Next Article in Journal
Sarcomas: New Biomarkers and Therapeutic Strategies
Next Article in Special Issue
Nrf2 Downregulation Contributes to Epithelial-to-Mesenchymal Transition in Helicobacter pylori-Infected Cells
Previous Article in Journal
Uncommon Variants of Mature T-Cell Lymphomas (MTCLs): Imaging and Histopathologic and Clinical Features with Updates from the Fourth Edition of the World Health Organization (WHO) Classification of Lymphoid Neoplasms
Previous Article in Special Issue
Beyond Chemotherapy, PD-1, and HER-2: Novel Targets for Gastric and Esophageal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Targets for Gastric Cancer Treatment and Future Perspectives from a Clinical and Translational Point of View

Department of Oncology, Gastroenterology, Hepatology, Pulmonology and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany
*
Authors to whom correspondence should be addressed.
Submission received: 8 September 2021 / Revised: 5 October 2021 / Accepted: 13 October 2021 / Published: 18 October 2021
(This article belongs to the Special Issue New Molecular Insights for GC Characterization and Treatment)

Abstract

:

Simple Summary

During the last several years, gastric cancer has been extensively studied on the molecular level and distinct molecular subtypes have been defined accordingly. Here, we review the current data on targeted treatment approaches for advanced or metastatic gastric cancer based on molecular alterations or immunological features, ranging from clinically approved treatment strategies to more innovative concepts which are currently in preclinical or clinical development. Information on the translational context and methodology is also provided, holding promise for the further improved development of personalized treatment strategies in the future.

Abstract

Gastric cancer is a leading cause of cancer death worldwide. Systemic treatment comprising chemotherapy and targeted therapy is the standard of care in advanced/metastatic gastric cancer. Comprehensive molecular characterization of gastric adenocarcinomas by the TCGA Consortium and ACRG has resulted in the definition of distinct molecular subtypes. These efforts have in parallel built a basis for the development of novel molecularly stratified treatment approaches. Based on this molecular characterization, an increasing number of specific genomic alterations can potentially serve as treatment targets. Consequently, the development of promising compounds is ongoing. In this review, key molecular alterations in gastric and gastroesophageal junction cancers will be addressed. Finally, the current status of the translation of targeted therapy towards clinical applications will be reviewed.

1. Introduction

Gastric cancer is one of the most common tumor entities worldwide with 1 million new cases per year and about 748,000 deaths per year [1]. Systemic treatment is recommended for patients with stage IV gastric or gastroesophageal junction (GEJ) adenocarcinomas. This is based on a broad body of evidence from clinical trials, demonstrating both prolonged survival and control of cancer-related symptoms, translating into improved quality of life [2]. Chemotherapy (i.e., mono- or polychemotherapy) represents an important backbone for systemic therapy. Trastuzumab, an anti-HER2 (human epidermal growth factor receptor 2) antibody, is used in combination with chemotherapy in the first-line treatment of HER2 positive gastric cancer [3]. Ramucirumab, an anti-VEGFR2 (vascular endothelial growth factor receptor 2) antibody, which targets angiogenesis, is approved for second-line treatment either as a monotherapy or in combination with Paclitaxel [4,5,6]. Recently, novel targets like programmed death 1 ligand-1/programmed cell death protein 1 (PD-L1/PD-1), fibroblast growth factor receptor 2 (FGFR2), and claudin 18.2 (CLDN18.2) are emerging. There is urgent need for advances, as prognosis with chemotherapy alone remains poor, with a median survival of about 12 months in metastatic disease.
Morphological and genetic heterogeneity are major hallmarks of gastric cancer and basic histological classifications like the Laurén classification (intestinal, diffuse and mixed subtype) do not allow precise patient stratification towards distinct treatment strategies. From this background, groups in Asia and the US have aimed to more comprehensively characterize the molecular features of gastric cancer. To this end, The Cancer Genome Atlas (TCGA) consortium has performed a comprehensive molecular analysis and classification of gastric adenocarcinomas from 295 patients based on next generation sequencing (NGS), whole-exomes DNA sequencing, mRNA, and miRNA sequencing. In addition, array-based analysis of DNA methylation and reverse-phase protein arrays have been performed. On this basis, four molecular subtypes have been defined: (i) EBV positive (EBV); (ii) microsatellite instable (MSI); (iii) genomically stable (GS) and iv) chromosomal instable (CIN) [7]. In addition, the Asian Cancer Research Group (ACRG) followed a different approach based primarily on gene expression profiling in a cohort of 300 Asian gastric cancer patients to identify four molecular subtypes: (i) microsatellite instable; (ii) microsatellite stable (MSS) TP53 inactive; (iii) MSS TP53 active, and iv) MSS with a gene expression profile related to Epidermal to Mesenchymal Transition (EMT). Furthermore, targeted sequencing and genome-wide copy number microarrays were used to describe key molecular alterations in the four subtypes [8]. The overlap between these two major classifications and key clinical and molecular features are summarized in Figure 1.
Due to differences within patient cohorts studied by these two consortia and due to different techniques used to analyze and define molecular subtypes, overlap is only partial. Specifically, while the TCGA approach relied on several technical platforms, as indicated above, the key technology used by the ACRG was gene expression profiling. There were differences between the cohorts regarding ethnicity and stage distribution (i.e., stage IV disease in 15% of patients in the TCGA data set vs. 26% for the ACRG data set). On the other hand, both classifications were demonstrated to be prognostic. In the TCGA approach ≥ 60% of tumor cell nuclei and ≤ 20% necrosis were required for inclusion, and the ACRG required a similar content of tumor cells. Thus, stroma-rich tumors and aspects related to the tumor microenvironment might be underestimated in both cohorts. Moreover, in both cohorts primary tumor material but not metastases were analyzed, causing another potential bias.
In this review, key molecular alterations in gastric cancer will be addressed and the current status of the translation of targeted therapy towards clinical applications will be comprehensively reviewed. Selected pivotal clinical trials are summarized in Table 1. Furthermore, promising targets under investigation in preclinical trials will be discussed.

2. Established Molecular Targets in Gastric Cancer

2.1. HER2

Human epidermal growth factor receptor 2 (HER2; syn. ERBB2) has been identified as a key molecular target for gastric cancer treatment even since before the era of comprehensive molecular characterization of gastric cancer began. HER2 belongs to the epidermal growth factor receptor (EGFR) family of tyrosine kinase receptors, which play a pivotal role in carcinogenesis as well as in propagation of tumor cell growth and survival [16,17]. About 20% of patients with gastric or GEJ cancers show an overexpression of the HER2 protein or HER2 gene amplification [18]. HER2 positivity (HER2+) is defined by either positive immunohistochemistry (IHC) score 3+ or by IHC score 2+ plus fluorescence in situ hybridization (FISH) positivity. No difference in frequencies were found between European and Asian patients, but there were significant differences according to the histological subtype (i.e., 31.8% in intestinal type vs. 6.1% in diffuse type) and location (i.e., 32.2% in GEJ vs. 21.4% in gastric tumors) in the screening cohort of the pivotal ToGA trial [19]. According to the TCGA data set, HER2 positivity rates in the different subtypes were: CIN 24%, EBV 12%, GS 7% and MSI 5% [7].
Targeting HER2 is a well-established clinical approach in metastatic HER2+ gastric and GEJ cancers. Several pharmacological approaches have been applied so far to target HER2 in gastric cancer, comprising monoclonal antibodies, antibody-drug conjugates and tyrosine kinase inhibitors (TKI). The pivotal ToGA trial was the first to show improved overall survival (OS) in patients with HER2+ metastatic gastric cancer by adding the anti-HER2 monoclonal antibody Trastuzumab to chemotherapy compared to chemotherapy alone (median OS; 13.8 vs 11.1 months; HR 0.74, 95% CI 0.60–0.91; p = 0.0046), and a subgroup with high HER2 expression (IHC score 2+ plus FISH+ or IHC score 3+) benefited most (median OS: 16.0 vs. 11.8 months; HR 0.65, 95% CI 0.51–0.83; p = 0.0368) [3]. In contrast, the TKI Lapatinib targeting both EGFR and HER2 did not improve clinical outcome in phase III studies for stage IV HER2+ gastric cancer patients in combination with standard chemotherapy compared to chemotherapy alone, in either first-line [20] or second-line [21] therapy. Moreover, addition of the HER2 targeting antibody Pertuzumab to Trastuzumab did not further improve overall survival in HER2+ metastatic disease in first-line [22]. Finally, the antibody drug conjugate Trastuzumab Emtansin was used in second-line following anti-HER2 therapy in the GATSBY study. Compared to standard taxan treatment there were no statistically significant improvements in clinical outcomes [23].
Trastuzumab Deruxtecan is a novel antibody–drug conjugate composed of an anti-HER2 antibody, a cleavable tetrapeptide-based linker and a cytotoxic topoisomerase I inhibitor. Mechanistically, it was shown that even HER2 negative tumor cells are killed due to bystander effects [24]. In the phase II DESTINY-Gastric01 study Trastuzumab Deruxtecan was compared with standard chemotherapy in HER2+ advanced gastric cancer patients having received at least two previous lines of therapy, including anti-HER2 treatment. Objective response rate (ORR; 51% vs. 14%, p < 0.001) and overall survival (median OS; 12.5 vs. 8.4 months; HR 0.59; 95% CI, 0.39 to 0.88; p = 0.01) were significantly improved compared to standard chemotherapy. Treatment related adverse events were higher in the experimental arm; however, most adverse events were manageable [9]. Therefore, Trastuzumab Deruxtecan was approved by the FDA for patients with advanced HER2+ gastric cancer, who have received a prior Trastuzumab-based regimen. Further studies investigating Trastuzumab Deruxtecan as a monotherapy (DESTINY-Gastric02 (NCT04014075, phase II, western study population), DESTINY-Gastric04 (NCT04704934, phase III)) or in combination with chemotherapy or immunotherapy (DESTINY-Gastric03 (NCT04379596, phase I/II)) are ongoing.
A number of mechanisms leading to resistance against HER2 targeted therapies have been identified. Preclinical models have been used for the analysis of resistance mechanisms for HER2 targeting. Shin et al. established gastric cancer HER2+ patient-derived xenografts (PDX, n = 5) which were treated with Trastuzumab, Pertuzumab and a novel HER2 directed antibody (1A12) as a monotherapy or dual blockade. Data demonstrated that different HER2 directed approaches can be analyzed in PDX, and that individual response evaluation can be obtained in this model [25]. Furthermore, a number of mechanisms promoting Trastuzumab resistance have been addressed preclinically, using different cell line models. On this basis, novel treatment approaches can further be validated in PDXs and finally move to early phase clinical trials [26,27,28,29,30,31,32].
From the clinical point of view, a key problem for successfully applying molecular targeted therapies in gastric cancer is intratumoral heterogeneity. With respect to HER2+ gastric cancer, significant inter-observer variability in HER2 assessment could be identified in the recently published VARIANZ study, especially in tumor specimens with an intermediate level of HER2 expression. Importantly, in this study, borderline HER2 positivity and heterogeneity of HER2 expression were proposed as resistance factors for HER2-targeting treatment [18]. Other mechanisms comprise loss of HER2 expression during Trastuzumab treatment [33], alterations in HER2 downstream signaling related to the RAS–phosphatidylinositol 3-kinase (PI3K) signaling pathway, and the activation of bypass pathways. For example, in the CIN subtype, amplification in other receptor tyrosine kinase pathways can be frequently found, including EGFR and MET. Such co-amplifications have been found to mediate resistance towards blockade of single pathways [34]. Moreover, amplification of CCNE1 coding for the cell cycle regulator cyclin E1 occurs in conjunction with HER2 amplification and can mediate resistance towards HER2 targeting therapy [35]. More generally, it has been demonstrated that HER2+ GEJ cancers harbor several such preexisting genomic alterations, which confer resistance, but which on the other hand would in principle be targetable [28]. Consequently, combined approaches to target the different pathways altered are promising in the CIN subtype. Comprehensive molecular profiling, however, represents an important prerequisite in order to identify respective alterations or pathway activations.
Furthermore, there are a number of developments which are promising to further improve HER2 directed treatment approaches. As immunotherapy based on the use of immune checkpoint inhibitors is successfully entering the field of gastric cancer treatment (see paragraph “Immune checkpoint inhibition”), combinations of HER2-targeted therapy with immunotherapy are on the horizon. Combination of HER2-targeted therapy with immunotherapy in HER2+ cancers is supported by a number of preclinical studies. Specifically, it has been shown that anti-HER2 treatment with Trastuzumab can lead to upregulation of PD-L1 [36], enhanced dendritic and T cell invasion, induction of HER2 specific T cell responses [37,38], and the initiation of innate immune response by antibody dependent cell-mediated cytotoxicity (ADCC) [39]. In a single arm phase II trial published by Janjigian et al., addition of Trastuzumab to Pembrolizumab in HER2+ metastatic gastric cancer patients (n = 37 patients) resulted in an ORR of 91% (95% CI 78–97%) with a median progression free survival (PFS) of 13 months (95% CI 8, 6-NA) and an OS of 27, 3 months (95% CI 18, 8-NA) [40]. This encouraging data resulted in an ongoing phase III trial (KEYNOTE-811) comparing chemotherapy with Trastuzumab plus Pembrolizumab or placebo. Interim analysis revealed a significant increase in ORR in the experimental arm, with durable responses and manageable adverse events [10]. This led to approval of the combination of chemotherapy plus Trastuzumab in combination with Pembrolizumab for previously untreated HER2+ advanced/metastatic gastric cancer by the Food and Drug Administration (FDA).
A novel anti-HER2 monoclonal antibody, Margetuximab, was combined with Pembrolizumab in a single arm phase Ib/II trial in patients with previously treated HER2+ adenocarcinoma of the gastroesophageal junction and stomach (n = 95 patients). Margetuximab was designed with an optimized Fc fragment to potentiate innate immune response through ADCC. This combination showed acceptable safety and tolerability. The ORR was 18% (95% CI 11.15–27.93). However, higher objective response rates were seen in HER2+ and PD-L1+ patients [41]. These results led to enforcement of a phase II/III clinical trial investigating Margetuximab in combination with other immunotherapeutic agents [42].
Other novel compounds in clinical development comprise ZW25 (Zanidatamab), a bispecific antibody directed toward two different HER2 epitopes representing the binding sites of Trastuzumab and Pertuzumab, which has shown promising efficacy in preclinical models of different HER2+ cancers [43]. Also, several pan-HER TKIs like Tucatinib showing already efficacy in extensively pretreated HER2+ breast cancer patients and has received market authorization for this indication in combination with Trastuzumab and Capecitabine [44] or Neratinib, which was investigated in in vitro experiments using gastric cancer cell lines [45].

2.2. Targeting Angiogenesis

Tumor angiogenesis is a prerequisite of neoplastic growth. Consequently, angiogenesis was identified as a target for cancer therapy decades ago [46,47,48,49,50]. One of the key regulators of tumor angiogenesis is Vascular Endothelial Growth Factor (VEGF). VEGF mediated angiogenesis can be targeted by the anti-VEGF antibody Bevacizumab, the anti-VEGF Receptor 2 (VEGFR2) directed antibody Ramucirumab, and a large number of TKIs targeting either different VEGF receptors (i.e., VEGFR1-3) alone or in combination with other pathways.
In gastric cancer, however, results from the first-line AVAGAST [51] and AVATAR study [52] investigating the combination of Bevacizumab with chemotherapy compared to chemotherapy alone did not show an improvement in overall survival. Interestingly, in AVAGAST, regional differences in efficacy were seen with patients from European and Pan-American regions benefiting more than those from Asia. The reasons for this finding, however, are still unclear. In contrast, the anti-VEGFR2 directed antibody Ramucirumab showed efficacy in second-line treatment either as a monotherapy (REGARD) [4] or in combination with Paclitaxel (RAINBOW) [5]. Moreover, the TKI Fruquintinib (i.e., targeting VEGFR1-3) was effective in combination with paclitaxel (i.e., disease control rate 68%) in heavily pretreated gastric cancer patients in a phase I/II study in Asian patients [53] and a phase III trial in second line with this combination is currently ongoing in Asia (NCT03223376). Another TKI, Apatinib, showed improved OS, again in heavily pretreated Chinese gastric cancer patients, compared to best supportive care [54]. These data, however, could not be confirmed in another international study [55]. Furthermore, all efforts to identify biomarkers predicting efficacy of antiangiogenic treatment in gastric cancer patients thus far have failed [56,57,58,59].
Notably, as no molecular stratification was possible at the time these trials were conducted, in view of the TCGA data set recurrent amplification of the gene encoding for VEGF has been found predominantly in the CIN subgroup of gastric cancer. From this background, it might be hypothesized that this respective subgroup might benefit most from antiangiogenic treatment in general.

2.3. Immune Checkpoint Inhibition

Nivolumab monotherapy improved OS compared to placebo in third- or later-line treatment of gastric or GEJ cancer patients from Asia (Japan, South Korea, Taiwan) in the ATTRACTION 2 study, leading to approval in these countries [12]. In a global Phase II study (KEYNOTE-059), pretreated gastric or GEJ cancer patients (cohort I of the trial, n = 259) were randomly assigned to receive Pembrolizumab or placebo as a third- or later-line therapy. The objective response rate was 11.6%. The PD-L1 positive cohort (according to the combined positive score, CPS) showed better objective response rates compared to the PD-L1 negative cohort (15.5% vs. 6.4%) [60]. Based on these data, Pembrolizumab was approved by the FDA for PD-L1 CPS ≥ 1 gastric and GEJ cancer patients after disease progression (i.e., at least two previous treatment lines). In contrast, the second-line study KEYNOTE-061 addressing the role of immune checkpoint monotherapy (i.e., Pembrolizumab) vs. standard of care chemotherapy did not show a survival benefit [61]. Furthermore, the JAVELIN gastric 100 study investigating immune checkpoint blockade with Avelumab as a maintenance strategy following induction chemotherapy was also negative [62]. Tumor mutational burden (TMB) was studied in the KEYNOTE-061 study as a biomarker beyond PD-L1 expression and predicted efficiency of Pembrolizumab treatment [61,63] even when patients with MSI were excluded [63]. Specifically, a TMB beyond the cut-off value of 175 mutations/exome was significantly associated with ORR, PFS and OS in patients treated with pembrolizumab [61]. Similar results were obtained with the alternative Foundation One® CDx test using a cut-off of 10 mutations/megabase [63].
Recent data on the addition of immune checkpoint blockade to chemotherapy in locally advanced irresectable or metastatic GEJ or gastric cancer in the first-line setting were in part contradictory. In the KEYNOTE-062 phase III study, Pembrolizumab monotherapy was non-inferior to chemotherapy [11]. However, Pembrolizumab mono or Pembrolizumab plus chemotherapy were not superior to chemotherapy alone regarding OS and PFS endpoints. In the Asian ATTRACTION 4 study, PFS was improved with the addition of Nivolumab to chemotherapy in first-line therapy of gastric or GEJ cancer patients compared to chemotherapy alone. However, no improvement in OS was found and PD-L1 (tumor positive score) did not identify a subgroup with improved outcomes [64,65]. TMB was studied in both trials on an exploratory basis as an additional biomarker for immune checkpoint therapy [13,66], indicating that there might be a predictive role. On the other hand, the CheckMate-649 study demonstrated a significant improvement both for OS and PFS when adding Nivolumab to chemotherapy versus chemotherapy alone in previously untreated patients with advanced gastric or GEJ cancer with a prespecified PD-L1 expression CPS ≥ 5 [67]. In the same direction, not focusing on gastric cancer patients, the KEYNOTE-590 demonstrated that addition of Pembrolizumab to chemotherapy in first-line treatment improved OS and PFS in GEJ adenocarcinoma patients with a PD-L1 CPS ≥ 10 compared to chemotherapy alone [68].
Regarding the TCGA classification, MSI and EBV subtypes have been shown to benefit extensively from immune checkpoint blockade [69]. High TMB resulting in extensive formation of cancer-specific neoantigens triggering robust anti-cancer immune responses is regarded as a key driver in the MSI subtype. Within the EBV subtype of gastric cancer, PD-L1 overexpression was shown to be triggered by two mechanisms: focal amplification of CD274 (PD-L1) and IFN-γ-mediated signaling via activation of IRF3 to increase PD-L1 expression [70]. Interestingly, 9p amplifications are enriched in the EBV subgroup (15% of tumors) according to the TCGA data set [7]. The genes CD274 and PDCD1LG2 are located in this region, and encode PD-L1 and PD-L2. Moreover, JAK2 amplification, also often present in the EBV gastric cancer subtype, may even enhance PD-L1 expression, as has been shown in the context of triple negative breast cancer. Remarkably, in classical Hodgkin´s Lymphoma (cHL), 9p amplifications are a defining feature [71]. Of note, cHL is extremely sensitive to immune checkpoint blockade [72].
Accordingly, in a prospective phase II study, an ORR of 100% was reported for EBV positive gastric cancer patients, and ORR was even slightly lower (i.e., 85.7%) in MSI gastric cancer patients [69]. Analysis of concurrent alterations in EBV positive gastric cancer (i.e., PIK3CA mutations, HER2 amplification) appear promising to identify mechanisms of resistance towards immunotherapy or to propose novel combinations [73]. Combinations of multi-TKI with immune checkpoint inhibitors are currently in clinical development. Promising data from a phase II trial exist for a combination of Regorafenib with Nivolumab in a cohort of microsatellite stable (MSS) mismatch repair proficient colorectal cancer patients [74], and in the Phase Ib REGONIVO, EPOC1603 trial toxicities were manageable in colorectal and gastric cancer patients. The gastric cancer cohort comprised 25 patients treated with ≥ 2 previous treatment lines. ORR of 44% and a median PFD of 5.6 months were promising [75]. Moreover, the multi TKI Lenvatinib was tested in combination with Pembrolizumab [76] in the phase II LEAP-005 study, and a phase III trial in patients with advanced or metastatic gastric or GEJ cancer in combination with chemotherapy in the first-line treatment is underway (NCT04662710).

3. Potential Novel Molecular Targets for Gastric Cancer Treatment

3.1. EGFR

The epidermal growth factor receptor (EGFR; syn. ERBB1) has been extensively studied as a target in gastric cancer based on preclinical findings that this pathway is involved in tumor progression and metastasis [17]. While overexpression of EGFR at the protein level was reported at variable frequencies ranging from 30–60% in diverse cohorts [16], EGFR amplification was found in only 4% of cases from the TCGA data set and pathway activation was reported in 21%, with a predominance in the CIN subtype (10%). Initial unselected phase III trials adding EGFR antibodies (Cetuximab or Panitumumab) to standard chemotherapy did not find improvement in clinical outcomes of patients with advanced or metastatic gastric or GEJ cancer in the first-line setting in the EXPAND or REAL3 trial, respectively [77,78]. Notably, in the REAL3 trial the OS outcome for the combination of EOX (i.e., Epirubicin, Oxaliplatin, Capecitabine) with Panitumumab was significantly worse compared with the standard arm of EOX [78]. Moreover, a study using the TKI Gefitinib in second to fourth line treatment did not demonstrate improved outcomes compared to standard chemotherapy [79]. Biomarker analysis from the EXPAND trial indicated that a very small subgroup of patients with very high EGFR expression (i.e., measured by IHC) might have a benefit from the addition of Cetuximab, while data on the amplification status were not available [80]. In a large cohort of gastric cancer patients from the U.S., EGFR amplification frequency was 6% and the retrospective cohort and patients in the prospective part of the study (n = 140; n = 8 amplified), treated at different treatment lines, showed a benefit for EGFR-directed therapy (i.e., Cetuximab or ABT-806, an investigational anti-EGFR antibody) according to the primary endpoint best overall response: complete response 3/7 (43%), partial response 1/7 (14%) and disease control 3/7 (43%) [79]. Importantly, alterations in related pathways as well as tumor heterogeneity were identified as resistance mechanisms (loss of EGFR amplification, PTEN deletions, KRAS amplifications/mutations, NRAS, MYK and HER2 amplifications) [81]. Furthermore, ctDNA analysis was able to monitor EGFR amplification status during treatment and correlated with the efficacy of anti-EGFR treatment. Finally, increased immune cell infiltration (i.e., CD3+ T-cells, NK-cells) as well as increased PD-L1 expression, which is otherwise low in EGFR amplified patients, were observed in on-treatment biopsies but were lost at progression. These findings, relying on small numbers, are hypothesis-generating, suggesting combinations of anti-EGFR antibodies with immune checkpoint inhibition [81]; however, this needs validation in larger cohorts. Recently, data from the REAL3 trial [78] have indicated that EGFR-amplified cases can been identified by analysis of circulating tumor DNA (ctDNA) using droplet polymerase chain reaction or in tumor tissue using FISH analysis. Interestingly, in contrast to previous results showing a benefit for anti-EGFR antibody monotherapy in EGFR amplified gastric cancer patients [81], EGFR-amplified patients from REAL3 treated with the EGFR antibody Panitumumab plus EOX chemotherapy had a particularly adverse survival outcome compared to chemotherapy alone [82]. Mechanistically, using tumor organoid models, an antagonistic effect between anti-EGFR agents and the chemotherapy drug Epirubicin specifically in EGFR-amplified organoids was found [81], indicating, that the chemotherapy backbone might have an impact on the outcome.

3.2. MET Alterations

The MET gene encodes Hepatocyte Growth Factor (HGF) receptor, which is a member of the receptor tyrosine kinase family involved in the regulation of cell growth and differentiation in health and disease. In gastric cancer, MET represents a major oncogenic driver (i.e., proto-oncogene), and activation of the pathway promotes disease progression [83] as well as resistance to HER2-targeted treatment strategies [84] and represents an adverse prognostic factor [85]. Important growth-promoting downstream pathways are activated by MET, such as the PI3K/AKT/mTOR pathway the RAS/RAF/ERK/MAPK pathway, the STAT-3 pathway and NFkB. Consequently, treatment strategies have been developed to target MET pathway activation. While up to 50% of gastric cancers show an overexpression on the protein level by IHC, only 3–4% are characterized by MET gene amplification [86] and this often occurs together with amplifications in EGFR, HER2 and other receptor tyrosine kinases and is related to the CIN subtype according to TCGA [7].
Following promising data from a phase II study in gastric cancer patients overexpressing MET [87], the monoclonal antibody Rilotumumab targeting the MET ligand HGF has been tested in phase III studies in first-line treatment of metastatic gastric cancer patients with MET positive tumors (i.e., according predefined IHC-based criteria), in combination with standard chemotherapy. The international RILOMET-1 study, however, failed to demonstrate an improvement in survival outcomes [88] and the RILOMET-2 trial [89], which was exclusively focused on Asian patients, was terminated prematurely due to an increased risk of death with the study drug Rilotuzumab in an interim safety analysis. Similarly, a phase III trial of the antibody directly targeting MET (Onartuzumab) in metastatic gastric cancer patients with MET positive tumors according predefined IHC-based criteria in combination with standard chemotherapy failed to demonstrate a survival benefit [90]. In addition, several TKIs targeting MET have been studied in gastric cancer patients in phase I or II trials. In the VIKTORY umbrella trial, there was a promising signal (i.e., ORR 50%) for the activity of the MET inhibitor Savolitinib in gastric cancer patients, and this compound is currently under further clinical development [91]. Mostly, however, efficacy of MET targeting TKI was marginal [83].
Key factors contributing to the disappointing results thus far are related to the heterogeneity of target expression found in gastric cancer, difficulties in reliably measuring MET overexpression/activation, and the activation of alternative signaling pathways specifically in the TCGA CIN subgroup, characterized by multiple concurrent amplifications in related pathways [83]. Moreover, translational research data from studies related to the VIKTORY trial indicated that novel occurring mutations in MET (i.e., MET D1228V/N/H and Y1230C) or high copy number MET gene amplifications contribute to the development of resistance against Savolitinib, which can be identified in the circulating tumor DNA (ctDNA) from blood samples [92]. Consequently, novel combination approaches targeting such related pathways and combinations with immune checkpoint inhibitors are promising; as for the latter, MET amplification has been identified as a resistance mechanism in lung cancer [93]. Moreover, MET Exon 14 skipping mutations, which leads to an activation of the MET pathway, can be targeted by specific TKIs (i.e., Capmatinib, Tepotinib). These compounds have already received market authorization for the treatment of Non-Smallcell Lung Cancer (NSCLC) patients harboring this alteration based on phase III studies [94,95]. Since MET Exon 14 skipping mutations have also been identified in gastric cancer in up to 7% of cases, these compounds are very promising for this subgroup.

3.3. FGFR2 Alterations

Alterations in FGF receptor genes are found in a wide variety of cancer entities ranging from < 1% in sarcomas to 32% in urothelial carcinoma [96]. In gastric and GEJ cancers, frequency ranges between 3–7% and amplifications are the most common alteration, followed by rearrangements and mutations [96]. In a cohort of 269 gastric cancer patients, the overall frequency of FGFR2 alterations was 4%, consisting of 72% amplifications, 13% mutations and 8.6% rearrangements. Multiple FGFR2 alterations were found in 6.3%. The level of amplification was shown to negatively affect chemotherapy response and prognosis [97]. FGFR2 pathway activation is overrepresented in the GS (9%) and CIN (8%) subtypes according to TCGA. With regard to biomarker development, FGFR2 amplification was shown to predict efficacy of the multi TKI Regorafenib, targeting FGFR2 amongst others, in preclinical models [97]. Small molecular compounds targeting FGFR have successfully been introduced in the clinic for the treatment of urothelial cancer (i.e., Erdafitinib, a pan-FGFR inhibitor) or cholangiocarcinoma (i.e., Pemigatinib, targeting FGFR1-3) and companion biomarkers related to FGFR alterations have been developed [98,99]. In gastric cancer, AZD4547, a selective FGFR-1, 2, 3 TKI which showed good preclinical activity in FGFR2 amplified gastric cancer patient derived xenograft models [100,101], failed to improve PFS in the second-line treatment of advanced gastric cancer compared to paclitaxel; intratumoral heterogeneity of FGFR2 expression was proposed as a possible explanation [102].
Moreover, a novel covalent, irreversible FGFR inhibitor (TAS-120, Frutibatinib) is in clinical development [103]. This TKI overcomes resistance to ATP-competitive FGFR inhibitors [104]. A clinical trial in a mixed cohort of advanced cancer patients including gastric cancer patients harboring FGFR2 alterations is currently underway [105]. Finally, Bemarituzumab, which represents a first-in-class humanized afucosylated IgG1 monoclonal antibody to FGFR2b (i.e., a splice variant of FGFR2) has shown promising clinical data in a phase I study in advanced solid tumors and FGFR2b selected gastric and GEJ cancer patients [15]. In the pretreated gastric and GEJ cohort characterized by FGFR2 amplification, a partial response rate of 17.9% was reached [15]. In a phase II study (FIGHT trial) Bemarituzumab plus chemotherapy (i.e., FOLFOX) was tested in first-line therapy in patients with unresectable locally advanced or metastatic HER2 negative gastric cancer with FGFR2b overexpression (i.e., IHC or FGFR2 amplification by circulating tumor DNA (ctDNA) analysis). Specifically, median PFS was improved to 9.5 months in the Bemarituzumab arm, compared to 7.4 months (hazard ratio (HR), 0.68; 95% confidence interval (CI), 0.44–1.04; p = 0.07) in the placebo arm. Median OS was not reached in the Bemarituzumab arm vs. 12.9 months in the placebo arm (HR, 0.58, 95% CI, 0.35–0.95; p = 0.03). ORR was improved from 40% to 53% in the Bemarituzumab arm [106,107]. While the treatment was overall well tolerated, eye toxicity (i.e., corneal adverse events) was more common in the Bemaritzumab arm (67% vs 10%) [108], representing a class effect of FRGR-inhibitors.

3.4. Claudin 18.2

The TCGA gastric cancer study has identified CLDN18-ARHGAP26/6 fusions in 15% of the patients, which predominantly occurred in the genomically stable subtype [7]. Nearly all tumors with CLDN18-ARHGAP26/6 fusions show claudin 18.2 (CLDN18.2) expression [109]. CLDN18.2 is an isoform of the claudin protein and belongs to structural components of tight junctions. It is expressed in various types of tumors but not in normal tissue except gastric mucosa [14]. In gastric cancer, about 40% of patients present with an overexpression according to immunohistochemistry [110]. The monoclonal antibody Zolbetuximab binds to CLDN18.2, and induced cancer cell death through antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. Zolbetuximab was studied in two phase II trials as a single agent therapy (MONO trial) and in combination with chemotherapy (FAST trial) in advanced CLDN18.2 positive gastric cancer patients [111,112]. The FAST trial showed beneficial effects of adding Zolbetuximab to standard first-line chemotherapy with prolonged PFS (HR 0.44; 95% CI 0.29–0.67; p < 0.0005) and OS (HR 0.55; 95% CI 0.39–0.77; p < 0.0005) in CLDN18.2 positive (>40% positive tumor cells) gastric or GEJ cancer [111]. Further validation of these results is currently being attempted in two phase III trials (NCT03504397, NCT03653507), which are investigating Zolbetuximab in combination with chemotherapy in the first-line setting in CLDN18.2 positive (>75% positive tumor cells) gastric and GEJ cancer. In addition, CLDN18.2 is being studied as a target for CAR-T cells (NCT04404595, NCT04467853) as well as for a bispecific T cell engager (BiTE) (NCT04260191) in phase I trials. The AMG 910 trial is investigating a CLDN18.2/CD3 directed BiTE to trigger tumor cell lysis by inducing T-cell activation and proliferation in advanced stage gastric and GEJ cancer patients.

3.5. MUC17

Another promising target in gastric and GEJ cancers is mucin 17 (MUC17), which belongs to the mucin family of glycosylated protein. Mucins are transmembrane proteins of the mucosal barrier of normal gastrointestinal epithelial cells [113]. MUC17 is overexpressed in gastric cancer cells, and expression in normal cells is limited. Therefore, a BiTE designed to engage MUC17 positive cells and CD3 positive cells is currently being investigated in a phase I trial ([114], NCT04117958).

3.6. Matrix Metalloproteinase-9

In cancer, matrix metalloproteinases are commonly upregulated in the tumor microenvironment. Matrix metalloproteinase-9 (MMP-9) is an extracellular enzyme which is involved in angiogenesis, matrix remodeling, tumor growth and metastases. It has been shown that inhibition of MMP-9 leads to improved penetration of chemotherapeutics and immune cells [115]. In early gastric cancer, MMP-9 expression levels were prognostic for survival [116]. To target MMP-9, the monoclonal antibody Andecaliximab was developed and encouraging results from a phase I trial investigating Andecaliximab in combination with chemotherapy in HER2 negative gastric and EGJ cancer led to initiation of a phase III trial in the first-line setting. Disappointingly, addition of Andecaliximab to chemotherapy did not improve OS in patients with untreated HER2-negative gastric or GEJ adenocarcinoma [117,118].

3.7. Homologous Recombination Deficiency

Homologous recombination deficiency (HRD) sensitizes tumor cells towards treatment with PARP inhibitors leading to synthetic lethality. This treatment approach is now well established in several cancer entities [119,120,121,122,123,124], and germline or somatic BRCA mutations represent the most widely used biomarkers. Other mutations are also related to HRD, such as ATM, PALB2, and RAD51. Furthermore, loss of heterozygosity represents another characteristic phenomenon in HRD deficient tumors. Accordingly, different methods have been used to identify HRD tumors. These comprise sequencing approaches to identify mutations in HRD related genes or to identify genomic scars resulting from HDR deficiency, and analysis of chromosomal aberrations as a consequence of HDR. On this basis, efforts are ongoing to establish and validate optimized biomarkers in order to define HDR in the context of treatment with PARP inhibitors [125]. In gastric cancer, the frequency of mutations in HDR related genes (22 genes analyzed) related to HDR was overall 32% in a large cohort (n = 395). Amongst them, ATM was the most frequently mutated with a frequency ranging from 19% in mucinous adenocarcinoma to 4% in diffuse type adenocarcinoma [126]. Another group has identified a mutation pattern characterized by the occurrence of large (i.e., ≥ 3 base pairs) indels to predict efficacy of Cisplatin treatment in pancreatic cancer even in the absence of BRCA mutations; this mutational pattern was found in up to 12% in gastric cancer, mostly in the mixed (9%) and the intestinal type (12%) according to Laurén classification [127]. Consequently, treatment with PARP inhibitors might have a role in a distinct subgroup of gastric cancer patients with HDR deficiency. In the REAL3 study patients were treated with chemotherapy (i.e., EOX) with or without Panitumumab and translational studies indicated that greater loss of heterogeneity was associated with longer OS, possibly attributable to the increased activity of platinum compounds (i.e., Oxaliplatin) in HDR deficient tumors [128]. In a cohort of familial diffuse gastric cancer not mutated in CDH1, germline mutations in HDR associated genes (i.e., BRCA1, PALB2, RAD51C) were identified in 6/31 patients (6.5%); such patients might also benefit from treatment with cisplatin and/or PARP inhibitors [129].
In the unselected phase II study 39 (NCT01063517), second-line treatment of Asian patients with Olaparib plus Paclitaxel versus placebo plus Paclitaxel showed an improvement in PFS. Moreover, a significant improvement in the secondary endpoint OS was seen in a subgroup of patients with ATM negative (i.e., according to IHC) tumors [130]. Based on these data, the phase III GOLD trial was performed, which failed to demonstrate an OS benefit in a similarly selected cohort of Asian patients [131]. Further translational analyses (i.e., NGS-based analysis of 15 HDR related genes and analysis of loss of heterogeneity) indicated better outcomes in the cohort (22 of 378 patients) with ATM mutations [132].
In general, how to best identify HDR is still a very important research area for predicting the efficacy of PARP inhibitors beyond BRCA testing. Furthermore, combinations of PARP inhibitors with immune checkpoint blockade, small molecule inhibitors or anti-angiogenic compounds are under way [133] and initial results are promising [134].

3.8. Potential Targets in Genomically Stable Gastric Cancer

The GS subtype of gastric cancer carries the most adverse prognosis. It is characterized by the activation of Epithelial to Mesenchymal Transition (EMT) programs. RHOA mutations and CLDN18-ARHGAP6 or ARHGAP26 fusions were found in this subtype, and the frequency of CDH1 mutations was 26% in the TCGA data set. Functionally, all these factors are related to cell motility and invasion. This corresponds very well with the finding that the GS subtype mostly corresponds to the diffuse histological subtype of the Laurén classification, characterized by a discohesive growth pattern. Currently, it is likely that CLDN18-ARHGAP6 or ARHGAP26 fusions will be targetable with CLDN18.2 antibodies, though still unproven. However, RHOA mutations were demonstrated to promote Focal Adhesion Kinase (FAK) activation and dependency [135]. Moreover, FAK representing a cytoplasmic protein tyrosine kinase, is overexpressed in several solid cancers, and the development of small molecule FAK inhibitors makes this pathway druggable [136]. On the other hand, sporadic early- onset diffuse gastric cancer is characterized by a high frequency of somatic CDH1 alterations, and as ROS-1 inhibition has been demonstrated to confer synthetic lethality in CDH1 mutated breast cancer [137]; this approach may represent an attractive strategy for the treatment of gastric cancer with CDH1 alterations as well.

4. Approaches to Further Improve Precision Medicine in Gastric Cancer

4.1. Molecular Tumor Boards

Despite recent development of established novel targeted therapies in gastric cancer, translation from bench to bedside often fails as most drugs selected by preclinical testing are not effective in phase II/III clinical trials [138]. The availability of high throughput sequencing technologies and of several targeted therapies has resulted in more personalized treatment approaches in late-stage disease. Molecular tumor boards which discuss individual patients’ high throughput sequencing reports in order to suggest individual treatment approaches are commonly available in comprehensive cancer centers. It has been shown that the implementation of molecular tumor boards and individual treatment decision making seem to lead to improved outcomes compared to conventional therapy [139]. However, these technologies are still expensive and not yet available for most patients from a global perspective. Additionally, the ultimate proof of individual treatment decisions for non-approved drugs based on molecular tumor board recommendations is still lacking.

4.2. Ex Vivo Tumor Models

Besides the genomic approach, patient-derived ex vivo tumor models have been investigated as an approach for individual patient stratification [140]. Essentially, in these models, tumor specimens are collected and utilized to establish patient derived xenografts, organoids or tissue cultures [141,142,143]. For gastric cancer, several studies of gastric cancer organoids have been performed and predictability of individual response to treatment and patient outcomes was observed [144,145,146]. Furthermore, patient derived tissue cultures have been established for gastric and GEJ cancer [147]. It was also shown that this model can be used to study PD-1 inhibition ex vivo, as immune contexture can be preserved in patient derived tissue cultures [148]. However, due to the complexity of this model, the predictive values for patient response to specific therapies have not yet been proved. Additionally, patient derived xenografts have been investigated by several groups and the potential for response prediction and analysis of targeted therapy for specific gastric cancer subtypes have been performed [149,150,151,152]. Although preclinical tumor models can be used for response prediction, analysis of resistance mechanism, and tumor biology, none have been implemented in daily clinical routine due to missing validation in bigger cohorts, high costs and extensive workload.

4.3. Novel Trial Designs

Novel clinical trial designs such as basket and umbrella trials are increasingly used to study the efficacy of biomarker directed targeted therapy strategies. In the area of gastric cancer, the PANGAE and VIKTORY studies are important examples [91,153].
The VIKTORY (targeted agent eValuation In gastric cancer basket KORea) trial aimed to attribute metastatic gastric cancer patients to specific targeted drugs based on eight different biomarker groups identified by NGS (RAS aberration, TP53 mutation, PIK3CA mutation/amplification, MET amplification, and MET overexpression all negative, TSC2 deficient, or RICTOR amplification). Based on the results, patients were assigned to 10 different associated clinical trials for second-line treatment with the following drugs: Capivasertib (AKT inhibitor), Savolitinib (MET inhibitor), Selumetinib (MEK inhibitor), Adavosertib (WEE1 inhibitor), and Vistusertib (TORC inhibitor), which were tested with or without chemotherapy. Some 772 patients with gastric cancer were enrolled, and 105 patients finally received biomarker-assigned drug treatment. Compared with standard second-line treatment, the biomarker-assigned treatment cohort had encouraging response rates and survival (mOS 9.8 vs. 6.9 months, median PFS 5.7 vs. 3.8 months) [91]. Moreover, ctDNA analysis demonstrated good correlation between high MET copy numbers and treatment efficacy, indicating that tumor heterogeneity can be captured by this method [91].
In the PANGEA phase II trial [153], a personalized treatment strategy was applied in gastric cancer patients: antibodies targeting HER2, MET, FGFR2 EGFR PD-L1 or VEGFR2 were added to chemotherapy in first- to third-line treatment in gastric or GEJ cancer patients. Treatment selection was done according to a panel of biomarkers (i.e., amplifications in HER2, MET, FGFR2, EGFR) or biomarkers related to efficiency of immune checkpoint therapy (i.e., MSI high, EBV positive, PD-L1 CPS ≥ 10 or TMB ≥ 15 mutations/megabase). One-year survival rate (n = 68 patients) was 66% and median OS was 15.7 months. Moreover, first-line response rate (74%), disease control rate (99%), and median progression-free survival (8.2 months) were superior to historical controls. Importantly, in case metastatic tissue specimens were not sufficient to complete all assays and biomarker assignment, ctDNA could be used for biomarker assignment, underscoring the potential future role of this method for targeted treatment approaches in gastric cancer. The clinical utility of ctDNA analysis in advanced gastrointestinal cancers has recently been validated [154] and this approach is very promising for assigning gastric cancer patients for targeted therapies in future clinical trials [155].

5. Conclusions

Various targeted therapy approaches have been investigated and specific biomarkers like HER2, MSI and PD-L1 currently play a pivotal role in clinical treatment decision making in gastric cancer. HER2 targeted therapy and anti-angiogenic treatment in combination with chemotherapy represent well-established therapeutic strategies in stage IV gastric cancer, and immune checkpoint blockade is rapidly evolving as a therapeutic strategy as well. Several clinical trials investigating targeted therapy in gastric cancer have failed, however, due to tumor heterogeneity and insufficient biomarker-based stratification approaches. Despite this drawback, new molecular targets (e.g., CLDN18.2, FGFR2) have been identified in subgroups of patients, and promising treatment efficacy was observed in several clinical trials. In addition, new treatment modalities like bispecific T-cell engager and CAR-T cells are under investigation. Finally, the TCGA dataset has built a roadmap for the development of novel molecularly stratified treatment approaches. Based on this, specific genomic alterations may serve as novel treatment targets, as they already are or will become druggable by the development of promising compounds in the future.

Author Contributions

Conceptualization, J.K., F.L. and U.T.H.; data curation, J.K. and U.T.H.; writing-original draft preparation, J.K. and U.T.H.; writing-review and editing, J.K., U.T.H. and F.L.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

F.L. declares the following conflict of interests: Bristol Myers Squibb (research funding, honoraria), Merck Sharp and Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA (research funding, honoraria), Amgen (honoraria), Astellas Pharma, Inc. (honoraria), AstraZeneca (honoraria), Eli Lilly (honoraria) Elsevier (honoraria), Incyte (honoraria), Medscape (honoraria), MedUpdate (honoraria), Novartis (honoraria), Roche (honoraria), Servier (honoraria), Springer Nature (honoraria), StreamedUp! (honoraria), Zymeworks (honoraria). J.K. and U.T.H. declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef]
  3. Bang, Y.J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; 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. [Google Scholar] [CrossRef]
  4. Fuchs, C.S.; Tomasek, J.; Yong, C.J.; Dumitru, F.; Passalacqua, R.; Goswami, C.; Safran, H.; dos Santos, L.V.; Aprile, G.; Ferry, D.R.; 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. [Google Scholar] [CrossRef]
  5. Wilke, H.; Muro, K.; Van Cutsem, E.; Oh, S.C.; Bodoky, G.; Shimada, Y.; Hironaka, S.; Sugimoto, N.; Lipatov, O.; Kim, T.Y.; 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. [Google Scholar] [CrossRef]
  6. Smyth, E.C.; Verheij, M.; Allum, W.; Cunningham, D.; Cervantes, A.; Arnold, D.; Committee, E.G. Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2016, 27, v38–v49. [Google Scholar] [CrossRef] [PubMed]
  7. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Cristescu, R.; Lee, J.; Nebozhyn, M.; Kim, K.M.; Ting, J.C.; Wong, S.S.; Liu, J.; Yue, Y.G.; Wang, J.; Yu, K.; et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 2015, 21, 449–456. [Google Scholar] [CrossRef]
  9. Shitara, K.; Bang, Y.J.; Iwasa, S.; Sugimoto, N.; Ryu, M.H.; Sakai, D.; Chung, H.C.; Kawakami, H.; Yabusaki, H.; Lee, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N. Engl. J. Med. 2020, 382, 2419–2430. [Google Scholar] [CrossRef]
  10. Janjigian, Y.Y.; Kawazoe, A.; Yanez, P.E.; Luo, S.; Lonardi, S.; Kolesnik, O.; Barajas, O.; Bai, Y.; Shen, L.; Tang, Y.; et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2+ metastatic gastric or gastroesophageal junction (G/GEJ) cancer: Initial findings of the global phase 3 KEYNOTE-811 study. J. Clin. Oncol. 2021, 39, 4013. [Google Scholar] [CrossRef]
  11. Shitara, K.; Van Cutsem, E.; Bang, Y.J.; Fuchs, C.; Wyrwicz, L.; Lee, K.W.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; 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. [Google Scholar] [CrossRef] [PubMed]
  12. Kang, Y.K.; Boku, N.; Satoh, T.; Ryu, M.H.; Chao, Y.; Kato, K.; Chung, H.C.; Chen, J.S.; Muro, K.; Kang, W.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, 2461–2471. [Google Scholar] [CrossRef]
  13. Wyrwicz, L.S.; Lee, K.W.; Van Cutsem, E.; Bang, Y.J.; Fuchs, C.S.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; Castro, H.R.; et al. 1442P Association of TMB using the Foundation Medicine Companion Diagnostic (F1CDx) with efficacy of first-line pembrolizumab (pembro) or pembro plus chemotherapy (pembro + chemo) versus chemo in patients with gastric cancer (gc) from KEYNOTE-062. Ann. Oncol. 2020, 31, S907–S908. [Google Scholar] [CrossRef]
  14. Sahin, U.; Koslowski, M.; Dhaene, K.; Usener, D.; Brandenburg, G.; Seitz, G.; Huber, C.; Tureci, O. Claudin-18 splice variant 2 is a pan-cancer target suitable for therapeutic antibody development. Clin. Cancer Res. 2008, 14, 7624–7634. [Google Scholar] [CrossRef] [Green Version]
  15. Catenacci, D.V.; Rasco, D.; Lee, J.; Rha, S.Y.; Lee, K.W.; Bang, Y.J.; Bendell, J.; Enzinger, P.; Marina, N.; Xiang, H.; 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, 2418–2426. [Google Scholar] [CrossRef] [PubMed]
  16. Arienti, C.; Pignatta, S.; Tesei, A. Epidermal Growth Factor Receptor Family and its Role in Gastric Cancer. Front. Oncol. 2019, 9, 1308. [Google Scholar] [CrossRef]
  17. Normanno, N.; De Luca, A.; Bianco, C.; Strizzi, L.; Mancino, M.; Maiello, M.R.; Carotenuto, A.; De Feo, G.; Caponigro, F.; Salomon, D.S. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene 2006, 366, 2–16. [Google Scholar] [CrossRef]
  18. Haffner, I.; Schierle, K.; Raimundez, E.; Geier, B.; Maier, D.; Hasenauer, J.; Luber, B.; Walch, A.; Kolbe, K.; Riera Knorrenschild, J.; et al. HER2 Expression, Test Deviations, and Their Impact on Survival in Metastatic Gastric Cancer: Results from the Prospective Multicenter VARIANZ Study. J. Clin. Oncol. 2021, 39, 1468–1478. [Google Scholar] [CrossRef] [PubMed]
  19. Van Cutsem, E.; Bang, Y.J.; Feng-Yi, F.; Xu, J.M.; Lee, K.W.; Jiao, S.C.; Chong, J.L.; Lopez-Sanchez, R.I.; Price, T.; Gladkov, O.; et al. HER2 screening data from ToGA: Targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer 2015, 18, 476–484. [Google Scholar] [CrossRef]
  20. Hecht, J.R.; Bang, Y.J.; Qin, S.K.; Chung, H.C.; Xu, J.M.; Park, J.O.; Jeziorski, K.; Shparyk, Y.; Hoff, P.M.; Sobrero, A.; 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. [Google Scholar] [CrossRef] [Green Version]
  21. Satoh, T.; Xu, R.H.; Chung, H.C.; Sun, G.P.; Doi, T.; Xu, J.M.; Tsuji, A.; Omuro, Y.; Li, J.; Wang, J.W.; 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. [Google Scholar] [CrossRef]
  22. Tabernero, J.; Hoff, P.M.; Shen, L.; Ohtsu, A.; Shah, M.A.; Cheng, K.; Song, C.; Wu, H.; Eng-Wong, J.; Kim, K.; 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. [Google Scholar] [CrossRef]
  23. Thuss-Patience, P.C.; Shah, M.A.; Ohtsu, A.; Van Cutsem, E.; Ajani, J.A.; Castro, H.; Mansoor, W.; Chung, H.C.; Bodoky, G.; Shitara, K.; 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. [Google Scholar] [CrossRef]
  24. Doi, T.; Shitara, K.; Naito, Y.; Shimomura, A.; Fujiwara, Y.; Yonemori, K.; Shimizu, C.; Shimoi, T.; Kuboki, Y.; Matsubara, N.; et al. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody–drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: A phase 1 dose-escalation study. Lancet Oncol. 2017, 18, 1512–1522. [Google Scholar] [CrossRef]
  25. Shin, S.H.; Park, S.S.; Ju, E.J.; Park, J.; Ko, E.J.; Hwang, J.J.; Suh, Y.A.; Jang, S.J.; Lee, J.S.; Ko, B.K.; et al. Establishment of a Patient-derived Xenograft for Development of Personalized HER2-targeting Therapy in Gastric Cancer. Anticancer Res. 2018, 38, 287–293. [Google Scholar] [CrossRef] [PubMed]
  26. Zheng, L.; Tan, W.; Zhang, J.; Yuan, D.; Yang, J.; Liu, H. Combining trastuzumab and cetuximab combats trastuzumab-resistant gastric cancer by effective inhibition of EGFR/ErbB2 heterodimerization and signaling. Cancer Immunol. Immunother. 2014, 63, 581–586. [Google Scholar] [CrossRef] [PubMed]
  27. Piro, G.; Carbone, C.; Cataldo, I.; Di Nicolantonio, F.; Giacopuzzi, S.; Aprile, G.; Simionato, F.; Boschi, F.; Zanotto, M.; Mina, M.M.; et al. An FGFR3 Autocrine Loop Sustains Acquired Resistance to Trastuzumab in Gastric Cancer Patients. Clin. Cancer Res. 2016, 22, 6164–6175. [Google Scholar] [CrossRef] [Green Version]
  28. Kim, J.; Fox, C.; Peng, S.; Pusung, M.; Pectasides, E.; Matthee, E.; Hong, Y.S.; Do, I.G.; Jang, J.; Thorner, A.R.; et al. Preexisting oncogenic events impact trastuzumab sensitivity in ERBB2-amplified gastroesophageal adenocarcinoma. J. Clin. Investig. 2014, 124, 5145–5158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Yoshioka, T.; Shien, K.; Takeda, T.; Takahashi, Y.; Kurihara, E.; Ogoshi, Y.; Namba, K.; Torigoe, H.; Sato, H.; Tomida, S.; et al. Acquired resistance mechanisms to afatinib in HER2-amplified gastric cancer cells. Cancer Sci. 2019, 110, 2549–2557. [Google Scholar] [CrossRef] [Green Version]
  30. Mezynski, M.J.; Farrelly, A.M.; Cremona, M.; Carr, A.; Morgan, C.; Workman, J.; Armstrong, P.; McAuley, J.; Madden, S.; Fay, J.; et al. Targeting the PI3K and MAPK pathways to improve response to HER2-targeted therapies in HER2-positive gastric cancer. J. Transl. Med. 2021, 19, 184. [Google Scholar] [CrossRef] [PubMed]
  31. Liu, J.; Pan, C.; Guo, L.; Wu, M.; Guo, J.; Peng, S.; Wu, Q.; Zuo, Q. A new mechanism of trastuzumab resistance in gastric cancer: MACC1 promotes the Warburg effect via activation of the PI3K/AKT signaling pathway. J. Hematol. Oncol. 2016, 9, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Shi, J.; Li, F.; Yao, X.; Mou, T.; Xu, Z.; Han, Z.; Chen, S.; Li, W.; Yu, J.; Qi, X.; et al. The HER4-YAP1 axis promotes trastuzumab resistance in HER2-positive gastric cancer by inducing epithelial and mesenchymal transition. Oncogene 2018, 37, 3022–3038. [Google Scholar] [CrossRef] [Green Version]
  33. Pietrantonio, F.; Caporale, M.; Morano, F.; Scartozzi, M.; Gloghini, A.; De Vita, F.; Giommoni, E.; Fornaro, L.; Aprile, G.; Melisi, D.; et al. HER2 loss in HER2-positive gastric or gastroesophageal cancer after trastuzumab therapy: Implication for further clinical research. Int. J. Cancer 2016, 139, 2859–2864. [Google Scholar] [CrossRef] [Green Version]
  34. Kwak, E.L.; Ahronian, L.G.; Siravegna, G.; Mussolin, B.; Borger, D.R.; Godfrey, J.T.; Jessop, N.A.; Clark, J.W.; Blaszkowsky, L.S.; Ryan, D.P.; et al. Molecular Heterogeneity and Receptor Coamplification Drive Resistance to Targeted Therapy in MET-Amplified Esophagogastric Cancer. Cancer Discov. 2015, 5, 1271–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lee, J.Y.; Hong, M.; Kim, S.T.; Park, S.H.; Kang, W.K.; Kim, K.M.; Lee, J. The impact of concomitant genomic alterations on treatment outcome for trastuzumab therapy in HER2-positive gastric cancer. Sci. Rep. 2015, 5, 9289. [Google Scholar] [CrossRef] [Green Version]
  36. Chaganty, B.K.; Qiu, S.; Gest, A.; Lu, Y.; Ivan, C.; Calin, G.A.; Weiner, L.M.; Fan, Z. Trastuzumab upregulates PD-L1 as a potential mechanism of trastuzumab resistance through engagement of immune effector cells and stimulation of IFNgamma secretion. Cancer Lett. 2018, 430, 47–56. [Google Scholar] [CrossRef] [PubMed]
  37. Taylor, C.; Hershman, D.; Shah, N.; Suciu-Foca, N.; Petrylak, D.P.; Taub, R.; Vahdat, L.; Cheng, B.; Pegram, M.; Knutson, K.L.; et al. Augmented HER-2 specific immunity during treatment with trastuzumab and chemotherapy. Clin. Cancer Res. 2007, 13, 5133–5143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Zum Buschenfelde, C.M.; Hermann, C.; Schmidt, B.; Peschel, C.; Bernhard, H. Antihuman epidermal growth factor receptor 2 (HER2) monoclonal antibody trastuzumab enhances cytolytic activity of class I-restricted HER2-specific T lymphocytes against HER2-overexpressing tumor cells. Cancer Res. 2002, 62, 2244–2247. [Google Scholar] [PubMed]
  39. Park, S.; Jiang, Z.; Mortenson, E.D.; Deng, L.; Radkevich-Brown, O.; Yang, X.; Sattar, H.; Wang, Y.; Brown, N.K.; Greene, M.; et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell 2010, 18, 160–170. [Google Scholar] [CrossRef] [Green Version]
  40. Janjigian, Y.Y.; Maron, S.B.; Chatila, W.K.; Millang, B.; Chavan, S.S.; Alterman, C.; Chou, J.F.; Segal, M.F.; Simmons, M.Z.; Momtaz, P.; 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, 821–831. [Google Scholar] [CrossRef]
  41. Catenacci, D.V.; Kang, Y.K.; Park, H.; Uronis, H.E.; Lee, K.W.; Ng, M.C.; Enzinger, P.C.; Park, S.H.; Gold, P.J.; Lacy, J.; et al. Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22–05): A single-arm, phase 1b–2 trial. Lancet Oncol. 2020, 21, 1066–1076. [Google Scholar] [CrossRef]
  42. Catenacci, D.V.; Rosales, M.; Chung, H.C.; Yoon, H.H.; Shen, L.; Moehler, M.; Kang, Y.K. MAHOGANY: Margetuximab combination in HER2+ unresectable/metastatic gastric/gastroesophageal junction adenocarcinoma. Future Oncol. 2021, 17, 1155–1164. [Google Scholar] [CrossRef]
  43. Weisser, N.; Wickman, G.; Davies, R.; Rowse, G. Abstract 31: Preclinical development of a novel biparatopic HER2 antibody with activity in low to high HER2 expressing cancers. Cancer Res. 2017, 77, 31. [Google Scholar] [CrossRef]
  44. Murthy, R.K.; Loi, S.; Okines, A.; Paplomata, E.; Hamilton, E.; Hurvitz, S.A.; Lin, N.U.; Borges, V.; Abramson, V.; Anders, C.; et al. Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N. Engl. J. Med. 2020, 382, 597–609. [Google Scholar] [CrossRef] [PubMed]
  45. Hamzehlou, S.; Momeny, M.; Zandi, Z.; Kashani, B.; Yousefi, H.; Dehpour, A.R.; Tavakkoly-Bazzaz, J.; Ghaffari, S.H. Anti-tumor activity of neratinib, a pan-HER inhibitor, in gastric adenocarcinoma cells. Eur. J. Pharmacol. 2019, 863, 172705. [Google Scholar] [CrossRef] [PubMed]
  46. Bergers, G.; Benjamin, L.E. Tumorigenesis and the angiogenic switch. Nat. Rev. Cancer 2003, 3, 401–410. [Google Scholar] [CrossRef] [PubMed]
  47. Carmeliet, P. Angiogenesis in health and disease. Nat. Med. 2003, 9, 653–660. [Google Scholar] [CrossRef]
  48. Ferrara, N.; Gerber, H.P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676. [Google Scholar] [CrossRef]
  49. Ferrara, N.; Kerbel, R.S. Angiogenesis as a therapeutic target. Nature 2005, 438, 967–974. [Google Scholar] [CrossRef] [PubMed]
  50. Ribatti, D.; Solimando, A.G.; Pezzella, F. The Anti-VEGF(R) Drug Discovery Legacy: Improving Attrition Rates by Breaking the Vicious Cycle of Angiogenesis in Cancer. Cancers 2021, 13, 3433. [Google Scholar] [CrossRef]
  51. Ohtsu, A.; Shah, M.A.; Van Cutsem, E.; Rha, S.Y.; Sawaki, A.; Park, S.R.; Lim, H.Y.; Yamada, Y.; Wu, J.; Langer, B.; 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. [Google Scholar] [CrossRef] [PubMed]
  52. Shen, L.; Li, J.; Xu, J.; Pan, H.; Dai, G.; Qin, S.; Wang, L.; Wang, J.; Yang, Z.; Shu, Y.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Xu, R.h.; Zhang, D.; Shen, L.; LI, J.; Huang, J.; Gong, J.; Guo, W.; Zhang, Y.; Fan, S.; Li, K.; et al. A Phase I/II trial of fruquintinib in combination with paclitaxel for second-line treatment in patients with advanced gastric cancer. J. Clin. Oncol. 2017, 35, 128. [Google Scholar] [CrossRef]
  54. Li, J.; Qin, S.; Xu, J.; Xiong, J.; Wu, C.; Bai, Y.; Liu, W.; Tong, J.; Liu, Y.; Xu, R.; 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. [Google Scholar] [CrossRef] [PubMed]
  55. Kang, Y.K.; Kang, W.K.; Di Bartolomeo, M.; Chau, I.; Yoon, H.H.; Cascinu, S.; Ryu, M.H.; Kim, J.G.; Lee, K.W.; Oh, S.C.; et al. Randomized phase III ANGEL study of rivoceranib (apatinib) + best supportive care (BSC) vs placebo + BSC in patients with advanced/metastatic gastric cancer who failed ≥2 prior chemotherapy regimens. Ann. Oncol. 2019, 30, v877–v878. [Google Scholar] [CrossRef]
  56. Fuchs, C.S.; Tabernero, J.; Tomasek, J.; Chau, I.; Melichar, B.; Safran, H.; Tehfe, M.A.; Filip, D.; Topuzov, E.; Schlittler, L.; et al. Biomarker analyses in REGARD gastric/GEJ carcinoma patients treated with VEGFR2-targeted antibody ramucirumab. Br. J. Cancer 2016, 115, 974–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Hacker, U.T.; Escalona-Espinosa, L.; Consalvo, N.; Goede, V.; Schiffmann, L.; Scherer, S.J.; Hedge, P.; Van Cutsem, E.; Coutelle, O.; Buning, H. Evaluation of Angiopoietin-2 as a biomarker in gastric cancer: Results from the randomised phase III AVAGAST trial. Br. J. Cancer 2016, 114, 855–862. [Google Scholar] [CrossRef] [Green Version]
  58. Van Cutsem, E.; de Haas, S.; Kang, Y.K.; Ohtsu, A.; Tebbutt, N.C.; Ming Xu, J.; Peng Yong, W.; Langer, B.; Delmar, P.; Scherer, S.J.; et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: A biomarker evaluation from the AVAGAST randomized phase III trial. J. Clin. Oncol. 2012, 30, 2119–2127. [Google Scholar] [CrossRef] [PubMed]
  59. Van Cutsem, E.; Muro, K.; Cunningham, D.; Bodoky, G.; Sobrero, A.; Cascinu, S.; Ajani, J.; Oh, S.C.; Al-Batran, S.E.; Wainberg, Z.A.; et al. Biomarker analyses of second-line ramucirumab in patients with advanced gastric cancer from RAINBOW, a global, randomized, double-blind, phase 3 study. Eur. J. Cancer 2020, 127, 150–157. [Google Scholar] [CrossRef] [Green Version]
  60. Fuchs, C.S.; Doi, T.; Jang, R.W.; Muro, K.; Satoh, T.; Machado, M.; Sun, W.; Jalal, S.I.; Shah, M.A.; Metges, J.P.; 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. [Google Scholar] [CrossRef]
  61. Fuchs, C.S.; Özgüroğlu, M.; Bang, Y.J.; Bartolomeo, M.D.; Mandalà, M.; Ryu, M.h.; Vivaldi, C.; Olesinski, T.; Caglevic, C.; Chung, H.C.; 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, 4512. [Google Scholar] [CrossRef]
  62. Moehler, M.; Dvorkin, M.; Boku, N.; Özgüroğlu, M.; Ryu, M.H.; Muntean, A.S.; Lonardi, S.; Nechaeva, M.; Bragagnoli, A.C.; Coşkun, H.S.; 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. [Google Scholar] [CrossRef] [PubMed]
  63. Shitara, K.; Özgüroğlu, M.; Bang, Y.J.; Bartolomeo, M.D.; Mandalà, M.; Ryu, M.h.; Vivaldi, C.; Olesinski, T.; Chung, H.C.; Muro, K.; et al. The association of tissue tumor mutational burden (tTMB) using the Foundation Medicine genomic platform with efficacy of pembrolizumab versus paclitaxel in patients (pts) with gastric cancer (GC) from KEYNOTE-061. J. Clin. Oncol. 2020, 38, 4537. [Google Scholar] [CrossRef]
  64. Boku, N.; Ryu, M.H.; Kato, K.; Chung, H.C.; Minashi, K.; Lee, K.W.; Cho, H.; Kang, W.K.; Komatsu, Y.; Tsuda, M.; 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, 250–258. [Google Scholar] [CrossRef] [Green Version]
  65. Boku, N.; Ryu, M.H.; Oh, D.Y.; Oh, S.C.; Chung, H.C.; Lee, K.W.; Omori, T.; Shitara, K.; Sakuramoto, S.; Chung, I.J.; 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, S1192. [Google Scholar] [CrossRef]
  66. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H.; 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. [Google Scholar] [CrossRef]
  67. Janjigian, Y.Y.; Shitara, K.; Moehler, M.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Campos Bragagnoli, A.; 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. [Google Scholar] [CrossRef]
  68. Kato, K.; Sun, J.M.; Shah, M.A.; Enzinger, P.C.; Adenis, A.; Doi, T.; Kojima, T.; Metges, J.P.; Li, Z.; Kim, S.B.; et al. LBA8_PR Pembrolizumab plus chemotherapy versus chemotherapy as first-line therapy in patients with advanced esophageal cancer: The phase 3 KEYNOTE-590 study. Ann. Oncol. 2020, 31, S1192–S1193. [Google Scholar] [CrossRef]
  69. Kim, S.T.; Cristescu, R.; Bass, A.J.; Kim, K.M.; Odegaard, J.I.; Kim, K.; Liu, X.Q.; Sher, X.; Jung, H.; Lee, M.; et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat. Med. 2018, 24, 1449–1458. [Google Scholar] [CrossRef] [PubMed]
  70. Nakano, H.; Saito, M.; Nakajima, S.; Saito, K.; Nakayama, Y.; Kase, K.; Yamada, L.; Kanke, Y.; Hanayama, H.; Onozawa, H.; et al. PD-L1 overexpression in EBV-positive gastric cancer is caused by unique genomic or epigenomic mechanisms. Sci. Rep. 2021, 11, 1982. [Google Scholar] [CrossRef]
  71. Roemer, M.G.; Advani, R.H.; Ligon, A.H.; Natkunam, Y.; Redd, R.A.; Homer, H.; Connelly, C.F.; Sun, H.H.; Daadi, S.E.; Freeman, G.J.; et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J. Clin. Oncol. 2016, 34, 2690–2697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 2015, 372, 311–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Borcoman, E.; De La Rochere, P.; Richer, W.; Vacher, S.; Chemlali, W.; Krucker, C.; Sirab, N.; Radvanyi, F.; Allory, Y.; Pignot, G.; et al. Inhibition of PI3K pathway increases immune infiltrate in muscle-invasive bladder cancer. Oncoimmunology 2019, 8, e1581556. [Google Scholar] [CrossRef] [PubMed]
  74. Fakih, M.; Raghav, K.P.; Chang, D.Z.; Bendell, J.C.; Larson, T.; Cohn, A.L.; Huyck, T.K.; Cosgrove, D.; Fiorillo, J.A.; Garbo, L.E.; et al. Single-arm, phase 2 study of regorafenib plus nivolumab in patients with mismatch repair-proficient (pMMR)/microsatellite stable (MSS) colorectal cancer (CRC). J. Clin. Oncol. 2021, 39, 3560. [Google Scholar] [CrossRef]
  75. Fukuoka, S.; Hara, H.; Takahashi, N.; Kojima, T.; Kawazoe, A.; Asayama, M.; Yoshii, T.; Kotani, D.; Tamura, H.; Mikamoto, Y.; 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, 2053–2061. [Google Scholar] [CrossRef]
  76. Chung, H.C.; Lwin, Z.; Gomez-Roca, C.; Longo, F.; Yanez, E.; Alvarez, E.C.; Graham, D.; Doherty, M.; Cassier, P.; Lopez, J.S.; et al. LEAP-005: A phase II multicohort study of lenvatinib plus pembrolizumab in patients with previously treated selected solid tumors—Results from the gastric cancer cohort. J. Clin. Oncol. 2021, 39, 230. [Google Scholar] [CrossRef]
  77. Lordick, F.; Kang, Y.K.; Chung, H.C.; Salman, P.; Oh, S.C.; Bodoky, G.; Kurteva, G.; Volovat, C.; Moiseyenko, V.M.; Gorbunova, V.; 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. [Google Scholar] [CrossRef]
  78. Waddell, T.; Chau, I.; Cunningham, D.; Gonzalez, D.; Okines, A.F.; Wotherspoon, A.; Saffery, C.; Middleton, G.; Wadsley, J.; Ferry, 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. [Google Scholar] [CrossRef] [Green Version]
  79. Dutton, S.J.; Ferry, D.R.; Blazeby, J.M.; Abbas, H.; Dahle-Smith, A.; Mansoor, W.; Thompson, J.; Harrison, M.; Chatterjee, A.; Falk, S.; 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. [Google Scholar] [CrossRef]
  80. Lordick, F.; Kang, Y.K.; Salman, P.; Oh, S.C.; Bodoky, G.; Kurteva, G.P.; Volovat, C.D.; Moiseyenko, V.; Sawaki, A.; Park, J.O.; et al. Clinical outcome according to tumor HER2 status and EGFR expression in advanced gastric cancer patients from the EXPAND study. J. Clin. Oncol. 2013, 31, 4021. [Google Scholar] [CrossRef]
  81. Maron, S.B.; Alpert, L.; Kwak, H.A.; Lomnicki, S.; Chase, L.; Xu, D.; O’Day, E.; Nagy, R.J.; Lanman, R.B.; Cecchi, F.; et al. Targeted Therapies for Targeted Populations: Anti-EGFR Treatment for EGFR-Amplified Gastroesophageal Adenocarcinoma. Cancer Discov. 2018, 8, 696–713. [Google Scholar] [CrossRef] [Green Version]
  82. Smyth, E.C.; Vlachogiannis, G.; Hedayat, S.; Harbery, A.; Hulkki-Wilson, S.; Salati, M.; Kouvelakis, K.; Fernandez-Mateos, J.; Cresswell, G.D.; Fontana, E.; et al. EGFR amplification and outcome in a randomised phase III trial of chemotherapy alone or chemotherapy plus panitumumab for advanced gastro-oesophageal cancers. Gut 2021, 70, 1632–1641. [Google Scholar] [CrossRef]
  83. El Darsa, H.; El Sayed, R.; Abdel-Rahman, O. MET Inhibitors for the Treatment of Gastric Cancer: What’s Their Potential? J. Exp. Pharmacol. 2020, 12, 349–361. [Google Scholar] [CrossRef] [PubMed]
  84. Sanchez-Vega, F.; Hechtman, J.F.; Castel, P.; Ku, G.Y.; Tuvy, Y.; Won, H.; Fong, C.J.; Bouvier, N.; Nanjangud, G.J.; Soong, J.; et al. EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2-Amplified Esophagogastric Cancer. Cancer Discov. 2019, 9, 199–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Peng, Z.; Zhu, Y.; Wang, Q.; Gao, J.; Li, Y.; Li, Y.; Ge, S.; Shen, L. Prognostic significance of MET amplification and expression in gastric cancer: A systematic review with meta-analysis. PLoS ONE 2014, 9, e84502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lee, H.E.; Kim, M.A.; Lee, H.S.; Jung, E.J.; Yang, H.K.; Lee, B.L.; Bang, Y.J.; Kim, W.H. MET in gastric carcinomas: Comparison between protein expression and gene copy number and impact on clinical outcome. Br. J. Cancer 2012, 107, 325–333. [Google Scholar] [CrossRef] [Green Version]
  87. Iveson, T.; Donehower, R.C.; Davidenko, I.; Tjulandin, S.; Deptala, A.; Harrison, M.; Nirni, S.; Lakshmaiah, K.; Thomas, A.; Jiang, Y.; et al. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: An open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study. Lancet Oncol. 2014, 15, 1007–1018. [Google Scholar] [CrossRef]
  88. Catenacci, D.V.; Tebbutt, N.C.; Davidenko, I.; Murad, A.M.; Al-Batran, S.E.; Ilson, D.H.; Tjulandin, S.; Gotovkin, E.; Karaszewska, B.; Bondarenko, 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. [Google Scholar] [CrossRef]
  89. Doi, T.; Kang, Y.K.; Muro, K.; Jiang, Y.; Jain, R.K.; Lizambri, R. A phase 3, multicenter, randomized, double-blind, placebo-controlled study of rilotumumab in combination with cisplatin and capecitabine (CX) as first-line therapy for Asian patients (pts) with advanced MET-positive gastric or gastroesophageal junction (G/GEJ) adenocarcinoma: The RILOMET-2 trial. J. Clin. Oncol. 2015, 33, TPS226. [Google Scholar] [CrossRef]
  90. Shah, M.A.; Bang, Y.J.; Lordick, F.; Alsina, M.; Chen, M.; Hack, S.P.; Bruey, J.M.; Smith, D.; McCaffery, I.; Shames, D.S.; 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. [Google Scholar] [CrossRef] [PubMed]
  91. Lee, J.; Kim, S.T.; Kim, K.; Lee, H.; Kozarewa, I.; Mortimer, P.G.; Odegaard, J.I.; Harrington, E.A.; Lee, J.; Lee, T.; et al. Tumor Genomic Profiling Guides Patients with Metastatic Gastric Cancer to Targeted Treatment: The VIKTORY Umbrella Trial. Cancer Discov. 2019, 9, 1388–1405. [Google Scholar] [CrossRef] [Green Version]
  92. Frigault, M.M.; Markovets, A.; Nuttall, B.; Kim, K.M.; Park, S.H.; Gangolli, E.A.; Mortimer, P.G.; Hollingsworth, S.J.; Hong, J.Y.; Kim, K.; et al. Mechanisms of Acquired Resistance to Savolitinib, a Selective MET Inhibitor in MET-Amplified Gastric Cancer. JCO Precis Oncol. 2020, 4, PO.19.00386. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, Y.; Yang, Q.; Zeng, X.; Wang, M.; Dong, S.; Yang, B.; Tu, X.; Wei, T.; Xie, W.; Zhang, C.; et al. MET amplification attenuates lung tumor response to immunotherapy by inhibiting STING. Cancer Discov. 2021, 11, 1–12. [Google Scholar] [CrossRef]
  94. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef]
  95. Wolf, J.; Seto, T.; Han, J.Y.; Reguart, N.; Garon, E.B.; Groen, H.J.; Tan, D.S.; Hida, T.; de Jonge, M.; Orlov, S.V.; et al. Capmatinib in MET Exon 14-Mutated or MET-Amplified Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef]
  96. Helsten, T.; Elkin, S.; Arthur, E.; Tomson, B.N.; Carter, J.; Kurzrock, R. The FGFR Landscape in Cancer: Analysis of 4,853 Tumors by Next-Generation Sequencing. Clin. Cancer Res. 2016, 22, 259–267. [Google Scholar] [CrossRef] [Green Version]
  97. Hur, J.Y.; Chao, J.; Kim, K.; Kim, S.T.; Kim, K.M.; Klempner, S.J.; Lee, J. High-level FGFR2 amplification is associated with poor prognosis and Lower response to chemotherapy in gastric cancers. Pathol. Res. Pract. 2020, 216, 152878. [Google Scholar] [CrossRef]
  98. Loriot, Y.; Necchi, A.; Park, S.H.; Garcia-Donas, J.; Huddart, R.; Burgess, E.; Fleming, M.; Rezazadeh, A.; Mellado, B.; Varlamov, S.; et al. Erdafitinib in Locally Advanced or Metastatic Urothelial Carcinoma. N. Engl. J. Med. 2019, 381, 338–348. [Google Scholar] [CrossRef]
  99. Abou-Alfa, G.K.; Sahai, V.; Hollebecque, A.; Vaccaro, G.; Melisi, D.; Al-Rajabi, R.; Paulson, A.S.; Borad, M.J.; Gallinson, D.; Murphy, A.G.; et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: A multicentre, open-label, phase 2 study. Lancet Oncol. 2020, 21, 671–684. [Google Scholar] [CrossRef]
  100. Xie, L.; Su, X.; Zhang, L.; Yin, X.; Tang, L.; Zhang, X.; Xu, Y.; Gao, Z.; Liu, K.; Zhou, M.; et al. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin. Cancer Res. 2013, 19, 2572–2583. [Google Scholar] [CrossRef] [Green Version]
  101. Jang, J.; Kim, H.K.; Bang, H.; Kim, S.T.; Kim, S.Y.; Park, S.H.; Lim, H.Y.; Kang, W.K.; Lee, J.; Kim, K.M. Antitumor Effect of AZD4547 in a Fibroblast Growth Factor Receptor 2–Amplified Gastric Cancer Patient–Derived Cell Model. Transl. Oncol. 2017, 10, 469–475. [Google Scholar] [CrossRef]
  102. Van Cutsem, E.; Bang, Y.J.; Mansoor, W.; Petty, R.D.; Chao, Y.; Cunningham, D.; Ferry, D.R.; Smith, N.R.; Frewer, P.; Ratnayake, J.; 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. [Google Scholar] [CrossRef] [PubMed]
  103. Sootome, H.; Fujita, H.; Ito, K.; Ochiiwa, H.; Fujioka, Y.; Ito, K.; Miura, A.; Sagara, T.; Ito, S.; Ohsawa, H.; et al. Futibatinib Is a Novel Irreversible FGFR 1-4 Inhibitor That Shows Selective Antitumor Activity against FGFR-Deregulated Tumors. Cancer Res. 2020, 80, 4986–4997. [Google Scholar] [CrossRef]
  104. Goyal, L.; Shi, L.; Liu, L.Y.; Fece de la Cruz, F.; Lennerz, J.K.; Raghavan, S.; Leschiner, I.; Elagina, L.; Siravegna, G.; Ng, R.W.; et al. TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion-Positive Intrahepatic Cholangiocarcinoma. Cancer Discov. 2019, 9, 1064–1079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Hollebecque, A.; Doi, T.; Saavedra, O.; Takahashi, O.; He, H.; Benhadji, K.A.; Shitara, K. A phase II study of futibatinib (TAS-120) in patients (pts) with advanced (adv) solid tumors harboring fibroblast growth factor receptor (FGFR) genomic aberrations. J. Clin. Oncol. 2020, 38, TPS470. [Google Scholar] [CrossRef]
  106. Catenacci, D.V.; Tesfaye, A.; Tejani, M.; Cheung, E.; Eisenberg, P.; Scott, A.J.; Eng, C.; Hnatyszyn, J.; Marina, N.; Powers, J.; et al. Bemarituzumab with modified FOLFOX6 for advanced FGFR2-positive gastroesophageal cancer: FIGHT Phase III study design. Future Oncol. 2019, 15, 2073–2082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Wainberg, Z.A.; Enzinger, P.C.; Kang, Y.K.; Yamaguchi, K.; Qin, S.; Lee, K.W.; Oh, S.C.; Li, J.; Turk, H.M.; Teixeira, A.C.; et al. Randomized double-blind placebo-controlled phase 2 study of bemarituzumab combined with modified FOLFOX6 (mFOLFOX6) in first-line (1L) treatment of advanced gastric/gastroesophageal junction adenocarcinoma (FIGHT). J. Clin. Oncol. 2021, 39, 160. [Google Scholar] [CrossRef]
  108. Cha, Y.; Kim, H.P.; Lim, Y.; Han, S.W.; Song, S.H.; Kim, T.Y. FGFR2 amplification is predictive of sensitivity to regorafenib in gastric and colorectal cancers in vitro. Mol. Oncol. 2018, 12, 993–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Nakayama, I.; Shinozaki, E.; Sakata, S.; Yamamoto, N.; Fujisaki, J.; Muramatsu, Y.; Hirota, T.; Takeuchi, K.; Takahashi, S.; Yamaguchi, K.; et al. Enrichment of CLDN18-ARHGAP fusion gene in gastric cancers in young adults. Cancer Sci. 2019, 110, 1352–1363. [Google Scholar] [CrossRef] [Green Version]
  110. Dottermusch, M.; Kruger, S.; Behrens, H.M.; Halske, C.; Rocken, C. Expression of the potential therapeutic target claudin-18.2 is frequently decreased in gastric cancer: Results from a large Caucasian cohort study. Virchows Arch. 2019, 475, 563–571. [Google Scholar] [CrossRef] [Green Version]
  111. Sahin, U.; Tureci, O.; Manikhas, G.; Lordick, F.; Rusyn, A.; Vynnychenko, I.; Dudov, A.; Bazin, I.; Bondarenko, I.; Melichar, B.; 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, 609–619. [Google Scholar] [CrossRef] [PubMed]
  112. Tureci, O.; Sahin, U.; Schulze-Bergkamen, H.; Zvirbule, Z.; Lordick, F.; Koeberle, D.; Thuss-Patience, P.; Ettrich, T.; Arnold, D.; Bassermann, F.; 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, 1487–1495. [Google Scholar] [CrossRef] [Green Version]
  113. Van Putten, J.P.M.; Strijbis, K. Transmembrane Mucins: Signaling Receptors at the Intersection of Inflammation and Cancer. J. Innate Immun. 2017, 9, 281–299. [Google Scholar] [CrossRef] [PubMed]
  114. Bailis, J.M.; Lutterbuese, P.; Thomas, O.; Locher, K.; Harrold, J.; Boyle, M.; Wahl, J.; Li, S.; Sternjak, A.; Henn, A.; et al. Abstract 3364: Preclinical evaluation of BiTE® immune therapy targeting MUC17 or CLDN18.2 for gastric cancer. Cancer Res. 2020, 80, 3364. [Google Scholar] [CrossRef]
  115. Coussens, L.M.; Fingleton, B.; Matrisian, L.M. Matrix metalloproteinase inhibitors and cancer: Trials and tribulations. Science 2002, 295, 2387–2392. [Google Scholar] [CrossRef] [PubMed]
  116. Lian, P.L.; Liu, Z.; Yang, G.Y.; Zhao, R.; Zhang, Z.Y.; Chen, Y.G.; Zhuang, Z.N.; Xu, K.S. Integrin alphavbeta6 and matrix metalloproteinase 9 correlate with survival in gastric cancer. World J. Gastroenterol. 2016, 22, 3852–3859. [Google Scholar] [CrossRef]
  117. Shah, M.A.; Ruiz, E.P.; Bodoky, G.; Starodub, A.; Cunningham, D.; Yip, D.; Wainberg, Z.A.; Bendell, J.C.; Thai, D.; Bhargava, P.; et al. A phase III, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of andecaliximab combined with mFOLFOX6 as first-line treatment in patients with advanced gastric or gastroesophageal junction adenocarcinoma (GAMMA-1). J. Clin. Oncol. 2019, 37, 4. [Google Scholar] [CrossRef]
  118. Shah, M.A.; Starodub, A.; Sharma, S.; Berlin, J.; Patel, M.; Wainberg, Z.A.; Chaves, J.; Gordon, M.; Windsor, K.; Brachmann, C.B.; et al. Andecaliximab/GS-5745 Alone and Combined with mFOLFOX6 in Advanced Gastric and Gastroesophageal Junction Adenocarcinoma: Results from a Phase I Study. Clin. Cancer Res. 2018, 24, 3829–3837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. De Bono, J.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2091–2102. [Google Scholar] [CrossRef]
  120. Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.O.; Hochhauser, D.; Arnold, D.; Oh, D.Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef]
  121. Hussain, M.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Survival with Olaparib in Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 383, 2345–2357. [Google Scholar] [CrossRef] [PubMed]
  122. Ledermann, J.A.; Harter, P.; Gourley, C.; Friedlander, M.; Vergote, I.; Rustin, G.; Scott, C.; Meier, W.; Shapira-Frommer, R.; Safra, T.; et al. Overall survival in patients with platinum-sensitive recurrent serous ovarian cancer receiving olaparib maintenance monotherapy: An updated analysis from a randomised, placebo-controlled, double-blind, phase 2 trial. Lancet Oncol. 2016, 17, 1579–1589. [Google Scholar] [CrossRef]
  123. Moore, K.; Colombo, N.; Scambia, G.; Kim, B.G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; Sonke, G.S.; et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2018, 379, 2495–2505. [Google Scholar] [CrossRef]
  124. Robson, M.; Im, S.A.; Senkus, E.; Xu, B.; Domchek, S.M.; Masuda, N.; Delaloge, S.; Li, W.; Tung, N.; Armstrong, A.; et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N. Engl. J. Med. 2017, 377, 523–533. [Google Scholar] [CrossRef] [PubMed]
  125. Ladan, M.M.; van Gent, D.C.; Jager, A. Homologous Recombination Deficiency Testing for BRCA-Like Tumors: The Road to Clinical Validation. Cancers 2021, 13, 1004. [Google Scholar] [CrossRef]
  126. Principe, D.R.; Narbutis, M.; Koch, R.; Rana, A. Frequency and prognostic value of mutations associated with the homologous recombination DNA repair pathway in a large pan cancer cohort. Sci. Rep. 2020, 10, 20223. [Google Scholar] [CrossRef]
  127. Alexandrov, L.B.; Nik-Zainal, S.; Siu, H.C.; Leung, S.Y.; Stratton, M.R. A mutational signature in gastric cancer suggests therapeutic strategies. Nat. Commun. 2015, 6, 8683. [Google Scholar] [CrossRef] [Green Version]
  128. Smyth, E.C.; Cafferkey, C.; Loehr, A.; Waddell, T.; Begum, R.; Peckitt, C.; Harding, T.C.; Nguyen, M.; Okines, A.F.; Raponi, M.; et al. Genomic loss of heterozygosity and survival in the REAL3 trial. Oncotarget 2018, 9, 36654–36665. [Google Scholar] [CrossRef]
  129. Sahasrabudhe, R.; Lott, P.; Bohorquez, M.; Toal, T.; Estrada, A.P.; Suarez, J.J.; Brea-Fernandez, A.; Cameselle-Teijeiro, J.; Pinto, C.; Ramos, I.; et al. Germline Mutations in PALB2, BRCA1, and RAD51C, Which Regulate DNA Recombination Repair, in Patients with Gastric Cancer. Gastroenterology 2017, 152, 983–986. [Google Scholar] [CrossRef] [Green Version]
  130. Bang, Y.J.; Im, S.A.; Lee, K.W.; Cho, J.Y.; Song, E.K.; Lee, K.H.; Kim, Y.H.; Park, J.O.; Chun, H.G.; Zang, D.Y.; 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. [Google Scholar] [CrossRef]
  131. Bang, Y.J.; Xu, R.H.; Chin, K.; Lee, K.W.; Park, S.H.; Rha, S.Y.; Shen, L.; Qin, S.; Xu, N.; Im, S.A.; 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. [Google Scholar] [CrossRef]
  132. Liu, Y.Z.; Hodgson, D.; Locker, G.; Lai, Z.; Balcerzak, D.; Sharpe, A.; Barrett, J.C.; Orr, M.; Gutjahr, T.S.; Dougherty, B.; et al. Olaparib plus paclitaxel sensitivity in biomarker subgroups of gastric cancer. Ann. Oncol. 2018, 29, viii25–viii26. [Google Scholar] [CrossRef]
  133. Wang, Y.; Zheng, K.; Huang, Y.; Xiong, H.; Su, J.; Chen, R.; Zou, Y. PARP inhibitors in gastric cancer: Beacon of hope. J. Exp. Clin. Cancer Res. 2021, 40, 211. [Google Scholar] [CrossRef]
  134. Bang, Y.J.; Kaufman, B.; Geva, R.; Stemmer, S.M.; Hong, S.H.; Lee, J.S.; Domchek, S.M.; Lanasa, M.C.; Tang, M.; Gresty, C.; et al. An open-label, phase II basket study of olaparib and durvalumab (MEDIOLA): Results in patients with relapsed gastric cancer. J. Clin. Oncol. 2019, 37, 140. [Google Scholar] [CrossRef]
  135. Zhang, H.; Schaefer, A.; Wang, Y.; Hodge, R.G.; Blake, D.R.; Diehl, J.N.; Papageorge, A.G.; Stachler, M.D.; Liao, J.; Zhou, J.; et al. Gain-of-Function RHOA Mutations Promote Focal Adhesion Kinase Activation and Dependency in Diffuse Gastric Cancer. Cancer Discov. 2020, 10, 288–305. [Google Scholar] [CrossRef]
  136. Sulzmaier, F.J.; Jean, C.; Schlaepfer, D.D. FAK in cancer: Mechanistic findings and clinical applications. Nat. Rev. Cancer 2014, 14, 598–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Bajrami, I.; Marlow, R.; van de Ven, M.; Brough, R.; Pemberton, H.N.; Frankum, J.; Song, F.; Rafiq, R.; Konde, A.; Krastev, D.B.; et al. E-Cadherin/ROS1 Inhibitor Synthetic Lethality in Breast Cancer. Cancer Discov. 2018, 8, 498–515. [Google Scholar] [CrossRef] [Green Version]
  138. Arrowsmith, J.; Miller, P. Trial watch: Phase II and phase III attrition rates 2011-2012. Nat. Rev. Drug Discov. 2013, 12, 569. [Google Scholar] [CrossRef]
  139. Larson, K.L.; Huang, B.; Weiss, H.L.; Hull, P.; Westgate, P.M.; Miller, R.W.; Arnold, S.M.; Kolesar, J.M. Clinical Outcomes of Molecular Tumor Boards: A Systematic Review. JCO Precis. Oncol. 2021, 5, PO.20.00495. [Google Scholar] [CrossRef] [PubMed]
  140. Klinghammer, K.; Walther, W.; Hoffmann, J. Choosing wisely—Preclinical test models in the era of precision medicine. Cancer Treat Rev. 2017, 55, 36–45. [Google Scholar] [CrossRef]
  141. Bhimani, J.; Ball, K.; Stebbing, J. Patient-derived xenograft models-the future of personalised cancer treatment. Br. J. Cancer 2020, 122, 601–602. [Google Scholar] [CrossRef]
  142. Powley, I.R.; Patel, M.; Miles, G.; Pringle, H.; Howells, L.; Thomas, A.; Kettleborough, C.; Bryans, J.; Hammonds, T.; MacFarlane, M.; et al. Patient-derived explants (PDEs) as a powerful preclinical platform for anti-cancer drug and biomarker discovery. Br. J. Cancer 2020, 122, 735–744. [Google Scholar] [CrossRef] [Green Version]
  143. Wensink, G.E.; Elias, S.G.; Mullenders, J.; Koopman, M.; Boj, S.F.; Kranenburg, O.W.; Roodhart, J.M. Patient-derived organoids as a predictive biomarker for treatment response in cancer patients. NPJ Precis. Oncol. 2021, 5, 30. [Google Scholar] [CrossRef] [PubMed]
  144. Seidlitz, T.; Merker, S.R.; Rothe, A.; Zakrzewski, F.; von Neubeck, C.; Grutzmann, K.; Sommer, U.; Schweitzer, C.; Scholch, S.; Uhlemann, H.; et al. Human gastric cancer modelling using organoids. Gut 2019, 68, 207–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Steele, N.G.; Chakrabarti, J.; Wang, J.; Biesiada, J.; Holokai, L.; Chang, J.; Nowacki, L.M.; Hawkins, J.; Mahe, M.; Sundaram, N.; et al. An Organoid-Based Preclinical Model of Human Gastric Cancer. Cell. Mol. Gastroenterol. Hepatol. 2019, 7, 161–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Yan, H.H.; Siu, H.C.; Law, S.; Ho, S.L.; Yue, S.S.; Tsui, W.Y.; Chan, D.; Chan, A.S.; Ma, S.; Lam, K.O.; et al. A Comprehensive Human Gastric Cancer Organoid Biobank Captures Tumor Subtype Heterogeneity and Enables Therapeutic Screening. Cell Stem. Cell 2018, 23, 882–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Koerfer, J.; Kallendrusch, S.; Merz, F.; Wittekind, C.; Kubick, C.; Kassahun, W.T.; Schumacher, G.; Moebius, C.; Gassler, N.; Schopow, N.; et al. Organotypic slice cultures of human gastric and esophagogastric junction cancer. Cancer Med. 2016, 5, 1444–1453. [Google Scholar] [CrossRef]
  148. Husstegge, M.; Hoang, N.A.; Rebstock, J.; Monecke, A.; Gockel, I.; Weimann, A.; Schumacher, G.; Bechmann, I.; Lordick, F.; Kallendrusch, S.; et al. PD-1 inhibition in patient derived tissue cultures of human gastric and gastroesophageal adenocarcinoma. Oncoimmunology 2021, 10, 1960729. [Google Scholar] [CrossRef]
  149. Choi, Y.Y.; Lee, J.E.; Kim, H.; Sim, M.H.; Kim, K.K.; Lee, G.; Kim, H.I.; An, J.Y.; Hyung, W.J.; Kim, C.B.; et al. Establishment and characterisation of patient-derived xenografts as paraclinical models for gastric cancer. Sci. Rep. 2016, 6, 22172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Peille, A.L.; Vuaroqueaux, V.; Wong, S.S.; Ting, J.; Klingner, K.; Zeitouni, B.; Landesfeind, M.; Kim, W.H.; Lee, H.J.; Kong, S.H.; et al. Evaluation of molecular subtypes and clonal selection during establishment of patient-derived tumor xenografts from gastric adenocarcinoma. Commun. Biol. 2020, 3, 367. [Google Scholar] [CrossRef]
  151. Reddavid, R.; Corso, S.; Moya-Rull, D.; Giordano, S.; Degiuli, M. Patient-Derived Orthotopic Xenograft models in gastric cancer: A systematic review. Updates Surg. 2020, 72, 951–966. [Google Scholar] [CrossRef]
  152. Wang, H.; Lu, J.; Tang, J.; Chen, S.; He, K.; Jiang, X.; Jiang, W.; Teng, L. Establishment of patient-derived gastric cancer xenografts: A useful tool for preclinical evaluation of targeted therapies involving alterations in HER-2, MET and FGFR2 signaling pathways. BMC Cancer 2017, 17, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Catenacci, D.V.; Moya, S.; Lomnicki, S.; Chase, L.M.; Peterson, B.F.; Reizine, N.; Alpert, L.; Setia, N.; Xiao, S.Y.; Hart, J.; et al. Personalized Antibodies for Gastroesophageal Adenocarcinoma (PANGEA): A Phase II Study Evaluating an Individualized Treatment Strategy for Metastatic Disease. Cancer Discov. 2021, 11, 308–325. [Google Scholar] [CrossRef] [PubMed]
  154. Nakamura, Y.; Taniguchi, H.; Ikeda, M.; Bando, H.; Kato, K.; Morizane, C.; Esaki, T.; Komatsu, Y.; Kawamoto, Y.; Takahashi, N.; et al. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat. Med. 2020, 26, 1859–1864. [Google Scholar] [CrossRef] [PubMed]
  155. Nakamura, Y.; Kawazoe, A.; Lordick, F.; Janjigian, Y.Y.; Shitara, K. Biomarker-targeted therapies for advanced-stage gastric and gastro-oesophageal junction cancers: An emerging paradigm. Nat. Rev. Clin. Oncol 2021, 18, 473–487. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular classification of gastric cancer into distinct subtypes according to TCGA and ACRG.
Figure 1. Molecular classification of gastric cancer into distinct subtypes according to TCGA and ACRG.
Cancers 13 05216 g001
Table 1. Selected pivotal clinical trials in gastric and esophagogastric junction cancers.
Table 1. Selected pivotal clinical trials in gastric and esophagogastric junction cancers.
TrialStudy ArmsEfficacy OutcomesClinical ImplicationsRef.
ORRmPFSmOS
HER2
ToGA
n = 594
phase III trial,
metastatic HER2+ G/GEJ cancers, first line
CTx (Capecitabine or 5- FU plus Cisplatin) 35%5.5 months11.1 monthsTrastuzumab plus CTx is standard of care in first-line treatment in metastatic HER2+ disease.[3]
CTx (Capecitabine or 5- FU plus Cisplatin) with Trastuzumab47%,
p = 0.0017
6.7 months,
p = 0.0002
13.8 months, p = 0.0046
DESTINY-Gastric01
n = 188
phase II trial, HER2+
Asian metastatic gastric cancer patients, third or later-line
CTx (Irinotecan or Paclitaxel) 14%3.5 months8.4 monthsFDA approval for Trastuzumab Deruxtecan in HER2+ G/GEJ cancer
patients who have received a prior Trastuzumab-based regimen. No approval in Europe yet.
[9]
Trastuzumab Deruxtecan51%,
p < 0.001
5.6 months,
p = 0.01
12.5 months
KEYNOTE-811
n = 264
phase III trial, metastatic HER2+ G/GEJ cancers,
first-line,
interim analysis
CTx (CAPOX or 5FU plus Cisplatin) plus Trastuzumab51.9% No dataNo dataNo mature data yet. Combination of HER2 targeting and immune checkpoint inhibition might have synergistic effects.[10]
CTx (CAPOX or 5FU plus Cisplatin) plus Trastuzumab with Pembrolizumab74.4%,
p = 0.00006
No dataNo data
VEGFR
REGARD
n = 355
phase III trial,
metastatic G/GEJ cancers, second-line
placebo3% 1.3 months3.8 monthsRamucirumab mono therapy is approved for second-line treatment in G/GEJ cancers.[4]
Ramucirumab3%, p = 0.762.1 months,
p < 0.0001
5.2 months,
p = 0.047
RAINBOW
n = 665
phase III trial, metastatic G/GEJ cancers, second-line
CTx (Paclitaxel)16%2.9 months7.4 monthsRamucirumab in
combination with
Paclitaxel therapy is approved for second-line treatment in G/GEJ cancers.
[5]
CTx (Paclitaxel) with Ramucirumab28%,
p = 0.0001
4.4 months,
p = 0.0001
9.6 months, p = 0.017
PD-1
ATTRACTION-4
n = 724
phase II trial, metastatic Asian G/GEJ cancer patients, first-line
CTx (S-1 or Capecitabine plus Oxaliplatin) 47.8%8.34 months17.15 monthsBiomarker based patient selection is needed. In second-line tretament or
later MSI high G/GEJ
cancers do benefit from immune checkpoint blockade.
[11]
CTx (S-1 or Capecitabine plus Oxaliplatin) with Nivolumab57.5%,
p = 0.0088
10.45 months,
p = 0.0007
17.45 months, p = 0.26
ATTRACTION-2
n = 493
phase III trial,
metastatic Asian G/GEJ cancer patients, third or later-line
placebo 0%1.45 months4.14 months[12]
Nivolumab11.2%1.61 months,
p < 0.0001
5.26 months, p < 0.0001
CheckMate-649
n = 1,581
phase III trial,
metastatic patients with oesophageal, gastric or
GEJ cancers,
first-line
CTx (Capecitabine plus Oxaliplatin or FOLFOX) 45%6.1 monthsCPS ≥ 5: 11.1 months;
All patients: 11.6 months
FDA approved Nivolumab
in combination with chemotherapy as a first-line therapy in metastatic
G/EGJ cancers.
EMA decision is still pending.
[13]
CTx (Capecitabine plus Oxaliplatin or FOLFOX) with
Nivolumab
60%,
p < 0.0001
7.7 months,
p < 0.0001
CPS ≥ 5: 14.4 months, p < 0.0001;
All patients: 13.8 months, p = 0.0002
Nivolumab plus IpilimumabNo dataNo dataNo dataNot published yet
Claudin 18.2
FAST
n = 161
phase II trial,
metastatic CLDN18.2+
(≥40% tumor cells)
G/GEJ cancers,
first-lin
CTx (ECX) 33.3%5.3 months8.4 monthsCLDN18.2 might be a promising target in
future. Further validation is currently ongoing.
[14]
CTx (ECX) with
Zolbetuximab
49.4%, p = 0.0227.5 months,
p < 0.0005
13.0 months,
p = 0.0008
FGFR2
FIGHT
n = 155
phase II trial,
metastatic G/GEJ cancers (FGFR2b overexpression),
first-line
CTx (modified FOLFOX6) 33%7.4 month12.9 monthsFGFR targeting might be reasonable for a small
cohort of gastric cancer patients. Validation studies need to be performed.
[15]
CTx (modified FOLFOX6) with Bemarituzumab47%9.5 months,
p = 0.073
Not reached, p = 0.027
Abbreviations: G: gastric; GEJ: gastroesophageal junction; ORR: objective response rate; mOS: median overall survival; mPFS: median progression free survival; FDA: Food and Drug Administration; EMA: European Medicines Agency; CTx: chemotherapy; ECX: Capecitabine plus Oxaliplatin and Epirubicin.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Körfer, J.; Lordick, F.; Hacker, U.T. Molecular Targets for Gastric Cancer Treatment and Future Perspectives from a Clinical and Translational Point of View. Cancers 2021, 13, 5216. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13205216

AMA Style

Körfer J, Lordick F, Hacker UT. Molecular Targets for Gastric Cancer Treatment and Future Perspectives from a Clinical and Translational Point of View. Cancers. 2021; 13(20):5216. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13205216

Chicago/Turabian Style

Körfer, Justus, Florian Lordick, and Ulrich T. Hacker. 2021. "Molecular Targets for Gastric Cancer Treatment and Future Perspectives from a Clinical and Translational Point of View" Cancers 13, no. 20: 5216. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13205216

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop