and tremelimumab was also assessed in a randomized multicentre phase II study in patients with advanced GEJ/G cancers and whilst response rates were low the 12-month OS rates were similar to those reported in CheckMate-032 (35).

The difficulty of displacing chemotherapy from its front-line position was seen in KEYNOTE-062. N=763 patients were randomized to pembrolizumab alone, pembrolizumab with chemotherapy or chemotherapy alone, in the first-line setting in unresectable or metastatic gastric and GEJ cancer with PD-L1 CPS ≥1 (24). Though pembrolizumab alone seemed to have benefit in PD-L1 CPS >10 this was not formally evaluated/pre-specified and neither pembrolizumab or pembrolizumab plus chemotherapy was superior to chemotherapy for the OS and PFS end points tested. Of concern though was the fact that though the 12-month survival was the same for the pembrolizumab-alone arm there seemed to be worse survival in the first few months of treatment due to the lack of chemotherapy in patients who were either slow to respond or would never respond to the ICI.

The other disappointing negative study evaluating a role for monotherapy in the front-line setting—this time in the maintenance setting—was the phase III Javelin Gastric 100 study of maintenance avelumab (36). This was a global (23% Asian), open-label study patients with untreated, unresectable, HER2-negative, gastric or junctional tumors who had not progressed after 12 weeks of up front oxaliplatin-based chemotherapy. In total, 499 of 805 patients met the criteria for randomization to receive maintenance avelumab or continue chemotherapy. Median OS was 10.4 (95% CI: 9.1 to 12.0) vs. 10.9 (95% CI: 9.6 to 12.4) months and 24-month OS rate was 22.1% vs. 15.5% with avelumab vs. chemotherapy, respectively. There was no benefit seen in the pre-specified PD-L1-positive population but in an exploratory analysis of those who were PD-L1-positive population by CPS—taking into account not just tumor cells but the immune microenvironment—median OS was 14.9 (95% CI: 8.7 to 17.3) months with avelumab vs. 11.6 (95% CI: 8.4 to 12.6) months with chemotherapy (Table 1).

Table 1

Resulted trials in the advanced disease setting

Trial Type Setting Arms ORR OS PFS
KEYNOTE-590 (13,14) Esophageal/GEJ cancer; global; n=749 Phase III; first-line advanced A: Pembrolizumab + chemotherapy (cisplatin & 5-FU). B: Placebo + chemotherapy A vs. B: 45.0% vs. 29.3% (P<0.0001) in all patients A vs. B: ESCC CPS ≥10, 13.9 vs. 8.8 months; ESCC, 12.6 vs. 9.8 months; CPS ≥10, 13.5 vs. 9.4 months; all patients, 12.4 vs. 9.8 months A vs. B: ESCC, 6.3 vs. 5.8 months; CPS ≥10, 7.5 vs. 5.5 months; all patients, 6.3 vs. 5.8 months
KEYNOTE-859 (15) Gastric/GEJ adenocarcinoma; global; n=1,579 Phase III; first-line advancer A: Pembrolizumab + chemotherapy (CAPOX or FP). B: Placebo + chemotherapy A vs. B: 51.3% vs. 42.0% (P=0.00009) A: 12.9 months; B: 11.5 months (HR: 0.78, 95% CI: 0.70–0.87, P<0.0001) A: 6.9 months; B: 5.6 months (HR: 0.76, 95% CI: 0.67–0.85; P<0.0001)
CheckMate-649 (16) Esophageal, GEJ, gastric adenocarcinoma; global; n=1,581 Phase III; first-line advanced A: Nivolumab + ipilimumab. B: Nivolumab + chemotherapy (CAPOX or FOLFOX). C: Chemotherapy CPS ≥5 ORRs were higher in nivolumab-treated patients (45% vs. 60%, P<0.0001) CPS ≥5, 3.3-month gain in OS (HR: 0.71, 98.4% CI: 0.59–0.86, P<0.0001) CPS ≥5, 1.7-month gain in PFS (HR: 0.68, 98.4% CI: 0.56–0.81, P<0.0001)
ATTRACTION-4 (17) Gastric/GEJ cancer; Asian; n=724 Phase III; first-line advanced A: Nivolumab + chemotherapy (SOX or CAPOX). B: Placebo + chemotherapy A vs. B: 57.5% vs. 47.8% (P=0.0088) No statistically significant difference A vs. B: 10.5 vs. 8.3 months (HR: 0.68, 98.51% CI: 0.51–0.90, P=0.0007)
ORIENT-16 (20) Gastric/GEJ adenocarcinoma; Asian; n=650 Phase III; first-line advanced A: Sintilimab + chemotherapy (oxaliplatin + capecitabine). B: Chemotherapy alone A vs. B: CPS ≥5, 72.8% vs. 59.6%; all patients: 65.1% vs. 58.7% A vs. B: CPS ≥5, 18.4 vs. 12.9 months (HR: 0.660, 95% CI: 0.505–0.864, P=0.0023); all patients, 15.2 vs. 12.3 months (HR: 0.766, 95% CI: 0.626–0.936, P=0.0090) A vs. B: reported as superior CPS ≥5 (HR: 0.628, 95% CI: 0.489–0.805, P=0.0002); all patients (HR: 0.636, 95% CI: 0.525–0.771, P<0.0001)
Rationale 305 (21) Gastric/GEJ adenocarcinoma; global; n=546 Phase 3; advanced A: Tislelizumab + chemotherapy (oxaliplatin + capecitabine or cisplatin + 5-FU). B: Placebo + chemotherapy A vs. B: 50.4% vs. 43.0% A vs. B: median OS 17.2 vs. 12.6 months (HR: 0.74, 95% CI: 0.59–0.94, one-sided P=0.0056) A vs. B: median PFS 7.2 vs. 5.9 months (HR: 0.67, 95% CI: 0.55–0.83)
KEYNOTE-012 (22) Gastric/GEJ adenocarcinoma; global; n=39 Phase 1b ORR: 22%
KEYNOTE-059 (23) Gastric/GEJ adenocarcinoma; global; n=56 Phase II; first-line metastatic A: Pembrolizumab and cisplatin/5-FU. B: Pembrolizumab A: 60.0% (95% CI: 38.7–78.9%). B: 25.8% (95% CI: 11.9–44.6%)
KEYNOTE-062 (24) Gastric/GEJ cancer; global; n=763 Phase III; first-line metastatic A: Pembrolizumab. B: Pembrolizumab + chemotherapy (cisplatin + 5-FU or capecitabine). C: chemotherapy Pembrolizumab noninferior to chemotherapy CPS ≥1: median 10.6 vs. 11.1 months (HR: 0.91, 99.2% CI: 0.69–1.18). Pembrolizumab monotherapy not superior to chemotherapy CPS ≥1. Pembrolizumab prolonged OS vs. chemotherapy in patients with CPS ≥10: median 17.4 vs. 10.8 months (HR: 0.69, 95% CI: 0.49–0.97) CPS ≥1, 6.9 vs. 6.4 months (HR: 0.84, 95% CI: 0.70–1.02, P=0.04)
KEYNOTE-061 (25) Gastric/GEJ cancer; global; n=592 Phase III; second-line metastatic A: Pembrolizumab. B: Paclitaxel 9.1 (95% CI: 6.2–10.7) months with pembrolizumab and 8.3 (95% CI: 7.6–9.0) months with paclitaxel (HR: 0.82, 95% CI: 0.66–1.03, one-sided P=0.0421) Median PFS 1·5 (95% CI: 1.4–2.0) months with pembrolizumab and 4.1 (95% CI: 3.1–4.2) months with paclitaxel (HR: 1.27, 95% CI: 1.03–1.57)
ATTRACTION-2 (26) Gastric/GEJ cancer; Asian; n=493 Phase III; refractory disease A: Nivolumab. B: Placebo A vs. B: 5.26 (95% CI: 4.60–6.37) vs. 4.14 (95% CI: 3.42–4.86) at 2-year follow-up
CheckMate-032 (33) Esophageal, GEJ, GC; US/Europe; n=160 Phase II; refractory disease A: NIVO3. B: NIVO1/IPI3. C: NIVO3/IPI1 A: 12%. B: 24%. C: 8% 12-month OS rates: A: 39%; B: 35%; C: 24% 12-month PFS rates: A: 8%; B: 17%; C: 10%
Javelin Gastric 100 (36) Gastric/GEJ cancer; global; n=805 Phase III; first-line maintenance metastatic A: Avelumab. B: Chemotherapy (continue oxaliplatin and 5-FU) A vs. B: median OS was 10.4 vs. 10.9 months; 24-month OS rate was 22.1% vs. 15.5%; PD-L1-positive population the HR for OS was 1.13; CPS ≥1 median OS was 14.9 vs. 11.6 months
LEAP-015 Phase III; gastroesophageal adenocarcinoma; n=15 (run in safety data) First-line A: Lenvatinib + pembrolizumab + chemotherapy (CAPOX or mFOLFOX). B: Chemotherapy ORR: 73% Disease control rate 93%

ORR, overall response rate; OS, overall survival; PFS, progression-free survival; GEJ, gastroesophageal junction; 5-FU, 5-fluorouracil; ESCC, esophageal squamous cell carcinoma; CPS, combined positive score; CAPOX, capecitabine with oxaliplatin; FP, infusional 5-fluorouracil; HR, hazard ratio; CI, confidence interval; FOLFOX, folinic acid, fluorouracil, and oxaliplatin; SOX, S-1 with oxaliplatin; GC, gastric cancer; NIVO, nivolumab; IPI, ipilimumab; mFOLFOX, modified FOLFOX.

Ongoing IO trials of note

Anti-vascular endothelial growth factor (anti-VEGF)

The combination of anti-angiogenic agents with ICIs has received a lot of attention in recent years with an increased understanding of the role of VEGF and other ‘pro-angiogenic’ mediators in maintaining an immune-suppressed microenvironment which facilitates immune-escape (37). The combination of lenvatinib—a multikinase inhibitor against VEGFR1, 2, and 3, fibroblast growth factor receptor (FGFR)1, 2, 3, and 4, platelet-derived growth factor receptor (PDGFR) alpha, c-Kit and rearranged during transfection (RET)—and pembrolizumab in particular has shown promising activity in endometrial and liver cancer (38,39). The LEAP-015 trial (NCT0466271) is a randomized phase III study evaluating this combination in patients with advanced/metastatic gastroesophageal adenocarcinoma (40). Safety run-in data was presented at ESMO 2022 which assessed the tolerability, safety and efficacy of the combination. N=15 patients were enrolled with an ORR of 73% demonstrated and a disease control rate of 93% (41). Part 2 of the study, which is open-label and randomized, is currently enrolling.

T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT)

TIGIT is an immune checkpoint which inhibits activation of T cells and natural killer cells and has been shown to play a role in tumor immune-surveillance (42). As such, the biologic rationale for anti-TIGIT antibodies in GC is similar to that for anti-PD-1 therapy (43). Pre-clinical studies have revealed a synergistic anti-tumor effect by combining anti-TIGIT antibodies with anti-PD-L1 ICIs, and up to 80 clinical trials are currently underway investigating anti-TIGIT antibodies across a host of solid tumors and hematological malignancies. Although phase II data for anti-TIGIT antibody in combination with anti-PD-L1 therapy in non-small cell lung cancer has been recently presented and shows greater promise in those with greater levels of PD-L1 expression (44), trials assessing anti-TIGIT therapy in combination with anti-PD-1/PD-L1 therapy in GC are currently in very early stages. These include SKYSCRAPER-11, a phase 2 trial which proposes to evaluate the safety and efficacy of tiragolumab (anti-TIGIT) and atezolizumab in patients with advanced solid tumors where anti-PD-1 therapy would be considered appropriate therapy (45), in addition to gastric/GEJ adenocarcinoma specific studies NCT04933227 (46) which looks at atezolizumab plus tiragolumab and chemotherapy in the first-line advanced disease setting and NCT05702229 (47), which will assess anti-TIGIT therapy with anti-PD-1 therapy.

Lymphocyte activation gene 3 (LAG-3)

LAG-3 is an immune checkpoint mediator limiting effector T-cell function and has been demonstrated to show expression in gastric carcinomas. CA224-060 is a phase II study which has completed recruitment and intends to assess the efficacy of relatlimab with nivolumab and chemotherapy in the first-line treatment of gastric and gastroesophageal adenocarcinoma (48).

Chimeric antigen receptor T-cell (CAR-T)

CAR-T works by utilizing viral vectors to introduce CARs to T-cells, thereby allowing the T-lymphocytes to identify tumor-associated antigens (such as surface glycoproteins) without the need for processing and presentation in context of major histocompatibility molecules (49). It has been used to great success in some hematological malignancies however as yet has not established a formal role in solid organ malignancies. Given the proposed promise of claudin 18.2 as a target in GC, which is outlined in greater detail below, CAR-T therapy using genetically engineered T-lymphocytes expressing claudin 18.2-targeted CAR has been developed. Interim phase I data has been published in previously treated claudin 18.2 positive cancers, including 28 patients with GC or GEJ cancer (50). In the GC cohort, an ORR of 57% (95% CI: 37.2%, 75.5%) was seen. In total, 42.9% of the gastric/GEJ cancer patients had received prior anti-PD-1 or anti-PD-L1 therapy previously. Additionally, although early phase data, an ORR of 70% was seen across all malignancy types in those without PD-L1 expression. As such, this represents an area of great promise for both immunogenic and low PD-L1 GC, notwithstanding the expense and toxicity of CAR-T therapy in its current form.

Perioperative/neoadjuvant use of ICIs

We have seen in lung cancer that the benefits of ICI seem to be greater when given earlier in the treatment paradigm, specifically pre-operatively (51). Neoadjuvant administration of ICIs is appealing for a variety of immunological reasons, with a potential increased efficiency of priming against in-situ tumor antigens leading to activated T-cells and better surveillance after surgery (52). In addition, the immune response is not complicated by the dramatic changes in VEGF levels and other mediators of wound healing that will occur in the days and weeks after surgery. In the GC area CHECKMATE 577 was practice-changing, though it enrolled patients with esophageal or GEJ cancer (with 71% having adenocarcinoma) (53). The Dante trial evaluated atezolizumab in combination with fluorouracil plus leucovorin, oxaliplatin, and docetaxel (FLOT) chemotherapy with interim results presented at ASCO 2022 (54). 295 patients were randomized with the addition of immunotherapy demonstrating greater downsizing (pT0 23% vs. 15% and pN0 in 68% vs. 54%). Centrally assessed pCR was seen in 46% vs. 24% in those with a PD-L1 CPS ≥10, though in all-comers comparable pCR rates were seen in both arms. A number of other studies in the peri-operative setting have not yet reported. EORTC VESTIGE, is a phase II study assessing adjuvant ipilimumab and nivolumab vs. chemotherapy in patients with gastric or EAC who have had a poor response to neoadjuvant chemotherapy and have incomplete (R1) resection or metastatic lymph nodes (N+) (55). KEYNOTE-585 is an ongoing phase III trial evaluating pembrolizumab in combination with perioperative chemotherapy in those with resectable gastric/GEJ adenocarcinoma (56), although it must be highlighted that FLOT, which represents the current standard of care chemotherapy, is not mandatory. MATTERHORN is a randomized controlled phase III study investigating peri-operative FLOT ± durvalumab (57). Omitting the chemotherapy completely in the subset of patients with MSI-high GC was evaluated by the NEONIPIGA phase II study (58). This trial evaluated neoadjuvant ipilimumab (1 mg/kg every 6 weeks) for two cycles in combination with nivolumab 240 mg every 2 weeks for six cycles, followed by surgery then nivolumab 480 mg every 4 weeks for nine cycles. N=32 patients were enrolled, and all had complete endoscopic response with tumor free biopsies. Three patients did not proceed to surgery, two of whom refused and a further patient had metastatic disease at inclusion. Of the 29 that proceeded to surgery, all had a R0 resection and 58.6% had pCR. This continues to recruit as a phase II trial (Table 2).

Table 2

Resulted and ongoing trials in the peri-operative setting

Trial Type Setting Arms DFS pCR
CheckMate-577 (53) Phase III; global; n=794 Resected (R0) stage II or III esophageal or GEJ junction. Post neoadjuvant chemoradiotherapy Nivolumab vs. placebo for 1 year (randomized 2:1) At median follow-up of 24.4 months. A vs. B: 22.4 (95% CI: 16.6 to 34.0) vs. 11.0 (95% CI: 8.3 to 14.3) months
DANTE (54) Phase IIb; n=295 (interim results) Resectable gastric/GEJ adenocarcinoma (cT2 and/or node positive) A: FLOT + atezolizumab ×4 cycles pre- and 4 cycles post-operative then maintenance atezolizumab. B: 4+4 cycles perioperative FLOT Downsizing favored arm A (pT0 in 23% vs. 15%; pN0 68% vs. 54%). Greater pathological regression rates seen in arm A vs. arm B
EORTC VESTIGE (55) Phase 2 Gastric/GEJ adenocarcinoma. Stage Ib–IVa post perioperative chemotherapy A: Ipilimumab and nivolumab ×1 year. B: Postoperative chemotherapy as per ESMO guidelines
KEYNOTE-585 (56) Phase III; global Stage II–IV previously untreated, localised gastric/GEJ adenocarcinoma with plan for surgery after preoperative chemotherapy A: Neoadjuvant ×3 and adjuvant ×3 chemotherapy (cisplatin + 5-FU ORR cisplatin + capecitabine with pembrolizumab then adjuvant pembrolizumab ×11 cycles. B: Neoadjuvant and adjuvant chemotherapy + placebo
MATTERHORN (57) Phase III; global; aims to randomize 900 patients Resectable gastric/GEJ adenocarcinoma. ≥ Stage II A: Neoadjuvant + adjuvant chemotherapy (FLOT) + durvalumab then maintenance durvalumab ×10 cycles. B: Neoadjuvant and adjuvant FLOT with placebo
NEONIPIGA (58) Phase II; n=32 dMMR/MSI-high gastric/GEJ adenocarcinoma. T2–T4NxM0 Neoadjuvant nivolumab and ipilimumab ×12 weeks then adjuvant nivolumab ×9 months Of 29 patients who proceeded to surgery all had an R0 resection, 58.6% (90% CI: 41.8% to 74.1%) had pCR

DFS, disease-free survival; pCR, pathologic complete response; GEJ, gastroesophageal junction; CI, confidence interval; FLOT, fluorouracil plus leucovorin, oxaliplatin, and docetaxel; 5-FU, 5-fluorouracil; ORR, overall response rate; dMMR, deficient mismatch repair; MSI, microsatellite instable.


Non-immune/immune-adjacent approaches

While there are continued advances in the immune-based therapy in GC, targeted therapy in this field has not quite kept the pace with a couple of exceptions. Over a decade ago, the ToGA trial demonstrated improved OS by adding trastuzumab to chemotherapy in the approximately 20% of gastroesophageal adenocarcinomas that are HER2-positive (3). Building on that success and introducing an antibody drug conjugate (ADC) to the GC treatment paradigm for the first time is T-DXd, an ADC consisting of an anti-HER2 antibody linked to a cytotoxic topoisomerase I inhibitor (4). DESTINY-Gastric01 is an open-label randomized phase 2 trial assessing T-DXd against chemotherapy in HER2-positive advanced GC previously treated with at least two lines of systemic therapy, including trastuzumab. N=125 patients were randomized 2:1 to T-DXd or chemotherapy. ORR of 51% with T-DXd was noted in comparison to 14% with chemotherapy (P<0.001). OS was 12.5 months in the T-DXd cohort vs. 8.4 months with chemotherapy [HR: 0.59, 95% CI: 0.39–0.88, P=0.01]. A phase 3 trial is currently recruiting. In a similar vein, DESTINY-Gastric03 is a phase 1b/II dose escalation and dose expansion trial with multiple arms investigating T-DXd as a single agent in addition to in combination with chemotherapy and/or anti-PD-1/PD-L1 therapy (59). Promising preliminary data demonstrating ORR of 50% was presented at ASCO Gastrointestinal in February 2022 and further results are eagerly awaited.

The fibroblast growth factor (FGF)/FGFR pathway plays a role in both angiogenesis and tumor cell proliferation. Alterations in the FGF/FGFR pathway, including FGFR2 gene amplification and FGFR2 overexpression, have been noted in gastroesophageal adenocarcinomas. It therefore represents another potential therapeutic target in GC. Bemarituzumab represents a first-in-class monoclonal antibody specific to the FGFR2b receptor, blocking FGF binding, and has demonstrated modest single agent activity in FGFR2b overexpressed advanced gastroesophageal adenocarcinoma with ORR of 17.9% (95% CI: 6.1–36.9%) (60). FIGHT is a randomized, doubled blind phase 2 trial investigating bemarituzumab in combination with modified infusional fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) vs. mFOLFOX6 with placebo in the first-line setting in FGFR2b overexpressed GC (5). Thirty percent [275] of N=910 tumors evaluated for inclusion were FGFR2b positive. N=155 patients were randomized to either mFOLFOX6 with bemarituzumab or placebo, with median PFS of 9.5 months in the bemarituzumab arm in comparison with 7.4 months in the placebo arm (HR: 0.68, 95% CI: 0.44–1.04, P=0.07). OS was the secondary endpoint, with median not yet reached in bemarituzumab arm and 12.9 months in the placebo arm (HR: 0.58, 95% CI: 0.35–0.95, P=0.03). Phase 3 data is awaited.

Furthermore, in the context of targeted therapy in GC, it is important to look again at claudin 18.2. Claudins are transmembrane proteins which form part of the tight junctions at the apical region of polarized endothelial or epithelial cells (61). Changes in their expression can result in alteration to signalling pathways and tumorigenesis. Claudin 18.2 is expressed only in the gastric epithelia, with prominent expression maintained in GC and aberrant expression seen in other malignancies (62). As such, it represents a potential target in the treatment of GC. A number of agents targeting claudin 18.2 are under development internationally however zolbetuximab from Astellas is currently at the forefront in terms of clinical development. It is a chimeric monoclonal immunoglobulin antibody that binds claudin 18.2 to bring about tumor cell death (63). Zolbetuximab monotherapy demonstrated only modest activity in advanced claudin 18.2 positive gastric/GEJ cancer in the second-line setting, with ORR of just 9% and marginally better ORR of 14% in those with moderate to high claudin 18.2 expression in ≥70% of tumor cells (64). Combination therapy looks to offer more efficacy. The FAST trial was a phase II randomized control trial assessing zolbetuximab with epirubicin, oxaliplatin, and capecitabine (EOX) vs. EOX alone in the first-line setting in claudin 18.2 positive advanced gastric, GEJ and esophageal adenocarcinoma. Inclusion criteria required moderate to strong claudin 18.2 expression in ≥40% of tumor cells and centrally assessed by immunohistochemistry. Of 730 patients that were screened, 686 had tumor samples amenable to central assessment, and of these 334 (49%) met the claudin 18.2 expression inclusion criteria. Median PFS was greater with zolbetuximab + EOX than EOX [7.5 (95% CI: 5.6–11.3) vs. 5.3 (95% CI: 4.1–7.1) months]. Zolbetuximab + EOX also demonstrated a greater median OS and reduction in risk of death, compared with EOX [13.0 (95% CI: 9.7–18.7) vs. 8.3 (95% CI: 6.9–10.2) months; HR: 0.55, 95% CI: 0.39–0.77, P<0.0005]. SPOTLIGHT and GLOW are two phase III randomized control trials currently in progress assessing mFOLFOX with zolbetuximab and CAPOX with zolbetuximab respectively (65,66). Data from the phase III Spotlight trial was presented at ASCO Gastrointestinal on Thursday Jan 19th 2023 by Shitara as late breaking abstract LBA292 (67). It assesses zolbetuximab plus mFOLFOX 6 vs. placebo plus mFOLFOX6 and reports statistically significant improvements in PFS and OS in the zolbetuximab arm. Median PFS with zolbetuximab + mFOLFOX6 was 10.61 vs. 8.67 months in the placebo arm (HR: 0.751, P=0.0066). Additionally, improved OS was also reported with zolbetuximab + mFOLFOX (median 18.23 vs. 15.54 months; HR: 0.750, P=0.0053). While the incidences of serious treatment related AEs were similar in both arms (44.8% vs. 43.5%), there were higher rates of nausea and vomiting in the zolbetuximab arm.

Finally, in the realms of non-immune approaches, brief consideration should be given to the notable advances over the past two decades in the prevention, early detection and enhanced surgical intervention in GC. As mentioned, considerable regional variations exist in the incidence of GC internationally, with considerably higher incidence in Asian populations than in the western world (68). Japan and South Korea demonstrate amongst the highest incidence internationally of GC and have both introduced population-based screening programmes, although the implementation of screening recommendations is noted to vary on a national level and, as with almost any new therapeutic or intervention, there are concerns about sustainability from a cost perspective (69). Significant advances have also been made, again particularly in the East Asian world, with novel and progressive surgical techniques. Minimally invasive interventions, such as endoscopic mucosal dissection (ESD) which can include the use of artificial intelligence (AI), and stomach sparing surgical techniques propose to offer improved quality of life outcomes for patients (70), At present, long-term survival data is outstanding, however such surgical advances may well play a role in the currently still evolving perioperative systemic therapy paradigm in GC.


Conclusions

There has been much progress in the management of GC over the past two decades. The role of chemotherapy has been refined and has steadily increased the proportion of patients cured by surgery. In the advanced disease setting, notwithstanding the disappointments of multiple targeted therapies, anti-HER2 treatments have been genuinely impactful and continue to evolve. More recently, we have seen the potential for immune-based approaches and already the standard of care has changed in the perioperative and advanced disease settings. This is not surprising given the heterogenous clinicopathologic subtypes and knowledge of their respective immunogenicity as evidenced by the prognostic relevance of finding tumor-reactive lymphocytes embedded in the tumor. Ongoing clinical trials seek to further these advances with many exciting strategies such as combining multikinase inhibitors like lenvatinib which can target multiple immune related pathways with PD-1 inhibitors and CAR-T therapy which, though in early stages of investigation, have demonstrated promising preliminary results. Other targeted therapies are showing promising early results, with agents under investigation targeting FGFR2b in addition to claudin 18.2. However, as treatments advance questions remain about their optimal use. Further clarity is needed regarding predictive biomarkers for ICIs, with limitations to using PD-L1 including its heterogeneity of expression, variation in assays and optimal cut-off levels. With the evolution of newer agents with alternative specific targets, similar questions may well develop with regard to FGFR2b and claudin 18.2 expression and assessment. Regardless, with continued advances in immune-based, immune-adjacent and non-immune therapeutic approaches in GC the future holds promise for better outcomes and improved survival.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editors (Adam Yopp and Matthew Porembka) for the series “Multimodal Management of Gastric and Gastroesophageal Cancers” published in Chinese Clinical Oncology. The article has undergone external peer review.

Peer Review File: Available at https://cco.amegroups.com/article/view/10.21037/cco-23-13/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-23-13/coif). The series “Multimodal Management of Gastric and Gastroesophageal Cancers” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  2. Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 2006;355:11-20. [Crossref] [PubMed]
  3. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687-97. [Crossref] [PubMed]
  4. Shitara K, Bang YJ, Iwasa S, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med 2020;382:2419-30. [Crossref] [PubMed]
  5. Wainberg ZA, Enzinger PC, Kang YK, et al. Bemarituzumab in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma (FIGHT): a randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol 2022;23:1430-40. [Crossref] [PubMed]
  6. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74. [Crossref] [PubMed]
  7. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol 2004;22:329-60. [Crossref] [PubMed]
  8. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513:202-9. [Crossref] [PubMed]
  9. Andre T, Tougeron D, Piessen G, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in patients (pts) with localized microsatellite instability-high (MSI)/mismatch repair deficient (dMMR) oeso-gastric adenocarcinoma (OGA): The GERCOR NEONIPIGA phase II study. J Clin Oncol 2022;40:abstr 244.
  10. 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;24:1449-58. [Crossref] [PubMed]
  11. Lee JS, Won HS, Sun S, et al. Prognostic role of tumor-infiltrating lymphocytes in gastric cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2018;97:e11769. [Crossref] [PubMed]
  12. Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 2007;13:1050-9. [Crossref] [PubMed]
  13. Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021;398:759-71. [Crossref] [PubMed]
  14. Metges JP, Kato K, Sun JM, et al. First-line pembrolizumab plus chemotherapy versus chemotherapy in advanced esophageal cancer: Longer-term efficacy, safety, and quality-of-life results from the phase 3 KEYNOTE-590 study. J Clin Oncol 2022;40:abstr 241.
  15. Rha SY, Wyrwicz LS, Weber PEY, et al. VP1-2023: Pembrolizumab (pembro) plus chemotherapy (chemo) as first-line therapy for advanced HER2-negative gastric or gastroesophageal junction (G/GEJ) cancer: Phase III KEYNOTE-859 study. Ann Oncol 2023;34:319-20. [Crossref]
  16. 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. [Crossref] [PubMed]
  17. Boku N, Ryu MH, Kato K, et al. Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: interim results of a randomized, phase II trial (ATTRACTION-4). Ann Oncol 2019;30:250-8. [Crossref] [PubMed]
  18. Janjigian YY, Ajani JA, Moehler M, et al. LBA7 Nivolumab (NIVO) plus chemotherapy (Chemo) or ipilimumab (IPI) vs chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer/esophageal adenocarcinoma (GC/GEJC/EAC): CheckMate 649 study. Ann Oncol 2021;32:S1329-30. [Crossref]
  19. Kang YK, Chen LT, Ryu MH, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:234-47. [Crossref] [PubMed]
  20. Xu J, Jiang H, Pan Y, et al. LBA53 Sintilimab plus chemotherapy (chemo) versus chemo as first-line treatment for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma (ORIENT-16): First results of a randomized, double-blind, phase III study. Ann Oncol 2021;32:S1331. [Crossref]
  21. Moehler MH, Kato K, Arkenau HT, et al. Rationale 305: Phase 3 study of tislelizumab plus chemotherapy vs placebo plus chemotherapy as first-line treatment (1L) of advanced gastric or gastroesophageal junction adenocarcinoma (GC/GEJC). J Clin Oncol 2023;41:abstr 286.
  22. Muro K, Chung HC, Shankaran V, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol 2016;17:717-26. [Crossref] [PubMed]
  23. 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. [Crossref] [PubMed]
  24. 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;6:1571-80. [Crossref] [PubMed]
  25. Shitara K, Özgüroğlu 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;392:123-33. [Crossref] [PubMed]
  26. 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;390:2461-71. [Crossref] [PubMed]
  27. Kim JH, Ryu MH, Park YS, et al. Predictive biomarkers for the efficacy of nivolumab as ≥ 3rd-line therapy in patients with advanced gastric cancer: a subset analysis of ATTRACTION-2 phase III trial. BMC Cancer 2022;22:378. [Crossref] [PubMed]
  28. Boku N, Satoh T, Ryu MH, et al. Nivolumab in previously treated advanced gastric cancer (ATTRACTION-2): 3-year update and outcome of treatment beyond progression with nivolumab. Gastric Cancer 2021;24:946-58. [Crossref] [PubMed]
  29. Curran MA, Montalvo W, Yagita H, et al. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A 2010;107:4275-80. [Crossref] [PubMed]
  30. Selby MJ, Engelhardt JJ, Johnston RJ, et al. Preclinical Development of Ipilimumab and Nivolumab Combination Immunotherapy: Mouse Tumor Models, In Vitro Functional Studies, and Cynomolgus Macaque Toxicology. PLoS One 2016;11:e0161779. [Crossref] [PubMed]
  31. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015;373:23-34. [Crossref] [PubMed]
  32. Escudier B, Motzer RJ, Tannir NM, et al. Efficacy of Nivolumab plus Ipilimumab According to Number of IMDC Risk Factors in CheckMate 214. Eur Urol 2020;77:449-53. [Crossref] [PubMed]
  33. 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;36:2836-44. [Crossref] [PubMed]
  34. Pietrantonio F, Randon G, Di Bartolomeo M, et al. Predictive role of microsatellite instability for PD-1 blockade in patients with advanced gastric cancer: a meta-analysis of randomized clinical trials. ESMO Open 2021;6:100036. [Crossref] [PubMed]
  35. Kelly RJ, Lee J, Bang YJ, et al. Safety and Efficacy of Durvalumab and Tremelimumab Alone or in Combination in Patients with Advanced Gastric and Gastroesophageal Junction Adenocarcinoma. Clin Cancer Res 2020;26:846-54. [Crossref] [PubMed]
  36. 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;39:966-77. [Crossref] [PubMed]
  37. Lee WS, Yang H, Chon HJ, et al. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp Mol Med 2020;52:1475-85. [Crossref] [PubMed]
  38. Finn RS, Ikeda M, Zhu AX, et al. Phase Ib Study of Lenvatinib Plus Pembrolizumab in Patients With Unresectable Hepatocellular Carcinoma. J Clin Oncol 2020;38:2960-70. [Crossref] [PubMed]
  39. Makker V, Taylor MH, Aghajanian C, et al. Lenvatinib Plus Pembrolizumab in Patients With Advanced Endometrial Cancer. J Clin Oncol 2020;38:2981-92. [Crossref] [PubMed]
  40. Tabernero J, Cohen D, Van Cutsem E, et al. P-154 A randomized phase 3 study evaluating the efficacy and safety of first-line pembrolizumab plus lenvatinib plus chemotherapy compared with chemotherapy in patients with advanced/metastatic gastroesophageal adenocarcinoma: LEAP-015. Ann Oncol 2021;32:S151-2. [Crossref]
  41. Yanez PE, Ben-Aharon I, Rojas C, et al. First-line lenvatinib plus pembrolizumab plus chemotherapy versus chemotherapy in advanced/metastatic gastroesophageal adenocarcinoma (LEAP-015): Safety run-in results. J Clin Oncol 2023;41:abstr 411.
  42. Solomon BL, Garrido-Laguna I. TIGIT: a novel immunotherapy target moving from bench to bedside. Cancer Immunol Immunother 2018;67:1659-67. [Crossref] [PubMed]
  43. Banta KL, Xu X, Chitre AS, et al. Mechanistic convergence of the TIGIT and PD-1 inhibitory pathways necessitates co-blockade to optimize anti-tumor CD8(+) T cell responses. Immunity 2022;55:512-26.e9. [Crossref] [PubMed]
  44. Cho BC, Rodriguez-Abreu D, Hussein M, et al. LBA2 Updated analysis and patient-reported outcomes (PROs) from CITYSCAPE: A randomised, double-blind, phase II study of the anti-TIGIT antibody tiragolumab+ atezolizumab (TA) versus placebo+ atezolizumab (PA) as first-line treatment for PD-L1+ NSCLC. Ann Oncol 2021;32:S1428. [Crossref]
  45. A Study to Evaluate the Safety and Pharmacokinetics of the Intravenous Fixed-Dose Combination (IV FDC) of Tiragolumab and Atezolizumab in Participants With Locally Advanced, Recurrent or Metastatic Solid Tumors (SKYSCRAPER-11). Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05661578
  46. A Study to Explore the Efficacy and Safety of Atezolizumab Plus Tiragolumab and Chemotherapy in 1st Line HER2 Negative Unresectable, Recurrent or Metastatic Gastric Cancer or Adenocarcinoma of Gastroesophageal Junction (GEJ). Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04933227
  47. Novel Combinations in Participants With Locally Advanced Unresectable or Metastatic Gastric or Gastroesophageal Junction Adenocarcinoma. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05702229
  48. Feeney K, Kelly R, Lipton LR, et al. CA224-060: A randomized, open label, phase II trial of relatlimab (anti-LAG-3) and nivolumab with chemotherapy versus nivolumab with chemotherapy as first-line treatment in patients with gastric or gastroesophageal junction adenocarcinoma. J Clin Oncol 2019;37:TPS4143. [Crossref]
  49. Bębnowska D, Grywalska E, Niedźwiedzka-Rystwej P, et al. CAR-T Cell Therapy-An Overview of Targets in Gastric Cancer. J Clin Med 2020;9:1894. [Crossref] [PubMed]
  50. Qi C, Gong J, Li J, et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med 2022;28:1189-98. [Crossref] [PubMed]
  51. Forde PM, Chaft JE, Smith KN, et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med 2018;378:1976-86. [Crossref] [PubMed]
  52. Krishnamoorthy M, Lenehan JG, Maleki Vareki S. Neoadjuvant Immunotherapy for High-Risk, Resectable Malignancies: Scientific Rationale and Clinical Challenges. J Natl Cancer Inst 2021;113:823-32. [Crossref] [PubMed]
  53. Kelly RJ, Ajani JA, Kuzdzal J, et al. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N Engl J Med 2021;384:1191-203. [Crossref] [PubMed]
  54. Al-Batran SE, Lorenzen S, Thuss-Patience PC, et al. Surgical and pathological outcome, and pathological regression, in patients receiving perioperative atezolizumab in combination with FLOT chemotherapy versus FLOT alone for resectable esophagogastric adenocarcinoma: Interim results from DANTE, a randomized, multicenter, phase IIb trial of the FLOT-AIO German Gastric Cancer Group and Swiss SAKK. J Clin Oncol 2022;40:abstr 4003.
  55. Smyth E, Knödler M, Giraut A, et al. VESTIGE: Adjuvant Immunotherapy in Patients With Resected Esophageal, Gastroesophageal Junction and Gastric Cancer Following Preoperative Chemotherapy With High Risk for Recurrence (N+ and/or R1): An Open Label Randomized Controlled Phase-2-Study. Front Oncol 2020;9:1320. [Crossref] [PubMed]
  56. Bang YJ, Van Cutsem E, Fuchs CS, et al. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol 2019;15:943-52. [Crossref] [PubMed]
  57. Janjigian YY, Van Cutsem E, Muro K, et al. MATTERHORN: phase III study of durvalumab plus FLOT chemotherapy in resectable gastric/gastroesophageal junction cancer. Future Oncol 2022;18:2465-73. [Crossref] [PubMed]
  58. André T, Tougeron D, Piessen G, et al. Neoadjuvant Nivolumab Plus Ipilimumab and Adjuvant Nivolumab in Localized Deficient Mismatch Repair/Microsatellite Instability-High Gastric or Esophagogastric Junction Adenocarcinoma: The GERCOR NEONIPIGA Phase II Study. J Clin Oncol 2023;41:255-65. [Crossref] [PubMed]
  59. Janjigian YY, Oh DY, Rha SY, et al. Dose-escalation and dose-expansion study of trastuzumab deruxtecan (T-DXd) monotherapy and combinations in patients (pts) with advanced/metastatic HER2+ gastric cancer (GC)/gastroesophageal junction adenocarcinoma (GEJA): DESTINY-Gastric03. J Clin Oncol 2022;40:abstr 295.
  60. Catenacci DVT, Rasco D, Lee J, et al. Phase I Escalation and Expansion Study of Bemarituzumab (FPA144) in Patients With Advanced Solid Tumors and FGFR2b-Selected Gastroesophageal Adenocarcinoma. J Clin Oncol 2020;38:2418-26. [Crossref] [PubMed]
  61. Hong JY, An JY, Lee J, et al. Claudin 18.2 expression in various tumor types and its role as a potential target in advanced gastric cancer. Transl Cancer Res 2020;9:3367-74. [Crossref] [PubMed]
  62. Ungureanu BS, Lungulescu CV, Pirici D, et al. Clinicopathologic Relevance of Claudin 18.2 Expression in Gastric Cancer: A Meta-Analysis. Front Oncol 2021;11:643872. [Crossref] [PubMed]
  63. Sahin U, Türeci Ö, Manikhas G, et al. FAST: a randomised phase II study of zolbetuximab (IMAB362) plus EOX versus EOX alone for first-line treatment of advanced CLDN18.2-positive gastric and gastro-oesophageal adenocarcinoma. Ann Oncol 2021;32:609-19. [Crossref] [PubMed]
  64. Türeci O, Sahin U, Schulze-Bergkamen H, et al. A multicentre, phase IIa study of zolbetuximab as a single agent in patients with recurrent or refractory advanced adenocarcinoma of the stomach or lower oesophagus: the MONO study. Ann Oncol 2019;30:1487-95. [Crossref] [PubMed]
  65. Shitara K, Al-Batran SE, Bang YJ, et al. 198TiP spotlight: phase III study of zolbetuximab+ mFOLFOX6 versus placebo+ mFOLFOX6 in first-line Claudin18. 2+/HER2 advanced or metastatic gastric or gastroesophageal junction adenocarcinoma (G/GEJ). Ann Oncol 2020;31:S1317. [Crossref]
  66. Xu RH, Ajani JA, Al-Batran SE, et al. 195TiP GLOW: Phase III study of first-line zolbetuximab+ CAPOX versus placebo+ CAPOX in Claudin18. 2+/HER2 advanced/metastatic gastric or gastroesophageal junction adenocarcinoma (G/GEJ). Ann Oncol 2020;31:S1315-6. [Crossref]
  67. Shitara K, Lordick F, Bang YJ, et al. Zolbetuximab+ mFOLFOX6 as first-line (1L) treatment for patients (pts) withclaudin-18.2+(CLDN18. 2+)/HER2− locally advanced (LA) unresectable or metastatic gastric or gastroesophageal junction (mG/GEJ) adenocarcinoma: primary results from phase 3 SPOTLIGHT study. J Clin Oncol 2023;41:LBA292. [Crossref]
  68. Morgan E, Arnold M, Camargo MC, et al. The current and future incidence and mortality of gastric cancer in 185 countries, 2020-40: A population-based modelling study. EClinicalMedicine 2022;47:101404. [Crossref] [PubMed]
  69. Mabe K, Inoue K, Kamada T, et al. Endoscopic screening for gastric cancer in Japan: Current status and future perspectives. Dig Endosc 2022;34:412-9. [Crossref] [PubMed]
  70. Li GZ, Doherty GM, Wang J. Surgical Management of Gastric Cancer: A Review. JAMA Surg 2022;157:446-54. [Crossref] [PubMed]
Cite this article as: Lynch E, Duffy AG, Kelly RJ. Brave new world—new and emerging treatments for gastric cancer. Chin Clin Oncol 2023;12(5):54. doi: 10.21037/cco-23-13

Download Citation