Skip to main content
Erschienen in: memo - Magazine of European Medical Oncology 1/2022

Open Access 19.10.2021 | short review

Innovative strategies in metastatic gastric cancer: a short review

verfasst von: Hannah Christina Puhr, MD, Aysegul Ilhan-Mutlu, MD PhD

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 1/2022

Summary

Recent innovative advances, especially concerning immunotherapeutic agents and targeted therapies, have changed the face of modern oncology. The year 2020 represents a milestone in the treatment of gastroesophageal cancer because several trials showed promising survival benefits, at least for a specific subgroup of patients. Not only immunotherapeutic agents, but also targeted therapies seem to be beneficial, particularly when the target is well defined and the threshold value is selected appropriately. Thus, many new innovative treatment strategies are underway and might lead to a further paradigm change in the therapy of patients with advanced gastric tumors. This review gives a concise overview of these new therapeutic options and recently approved strategies as well as ongoing studies.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CI
Confidence interval
CPS
Combined Positive Score
DoR
Duration of response
EMA
European Medicines Agency
FDA
Food and Drug Administration
FGFR2b
Fibroblast growth factor receptor 2b
GEJ
Gastroesophageal junction
HER2
Human epidermal growth factor receptor 2
HR
Hazard ratio
MSI‑H
High microsatellite instability
n.r.
Not reached
ORR
Overall response rate
OS
Overall survival
PD-(L)1
Programmed cell death receptor (ligand) 1
PFS
Progression-free survival
TMB‑H
High tumor mutational burden

Introduction

Gastric cancer is a major contributor to global disease burden and, thus, new treatment options are desperately needed to improve the outcome of patients suffering from this devastating disease [1]. Although advances in immunotherapy as well as targeted therapy at the beginning of the 21st century have led to major breakthroughs in various types of cancer, these high expectations could only recently be met in trials concerning gastric cancer [2]. Thus, new innovative targets and drugs are underway to improve the care of patients with gastric cancer. The aim of this mini-review is to concisely highlight some promising new treatment approaches of metastatic gastric cancer and their clinical relevance. Trials, which showed clinical and statistical relevant data leading to approval of new therapeutic strategies by appropriate authorities for advanced gastric cancer patients in recent years, are listed in Table 1.
Table 1
Innovative therapeutic strategies for advanced gastric cancer, which are already approved by authorities
Study name
Therapeutic agent
Phase
Histology
Therapy line
Control group
Findings
Population
Approval
Reference number
ATTRACTION‑2
Nivolumab
III
Advanced gastric or GEJ cancer
≥ 3
Placebo
Median OS: 5.26 vs 4.14 months (HR 0.63, 95% CI 0.51–0.78); p < 0.0001
Asia
2017–2019 (PD-L1 independent): Japan, South Korea, Taiwan, Singapore and Switzerland
[4, 6]
KEYNOTE-059
Pembrolizumab
II
Advanced gastric and GEJ cancer
≥ 2
No control group
ORR: 15.5%; 95% CI 10.1–22.4%
North and South America, Asia, Europe
2017 (CPS ≥ 1): FDA → 07/2021 withdrawal by company
[5, 7]
CheckMate 649
Nivolumab + chemotherapy;
(ipilimumab + nivolumab)
III
Advanced non-HER2-positive gastric, GEJ or esophageal adenocarcinoma
1
Chemotherapy
Median OS: nivo + CHT 14.4 (95% CI 13.1–16.2) vs CHT 11.1 months (95% CI 10.0–12.1) in PD-L1 CPS ≥ 5 pts
North and South America, Australia, Asia, Europe
2021 (PD-L1 independent): FDA
[8, 9]
5 trials (KEYNOTE: 016, 164, 012, 028, and 158)
Pembrolizumab
Ib, II
149 patients with MSI‑H cancers
≥ 2
No control group
ORR (retrospective analysis): 39.6% (95% CI: 31.7, 47.9)
North and South America, Australia, Asia, Europe
2017 (MSI‑H; without satisfactory alternative treatment options): FDA
[11]
KEYNOTE-158
Pembrolizumab
II
MSI‑H advanced non-colorectal cancer
≥ 2
No control group
TMB‑H (retrospective analysis): ORR: 29% (95% CI 21–39); DoR: not reached
North and South America, Australia, Asia, Europe
2020 (TMB‑H; without satisfactory alternative treatment options): FDA
[12]
KEYNOTE-811
Pembrolizumab + trastuzumab and chemotherapy
III
HER2-positive metastatic gastric/GEJ cancer
1
Placebo + trastuzumab and chemotherapy
ORR: pembro 74% (95% CI 66–82) vs placebo 52% (95% CI 43–61) arm; one-sided p-value < 0.0001
North and South America, Australia, Asia, Europe
2021 (HER2-positive, PD-L1 independent): FDA
[24, 25]
DESTINY-Gastric01
Trastuzumab deruxtecan
II
HER2-positive advanced gastric/GEJ cancer
≥ 3
Chemotherapy
Median OS: 12.5 (95% CI 9.6–14.3) vs 8.4 months (95% CI 6.9–10.7); HR 0.59 (95% CI 0.39–0.88); p = 0.0097
Asia
2021 (HER2-positive; prior trastuzumab-based regimen): FDA, Japan
[26, 27]
FIGHT
Bemarituzumab + chemotherapy
II
FGFR2b-positive, HER2-negative advanced gastric/GEJ cancer
1
Chemotherapy + placebo
Median OS: 25.4 months (95% CI: 13.8, n.r.) vs 11.1 months (95% CI: 8.4, 13.8) for pts with ≥ 10% FGFR2b+; HR 0.41 (95% CI 0.23–0.74)
North America, Australia, Asia, Europe
2021 (FGFR2b-positive, HER2-negative): FDA
[29, 30]
CI confidence interval, CPS Combined Positive Score, DoR Duration of response, EMA European Medicines Agency, FDA Food and Drug Administration, FGFR2b fibroblast growth factor receptor 2b, GEJ gastroesophageal junction, HER2 human epidermal growth factor receptor 2, HR hazard ratio, MSI‑H high microsatellite instability, n.r. not reached, ORR overall response rate, OS overall survival, PD-(L)1 programmed cell death receptor (ligand) 1, PFS progression-free survival, TMB‑H high tumor mutational burden

Immunotherapy for gastric cancer

Current treatment landscape with immunotherapeutic agents

So far, the most promising immunotherapeutic target in gastric cancer patients was the programmed cell death receptor 1 (PD-1)/programmed cell death ligand 1 (PD-L1) checkpoint axis [3]. Although some studies showed a significant benefit for all patients enrolled [4], most trials indicate that specific patient subgroups (such as high PD-L1 expression or high microsatellite instability [MSI-H]) profit most from immune checkpoint inhibition [5]. Thus, the evaluation of PD-L1 expression using Combined Positive Score (CPS) became part of the routine diagnostic work-up of tumor tissue. Yet, the cut-off levels for therapeutic approvals are still widely discussed. The results of the ATTACTION-II trial were published independent of PD-L1 expression and led to an approval of nivolumab independent of CPS expression [6].
However, in the phase II KEYNOTE-059 trial the cut-off level for PD-L1-positivity was defined as CPS ≥ 1 (median response duration was 16.3 [1.6+ to 17.3+] months in patients with PD-L1-positive and 6.9 [2.4 to 7.0+] months and PD-L1-negative tumors) and, thus, pembrolizumab was approved by the Food and Drug Administration (FDA) as a third and further line therapy in CPS ≥ 1 patients with metastatic gastroesophageal adenocarcinoma [5, 7]. However, by July 2021, the company voluntarily withdrew this accelerated approval indication.
However, the CheckMate 649 trial found that patients with CPS ≥ 5 showed the most significant benefit (overall survival hazard ratio [OS HR] 0.71, 98.4% CI 0.59–0.86; p < 0.0001) when adding nivolumab to standard chemotherapy. Interestingly, this combination was approved by the FDA in 2021 regardless of PD-L1 expression status and is currently under investigation by the European Medicines Agency (EMA) [8, 9].
Other important and already established biomarkers that indicate response to checkpoint inhibition are MSI, which is surmised to be high (MSI-H) in around 4–5% of all advanced Western gastric tumor cases, and tumor mutational burden (TMB) [10]. Thus, the FDA approved immunotherapy with pembrolizumab for the treatment of unresectable or metastatic MSI‑H and TMB‑H solid tumors that have progressed following prior treatment independent of tumor location and which have no satisfactory alternative treatment options (“tissue agnostic approvals”) [11, 12].
These findings underline the importance of patient selection and changed the face of gastric cancer treatment.

Strategies to overcome the immune cold tumors

As mentioned above, a major issue concerning immunotherapy is that only a subset of patients achieve responses. Thus, the identification of underlying mechanisms for primary resistance to immunotherapy are of major concern [13]. Recent studies characterized these immunologically “cold” tumors by a lack of infiltrating T cells in the tumor microenvironment. Consequentially, tumor cells stay unrecognized by the immune system and, thus, do not respond to checkpoint inhibition.
However, it is surmised that these “cold” tumors can be transformed into “hot” and inflamed tumors by several strategies including neutralizing immunosuppression at the tumor site by combining immunotherapeutic approaches, modifying the tumor vasculature by targeting endothelial growth, targeting the tumor cells themselves with chemotherapy, inducing local inflammation with radiation therapy, or increasing the frequency of tumor-specific T cells with personalized approaches such as CAR T cell therapy [13].

Combination with targeted therapies

Especially, the combination with targeted therapy for angiogenesis and growth pathways has gained importance in recent years.
The combination of nivolumab and regorafenib is currently under investigation. The recently published phase Ib REGONIVO trial found encouraging antitumor activity in patients with gastric cancer in a third and further line setting (median PFS 5.6 months), thus, warranting additional investigations in larger cohorts [14]. Furthermore, ramucirumab was investigated in combination with paclitaxel plus nivolumab and showed promising anti-tumor activity (median OS 13.8 months [95% CI 8.0–19.5 months] in CPS ≥ 1 patients) [15]. Another approach evaluated the combination treatment of lenvatinib, a multikinase inhibitor of VEGF receptors and other receptor tyrosine kinases, with pembrolizumab and a response rate of 69% (95% CI 49–85) was demonstrated [16].
There are several ongoing trials evaluating the combination of the checkpoint inhibitor durvalumab with targeted therapies, e.g., with cabozantinib in CAMILLA trial and with raumucirumab in a phase Ib trial [17, 18].

Combination of anti-PD-1/PD-L1 drugs with other checkpoint inhibitors

Although the concept of neutralizing immunosuppression at the tumor site by combining immunotherapeutic approaches, the combination of anti-PD-1/PD-L1 drugs with other checkpoint inhibitors, so far no practice changing trials can be reported in gastric cancer. However, several studies suggest promising new combination strategies including combination with cytotoxic T lymphocyte antigen‑4 (CTLA‑4; drug: ipilimumab; trials: CheckMate-032 [19], Moonlight trial [20]), lymphocyte activation gene‑3 (LAG3; drug: relatlimab; trials: FRACTION [21], REACTION), T cell immunoglobulin and mucin-domain containing‑3 (TIM-3), T cell immunoglobulin and ITIM domain (TIGIT), V‑domain Ig suppressor of T cell activation (VISTA), OX40 (CD134) and B and T cell lymphocyte attenuator (BTLA) [22, 23].

Targeted therapy for gastroesophageal tumors

Targeting HER2 beyond trastuzumab:

In the field of HER2-positive (human epidermal growth factor receptor 2) gastroesophageal cancer, a recently published phase II trial evaluating first-line pembrolizumab and trastuzumab in combination with chemotherapy showed promising response rates and, thus, was the impulse for a randomized phase 3 clinical trial (KEYNOTE-811) [24]. First results of the KEYNOTE-811 trial found that the overall response rate (ORR) was 74% (95% CI 66–82) in the pembrolizumab arm and 52% (95% CI 43–61) in the placebo arm (one-sided p-value < 0.0001) and, thus, the combination of pembrolizumab, trastuzumab and chemotherapy (ToGA regimen) has been recently approved by the FDA for the first-line treatment of HER2-positive advanced gastric cancer [25].
Another new approach that was recently approved by the FDA for HER2-positive patients who have received a prior trastuzumab-based regimen is the drug trastuzumab deruxtecan, a novel antibody-drug conjugate [26]. The drug was evaluated by the phase II DESTINY-Gastric01 trial in a third and further line setting in comparison to chemotherapy and showed a significantly longer median OS (12.5 vs. 8.4 months; HR 0.59; 95% CI 0.39–0.88; p = 0.01) [27].

Attempts for novel targets

Claudin 18.2 (CLDN18.2) protein is physiologically confined to gastric mucosa tight junctions and is exposed on the cancer cell surface upon malignant transformation.
In the phase II FAST trial, patients with advanced gastric cancer and a CLDN18.2 expression in ≥ 70% of tumor cells were treated with the anti-Claudin 18.2 antibody zolbetuximab in combination with chemotherapy. Zolbetuximab generated prolonged OS rates in this patient subgroup (HR 0.55; 95% CI 0.39–0.77; p < 0.0005) [28]. Based on this data, two phase III trials have been initiated, which will reveal first results within the next few years.
Fibroblast growth factor receptor 2b (FGFR2b) is another very interesting target, which was tested in the FIGHT study. The anti-FGFR2b antibody bemarituzumab in combination with chemotherapy as first-line revealed very promising results in FGFR2b-positive patients, which led to the designation of bemarituzumab as a breakthrough therapy by the FDA (OS HR 0.66; 95% CI 0.39–1.12) [29, 30].

Discussion

Despite the improvement of modern cancer medicine including immunotherapies and targeted therapies, new therapeutic approaches seem to be efficiently in only specific subgroups of patients. To improve patient outcome with these new treatment options, it is of highest importance to define these subgroups more accurately. The evaluation and implementation of new biomarkers seems to be the key for adequate patient selection leading to high treatment efficacy. In case of immunotherapy, this selection is mainly based on PD-L1 expression, MSI status and, more recently, on TMB. However, looking at response rates it is evident that despite these biomarkers there is still significant percentage of patients who do not respond to treatment. Thus, underlining the fact that further strategies should be implemented to develop predictive markers.
Another critical issue is to overcome so called “cold” tumors, thereby improving treatment response. New combination strategies including combinations of the inhibition of the PD-(L)1 axis with chemotherapy, radiation therapy, other immune checkpoint inhibitors and targeted therapies are underway into clinical practice to overcome this treatment resistance.
Finally, critical issues to consider for new drug approvals are tolerability, impact on quality of life, and financial considerations. Future studies need to include these major considerations in clinical trial design in order to achieve more adequate implementation of new therapeutic agents into real-life cohorts and guidelines.
In conclusion, recently published studies led to a paradigm change of advanced gastric cancer treatment and several new innovative approaches are underway to further improve the management of patient subgroups.
Take home message
New innovative targets and drugs are underway to improve the care of patients with advanced gastric cancer. This mini-review highlights promising new treatment approaches and their clinical relevance.

Funding

The authors gratefully acknowledge Medical University of Vienna’s core funding to the Department of Medicine I.

Conflict of interest

H.C. Puhr has received travel support from Eli Lilly, MSD, Novartis, Pfizer and Roche. A. Ilhan-Mutlu participated in advisory boards from MSD, BMS and Servier, received lecture honoraria from Eli Lilly, MSD, BMS and Servier, is the local PI for clinical trials sponsored by BMS and Roche and received travel support from BMS, Roche, Eli Lilly and Daiichi Sankyo.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144(8):1941–53.CrossRef Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144(8):1941–53.CrossRef
2.
Zurück zum Zitat Vrána D, Matzenauer M, Neoral Č, Aujeský R, Vrba R, Melichar B, et al. From tumor immunology to immunotherapy in gastric and esophageal cancer. Int J Mol Sci. 2018;20(1):13.CrossRef Vrána D, Matzenauer M, Neoral Č, Aujeský R, Vrba R, Melichar B, et al. From tumor immunology to immunotherapy in gastric and esophageal cancer. Int J Mol Sci. 2018;20(1):13.CrossRef
3.
Zurück zum Zitat Lordick F, Shitara K, Janjigian YY. New agents on the horizon in gastric cancer. Ann Oncol. 2017;28(8):1767–75.CrossRef Lordick F, Shitara K, Janjigian YY. New agents on the horizon in gastric cancer. Ann Oncol. 2017;28(8):1767–75.CrossRef
4.
Zurück zum Zitat Kang Y‑K, Boku N, Satoh T, Ryu M‑H, Chao Y, Kato K, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71.CrossRef Kang Y‑K, Boku N, Satoh T, Ryu M‑H, Chao Y, Kato K, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71.CrossRef
5.
Zurück zum Zitat Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, Machado M, et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE-059 trial. JAMA Oncol. 2018;4(5):e180013.CrossRef Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, Machado M, et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE-059 trial. JAMA Oncol. 2018;4(5):e180013.CrossRef
6.
Zurück zum Zitat Chen L‑T, Satoh T, Ryu M‑H, Chao Y, Kato K, Chung HC, et al. A phase 3 study of nivolumab in previously treated advanced gastric or gastroesophageal junction cancer (ATTRACTION-2): 2‑year update data. Gastric Cancer. 2020;23(3):510–9.CrossRef Chen L‑T, Satoh T, Ryu M‑H, Chao Y, Kato K, Chung HC, et al. A phase 3 study of nivolumab in previously treated advanced gastric or gastroesophageal junction cancer (ATTRACTION-2): 2‑year update data. Gastric Cancer. 2020;23(3):510–9.CrossRef
8.
Zurück zum Zitat Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, 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(10294):27–40.CrossRef Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, 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(10294):27–40.CrossRef
10.
Zurück zum Zitat Polom K, Marano L, Marrelli D, De Luca R, Roviello G, Savelli V, et al. Meta-analysis of microsatellite instability in relation to clinicopathological characteristics and overall survival in gastric cancer. Br J Surg. 2018;105(3):159–67.CrossRef Polom K, Marano L, Marrelli D, De Luca R, Roviello G, Savelli V, et al. Meta-analysis of microsatellite instability in relation to clinicopathological characteristics and overall survival in gastric cancer. Br J Surg. 2018;105(3):159–67.CrossRef
13.
Zurück zum Zitat Ochoa de Olza M, Navarro Rodrigo B, Zimmermann S, Coukos G. Turning up the heat on non-immunoreactive tumours: opportunities for clinical development. Lancet Oncol. 2020;21(9):e419–e30.CrossRef Ochoa de Olza M, Navarro Rodrigo B, Zimmermann S, Coukos G. Turning up the heat on non-immunoreactive tumours: opportunities for clinical development. Lancet Oncol. 2020;21(9):e419–e30.CrossRef
14.
Zurück zum Zitat Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, et al. Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 2020;38(18):2053–61.CrossRef Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, et al. Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 2020;38(18):2053–61.CrossRef
15.
Zurück zum Zitat Hironaka S, Kadowaki S, Izawa N, Nishina T, Yamanaka T, Minashi K, et al. A phase I/II study of nivolumab, paclitaxel, and ramucirumab as second-line in advanced gastric cancer. J Clin Oncol. 2020;38(4_suppl):352.CrossRef Hironaka S, Kadowaki S, Izawa N, Nishina T, Yamanaka T, Minashi K, et al. A phase I/II study of nivolumab, paclitaxel, and ramucirumab as second-line in advanced gastric cancer. J Clin Oncol. 2020;38(4_suppl):352.CrossRef
16.
Zurück zum Zitat Kawazoe A, Fukuoka S, Nakamura Y, Kuboki Y, Wakabayashi M, Nomura S, et al. Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(8):1057–65.CrossRef Kawazoe A, Fukuoka S, Nakamura Y, Kuboki Y, Wakabayashi M, Nomura S, et al. Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(8):1057–65.CrossRef
17.
Zurück zum Zitat Saeed A, Koestler D, Williamson SK, Baranda JC, Sun W, Al-Rajabi RMT, et al. A phase Ib trial of cabozantinib in combination with durvalumab (MEDI4736) in previously treated patients with advanced gastroesophageal cancer and other gastrointestinal (GI) malignancies (CAMILLA). J Clin Oncol. 2019;37(8_suppl):TPS56.CrossRef Saeed A, Koestler D, Williamson SK, Baranda JC, Sun W, Al-Rajabi RMT, et al. A phase Ib trial of cabozantinib in combination with durvalumab (MEDI4736) in previously treated patients with advanced gastroesophageal cancer and other gastrointestinal (GI) malignancies (CAMILLA). J Clin Oncol. 2019;37(8_suppl):TPS56.CrossRef
18.
Zurück zum Zitat Bang Y‑J, Golan T, Lin C‑C, Dahan L, Fu S, Moreno V, et al. Ramucirumab (Ram) and durvalumab (Durva) treatment of metastatic non-small cell lung cancer (NSCLC), gastric/gastroesophageal junction (G/GEJ) adenocarcinoma, and hepatocellular carcinoma (HCC) following progression on systemic treatment(s). J Clin Oncol. 2019;37(15_suppl):2528.CrossRef Bang Y‑J, Golan T, Lin C‑C, Dahan L, Fu S, Moreno V, et al. Ramucirumab (Ram) and durvalumab (Durva) treatment of metastatic non-small cell lung cancer (NSCLC), gastric/gastroesophageal junction (G/GEJ) adenocarcinoma, and hepatocellular carcinoma (HCC) following progression on systemic treatment(s). J Clin Oncol. 2019;37(15_suppl):2528.CrossRef
19.
Zurück zum Zitat Janjigian YY, Bendell J, Calvo E, Kim JW, Ascierto PA, Sharma P, 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(28):2836–44.CrossRef Janjigian YY, Bendell J, Calvo E, Kim JW, Ascierto PA, Sharma P, 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(28):2836–44.CrossRef
20.
Zurück zum Zitat Al-Batran S‑E, Pauligk C, Goetze TO, Riera-Knorrenschild J, Goekkurt E, Angermeier S, et al. Modified FOLFOX versus modified FOLFOX plus nivolumab and ipilimumab in patients with previously untreated advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction: Moonlight, a randomized phase 2 trial of the German Gastric Group of the AIO. J Clin Oncol. 2019;37(15_suppl):TPS4144.CrossRef Al-Batran S‑E, Pauligk C, Goetze TO, Riera-Knorrenschild J, Goekkurt E, Angermeier S, et al. Modified FOLFOX versus modified FOLFOX plus nivolumab and ipilimumab in patients with previously untreated advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction: Moonlight, a randomized phase 2 trial of the German Gastric Group of the AIO. J Clin Oncol. 2019;37(15_suppl):TPS4144.CrossRef
21.
Zurück zum Zitat Aanur P, Gutierrez M, Kelly RJ, Ajani JA, Ku GY, Denlinger CS, et al. FRACTION (Fast Real-time Assessment of Combination Therapies in Immuno-Oncology)-gastric cancer (GC): a randomized, open-label, adaptive, phase 2 study of nivolumab in combination with other immuno-oncology (IO) agents in patients with advanced GC. J Clin Oncol. 2017;35(15_suppl):TPS4137.CrossRef Aanur P, Gutierrez M, Kelly RJ, Ajani JA, Ku GY, Denlinger CS, et al. FRACTION (Fast Real-time Assessment of Combination Therapies in Immuno-Oncology)-gastric cancer (GC): a randomized, open-label, adaptive, phase 2 study of nivolumab in combination with other immuno-oncology (IO) agents in patients with advanced GC. J Clin Oncol. 2017;35(15_suppl):TPS4137.CrossRef
22.
Zurück zum Zitat Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. Novel immune checkpoint targets: moving beyond PD‑1 and CTLA‑4. Mol Cancer. 2019;18(1):155.CrossRef Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. Novel immune checkpoint targets: moving beyond PD‑1 and CTLA‑4. Mol Cancer. 2019;18(1):155.CrossRef
23.
Zurück zum Zitat Alves Costa SC, Facchinetti F, Routy B, Derosa L. New pathways in immune stimulation: targeting OX40. ESMO Open. 2020;5(1):e573.CrossRef Alves Costa SC, Facchinetti F, Routy B, Derosa L. New pathways in immune stimulation: targeting OX40. ESMO Open. 2020;5(1):e573.CrossRef
24.
Zurück zum Zitat Janjigian YY, Maron SB, Chatila WK, Millang B, Chavan SS, Alterman C, et al. First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(6):821–31.CrossRef Janjigian YY, Maron SB, Chatila WK, Millang B, Chavan SS, Alterman C, et al. First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(6):821–31.CrossRef
27.
Zurück zum Zitat Shitara K, Bang Y‑J, Iwasa S, Sugimoto N, Ryu M‑H, Sakai D, et al. Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer. N Engl J Med. 2020;382(25):2419–30.CrossRef Shitara K, Bang Y‑J, Iwasa S, Sugimoto N, Ryu M‑H, Sakai D, et al. Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer. N Engl J Med. 2020;382(25):2419–30.CrossRef
28.
Zurück zum Zitat Sahin U, Türeci Ö, Manikhas G, Lordick F, Rusyn A, Vynnychenko I, 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(5):609–19.CrossRef Sahin U, Türeci Ö, Manikhas G, Lordick F, Rusyn A, Vynnychenko I, 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(5):609–19.CrossRef
29.
Zurück zum Zitat Catenacci DVT, Kang Y‑K, Saeed A, Yamaguchi K, Qin S, Lee K‑W, et al. FIGHT: a randomized, double-blind, placebo-controlled, phase II study of bemarituzumab (bema) combined with modified FOLFOX6 in 1L FGFR2b+ advanced gastric/gastroesophageal junction adenocarcinoma (GC). J Clin Oncol. 2021;39(15_suppl):4010.CrossRef Catenacci DVT, Kang Y‑K, Saeed A, Yamaguchi K, Qin S, Lee K‑W, et al. FIGHT: a randomized, double-blind, placebo-controlled, phase II study of bemarituzumab (bema) combined with modified FOLFOX6 in 1L FGFR2b+ advanced gastric/gastroesophageal junction adenocarcinoma (GC). J Clin Oncol. 2021;39(15_suppl):4010.CrossRef
Metadaten
Titel
Innovative strategies in metastatic gastric cancer: a short review
verfasst von
Hannah Christina Puhr, MD
Aysegul Ilhan-Mutlu, MD PhD
Publikationsdatum
19.10.2021
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 1/2022
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-021-00762-w

Weitere Artikel der Ausgabe 1/2022

memo - Magazine of European Medical Oncology 1/2022 Zur Ausgabe