Next Article in Journal
Shaping the Landscape of Lung Cancer: The Role and Therapeutic Potential of Matrix Metalloproteinases
Previous Article in Journal
The Successful and Safe Real-Time TDM-Guided Treatment of Invasive Pulmonary Aspergillosis Using Isavuconazole Administered by Enteral Tube
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tissue Biomarkers in Gastric Cancer Treatment: Present and Future

1
Medical Oncology, University Hospital of Parma, 43100 Parma, Italy
2
Department of Medicine and Surgery, University of Parma, 43100 Parma, Italy
3
GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43100 Parma, Italy
4
Pathological Anatomy, Laboratory Valdès, 81200 Cagliari, Italy
5
Biobank Lab, Department of Oncobiology and Epigenetics, University of Lodz, 90-237 Lodz, Poland
6
Medical Oncology Unit, University Hospital of Cagliari, University of Cagliari, 09100 Cagliari, Italy
7
Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Transl. Med. 2024, 4(4), 640-660; https://doi.org/10.3390/ijtm4040045
Submission received: 30 August 2024 / Revised: 15 November 2024 / Accepted: 18 November 2024 / Published: 22 November 2024

Abstract

:
The aggressive nature of gastric cancer often leads to late diagnosis and poor prognosis. Chemotherapy and the more recently added immunotherapy remain key treatments for this disease. Several studies have focused on identifying tissue biomarkers with prognostic and/or predictive roles and therefore the therapeutic options are rapidly growing. In this narrative review, we summarize the major tissue biomarkers routinely assessed in clinical practice. In addition, we focus on new evidence about emerging tissue biomarkers that could have a predictive role in future therapeutic approaches and also on the potential role of liquid biopsy in this neoplasm.

1. Introduction

Gastric cancer (GC) accounted for approximately 5.6% of newly diagnosed cancer in 2020 and the incidence will continue to increase due to the ageing population and rising exposure to risk factors. The incidence of GC is higher in men, and it varies by region; the highest rates are observed in Eastern Asia, Central and Eastern Europe and South America [1]. These differences in terms of incidence could be explained by the prevalence of helicobacter pylori and Epstein–Barr virus (EBV) infections, alcohol habits, high-salt diets, low fruit and vegetable intake, obesity and gastro-esophageal reflux [2]. GC aggressiveness relies on rapid tumor growth, which makes it hard to detect the malignancy in its early stages. Overall, about 60% of patients with GC are not eligible for curative treatment due to the advanced stage of the disease or comorbidities [3]. Even though the therapeutic options are rapidly growing, the prognosis of GC remains poor, with a 5-year relative survival rate of 36% [4].
Since the first evidence in 1957 [5], the fluoropyrimidine-based chemotherapy (CT) combination has remained the backbone of perioperative treatment for locally advanced disease [6,7,8] and the frontline treatment for advanced stage disease [9,10,11,12]. Recently, immunotherapy with programmed cell death-1 (PD-1) inhibitors in combination with CT has been introduced in specific subtypes in the advanced disease [13,14].
Despite initially high chemosensitivity, the rapid onset of chemoresistance dramatically impacts on patients’ prognosis; survival in clinical trials assessing the value of chemotherapy has historically been <1 year in non-Asian countries [15]. The standard second-line treatments have consisted of mono-chemotherapy paclitaxel, docetaxel or irinotecan for years, although recently the addiction of the anti-vascular endothelial growth factor receptor 2 (VEGFR2) antibody, ramucirumab, to paclitaxel, or as a single agent [16], became the new second-line standard treatment [17].
In this poor scenario, several studies have focused on identifying tissue biomarkers with prognostic and/or predictive roles. Their differentiation into intestinal and diffuse types according to the Lauren classification [18] and into papillary, tubular, mucinous (colloid) and poorly cohesive carcinomas according to the system proposed by the World Health Organization [19] showed no clinical utility. The Lauren classification [18] divides GC as follows:
(a)
Diffuse type (poor cohesion between neoplastic cells, variable component of cells showing signet ring cell morphology);
(b)
Intestinal type (composed of tubular or glandular structures similar to intestinal adenocarcinoma);
(c)
Mixed;
(d)
Undifferentiated.
The WHO classification [19] of GC comprises a more detailed classification based on the morphological and, in some cases, immune expression profile of tumors:
(a)
Adenocarcinoma NOS (tubular, parietal, papillary, micropapillary, mucoepidermoid, mucinous, signet ring, poorly cohesive, medullary, hepatoid, Paneth cell);
(b)
Squamous cell carcinoma;
(c)
Adenosquamous carcinoma;
(d)
Undifferentiated carcinoma (large cell, pleomorphic, sarcomatoid);
(e)
Gastroblastoma;
(f)
Neuroendocrine tumor.
Some subtypes can be recognized in both classifications: the Lauren intestinal subtype corresponds to the papillary and well- and moderately differentiated tubular carcinoma in the WHO classification. The Lauren diffuse subtype corresponds with the poorly cohesive categories of the WHO classification [20].
The Cancer Genome Atlas (TCGA) molecular classification of GC has identified four major genomic classes, each characterized by distinct pathway alterations. These alterations hold potential for identifying clinically relevant biomarkers and developing new therapeutic options [21]. There is some overlap in the genomic features defining each molecular subtype among the four classes.
The first group comprises Epstein–Barr Virus (EBV)-infected tumors, which exhibit a significant enrichment of a high EBV burden and extensive DNA promoter hypermethylation and are associated with a better prognosis [22]. The second group consists of microsatellite instability (MSI) tumors, which are enriched for MSI and exhibit elevated mutation rates and hypermethylation (including at the MLH1 promoter). Chromosomal instability (CIN) and genomically stable (GS) types are distinguished by the presence or absence, respectively, of extensive somatic copy-number aberrations. The GS subtype is associated with the worst prognosis [22]. Notably, receptor tyrosine kinase oncogene amplifications, such as human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), fibroblast growth factor receptor 2 (FGFR2) and MET, are particularly described in the CIN subtype [21].
Understanding these pathway alterations has led to the development of targeted cancer therapies. However, these therapies are more effective in specific subgroups of GC expressing specific biomarkers. The aim of this review is to provide an overview on the tissue biomarkers that are currently useful in clinical practice and other biomarkers with potential utility in the future.

2. Tissue Biomarkers in Standard Treatments

2.1. Programmed Cell Death Ligand 1

The programmed cell death ligand 1 (PD-L1) is a protein expressed mainly in activated T-lymphocytes and other immune system cells and it represents the primary ligand of PD-1. This interaction is among the multiple co-inhibitory signals that modulate T cell activation against self-proteins for the maintenance of self-tolerance as well as infections and tumor antigens [23]. The expression of PD-L1 can be exploited by cancer cells as an important mechanism of immune escape [24]. Thus, to guide patient selection for immune checkpoint inhibitor (ICI) therapy, PD-L1 biomarker expression is routinely assessed by immunohistochemistry (IHC). PD-L1 expression can be explicit as a tumor proportion score (TPS), which is the ratio of PD-L1-expressing cells to all viable tumor cells; however, in GC, the combined positive score (CPS), which also includes the number of PD-L1-positive lymphocytes and macrophages, is considered a more reliable predictive biomarker for immunotherapy efficacy [25]. Therefore, major guidelines recommend the assessment of the PD-L1 combined positive score (CPS) in patients with metastatic GC to tailor first-line treatment in combination with chemotherapy [26,27]. Different IHC PD-L1 antibody assays (Dako 22C3, Dako 28–8, Ventana SP-142 and Ventana SP-263) have been recognized by the Food and Drug Administration (FDA) as companion diagnostics to assess PD-L1 and candidate patients for different ICI treatments in various tumor types [28].
One of the first evidence of ICI activity in GC was reported by the KEYNOTE-059 phase II study, evaluating the efficacy of monotherapy with pembrolizumab in heavily pretreated GC. Based on these results, the Food and Drug Administration (FDA) approved pembrolizumab as a third-line treatment for GC patients with a CPS ≥ 1 [29]. However, in the subsequent KEYNOTE-061 phase III trial, pembrolizumab failed to demonstrate a significant improvement in overall survival (OS) versus paclitaxel as a second-line therapy for patients with a PD-L1 CPS ≥ 1 GC [30]. The antibody used to determine the CPS in KEYNOTE-059 and KEYNOTE-061 was the Dako 22C3 [29,30].
The CheckMate-649 trial compared the addition of nivolumab to CT versus CT alone (either capecitabine/oxaliplatin or 5-FU/leucovorin/oxaliplatin) in treatment-naive gastric, esophago-gastric junction (OGJ) or esophageal cancer: in patients with a PD-L1 CPS ≥ 5 (evaluated with Dako 28-8 antibody) GC, the addition of nivolumab to CT improved OS with a hazard ratio (HR) of 0.71 (98% CI, 0.59 to 0.86; p < 0·0001) and progression-free survival (PFS) (HR 0.68, 98% CI, 0.56 to 0.81; p < 0·0001) [13]. Based on this evidence, ICIs targeting the PD-1/PD-L1 pathway combined with fluoropyrimidine and platinum-based CT are now standard first-line treatment for patients with HER2-negative PD-L1 CPS ≥ 5 GC [14].
Table 1 shows clinical trials and evidence on the combination of immunotherapy and chemotherapy in GC patients.
In the primary clinical trials involving these two assays, the question of whether the 22C3 and 28-8 pharmDx assays could be used interchangeably in GC remained unanswered. A recent study demonstrated a high overall percentage agreement between the PD-L1 22C3 and 28-8 pharmDx assays at CPS cut-offs of 1, 10 and 50 [31]. However, these findings were not corroborated by another study, which suggested that in a larger cohort, tumors that tested positive for PD-L1 using the 28-8 assay were twice as prevalent compared to those tested using the 22C3 and other assays [32]. Other criticisms regarding the use of this biomarker in clinical practice stem from variations in cut-off values and the diverse range of assays employed in clinical trials. Additionally, PD-L1 expression exhibits spatial and temporal heterogeneity [33,34], potentially leading to inconsistencies in PD-L1 expression between surgical samples and their corresponding biopsy specimens. Moreover, CT exposure can alter PD-L1 expression levels in tumor samples [35]. Various thresholds for the TPS and CPS have been employed across studies, with a CPS cut-off of ≥1 indicating PD-L1-positive tumors; the prevalence of PD-L1 CPS ≥ 1 tumors ranges between 50% and 60% [25,36].
Studying PD-L1 expression differences between core biopsies and surgical resection samples in GC remains an unmet need. Ye et al. addressed this gap by conducting a study aimed at determining the number of core biopsies needed to accurately reflect the PD-L1 expression status of resection specimens [37]. The study highlighted the spatial heterogeneity of this biomarker, revealing variations in PD-L1 staining status among different cores from the same tumor. Moreover, the sensitivity and concordance rates of a single tissue microarray core in predicting the PD-L1 status of the entire tumor were found to be very low (0.49 and 60.4%, respectively). Consequently, the study suggests that a minimum of five core biopsies is necessary to achieve high concordance with the overall tumor PD-L1 status [37].

2.2. Human Epidermal Growth Factor Receptor 2 (HER2)

HER2 is a transmembrane protein member of the epidermal growth factor receptor (EGFR) family. HER2 heterodimerization initiates a signal transduction cascade that promotes cell proliferation [38]. When amplificated and/or overexpressed, HER2 acts as an oncogene in numerous types of human cancer, especially in breast cancer and GC [39].
The overexpression of HER2 is reported in approximately 20% of patients with GC, being more prevalent in GEJ tumors, in the intestinal subtype and in well-differentiated GC [40,41]. Several studies have demonstrated that HER2 overexpression represents an independent negative prognostic marker in GC as it correlates with tumor size and lympho-vascular invasion [42,43,44]. After the development and success of molecular targeted drugs, HER2 expression has become an important predictive biomarker; thus, its accurate determination is crucial to identify patients who might benefit from HER2-targeting therapy.
According to the guidelines, HER2 status determined by IHC to assess protein overexpression or by fluorescence in situ hybridization (FISH) to test for HER2 gene amplification is recommended to guide the choice of first-line therapy [45,46]. Since 2010, the combination of trastuzumab and standard CT has represented the first-line treatment for patients with advanced HER2-overexpressing GC: in the phase III ToGA trial, the addition of trastuzumab to platinum/fluoropyrimidine-based CT significantly improved both the PFS (6.7 vs. 5.5. months, HR of 0.71, p = 0.0002) and OS (13.8 vs. 11.1 months; HR 0.74, p = 0.0046) [47].
In recent years, preclinical research has investigated the crosstalk between the PDL-1/PD1 axis and HER2, leading to clinical trials aimed at assessing the potential synergistic activity of a dual PD-1/HER2 blockade [48,49]. Recently, in the phase III KEYNOTE-811 trial, the combination of pembrolizumab with trastuzumab and CT as the first-line treatment for HER2-positive advanced GC showed higher overall response rate (ORR) and significantly improved PFS compared to the standard of care (SoC), especially in patients with a PD-L1 CPS ≥ 1 (10.8 months versus 7.2 months; HR 0.70). Based on these findings, both the FDA and EMA granted accelerated approval for the pembrolizumab/trastuzumab/chemotherapy combination, which currently represents the treatment of choice for first-line therapy in HER2 advanced GC with a PD-L1 CPS ≥ 1 [50].
The treatment landscape is rapidly evolving, and new molecules have demonstrated promising activity. Regarding first-line therapy, the use of zanidatamab (ZW25), an HER2-targeted bispecific antibody, combined with chemotherapy, demonstrated durable disease control, with encouraging PFS and OS results: an ongoing global phase III study (HERIZON-GEA-01) is evaluating zanidatamab plus standard chemo with or without the PD-1 inhibitor, tislelizumab, for the first-line treatment of advanced HER2-positive advanced GC [51].
Margetuximab is an Fc-engineered, anti-HER2 monoclonal antibody designed to increase antibody-dependent cytotoxicity (ADCC) and enhance PD-1/PD-L1 axis expression [52]. In cohort A of the phase II/III MAHOGANY trial, the combination of margetuximab and retifanlimab, an anti-PD1 agent, demonstrated a favorable toxicity profile and ORR compared with the historical outcomes of CT plus trastuzumab in first-line HER2-positive/PD-L1-positive advanced GC [53].
Moving to the second-line setting, several phase III trials with other HER2-targeting agents, such as pertuzumab, lapatinib and trastuzumab emtansine (T-DM1), failed to demonstrate any significant clinical benefit for HER2-positive GC or GEJ cancer [54,55,56]. This discrepancy may be due to both intra- and inter-tumoral heterogeneity in HER2 expression and its downregulation following trastuzumab therapy [57,58]. For these reasons, the reassessment of HER2 status through tissue biopsy is generally required in clinical trials and is recommended, even though recent studies suggest the potential role of the less invasive circulating tumor cell DNA in tracking trastuzumab resistance and evaluating HER2 expression [59,60,61]. Trastuzumab deruxtecan (T-DXd) is an antibody–drug conjugate (ADC) consisting of an anti-HER2 antibody, a cleavable tetrapeptide-based linker and a cytotoxic topoisomerase I inhibitor. With its high drug-to-antibody ratio (8:1) and membrane permeability of its payload, T-DXd does not require high-level HER2 expression and can be active against cancers with low HER2 expression using the bystander effects of the payload [62]. T-DXd proved to be effective in advanced HER2-positive GC: DESTINY-Gastric01 showed an ORR of 51% of the patients treated with T-DXd versus 14% of those in the physician’s choice group (p < 0.001) and an improvement in OS in heavily treated patients, and DESTINY-Gastric02 also confirmed the results in a second-line setting [63,64]. The ongoing randomized phase III DESTINY-Gastric04 study is investigating the efficacy and safety of T-DXd compared with the SoC ramucirumab and paclitaxel and will define the optimal second-line therapy in patients progressed from a first-line treatment containing trastuzumab [65].
Concerning third- or later-line treatment options, a randomized ongoing phase III study, RC48-C007, want to compare a novel ADC, disitamab vedotin, with the physician-selected standard treatment in patients with heavily pretreated advanced GC with HER2 overexpression [66].
Table 2 summarizes the anti-HER2 drugs and their level of development.

2.3. Microsatellite Instability (MSI)

The mismatch repair (MMR) system comprises a set of proteins involved in repairing DNA errors generated during DNA replication [67]. The MMR proteins with clinical relevance in human cancer biology are MLH-1, MSH-2, MSH-6 and PMS-2 [68]. Deficient mismatch repair (dMMR) function leads to the accumulation of mutations in the genome, which is a common occurrence in cancer development. This can arise from either germline mutations or somatic events [69]. The most common cause of somatic MMR gene inactivation involves MLH-1, often due to the hypermethylation of its promoter sequence. It is typically linked to widespread genomic hypermethylation, known as CpG island methylator phenotype (CIMP) high status. Defective MMR results in an elevated tumor mutational burden (TMB), associated with the expression of numerous abnormal proteins acting as neoantigens. These neoantigens serve as targets for cancer immunotherapies [70].
Defective MMR can be identified through IHC to assess MMR protein expression or by detecting microsatellite instability (MSI) using the polymerase chain reaction (PCR) or next-generation sequencing (NGS) methods [71].
MSI-high (MSI-H) phenotypes represent a distinct subclass, and they are mutually exclusive with other biomarkers usually used in clinical practice [72]. Patients with MSI-H GCs who have undergone radical resection have a better prognosis compared with patients with non-MSI-H subtypes [23].
In the metastatic setting, the MSI-H phenotype has shown high responsiveness to immunotherapy, as evidenced by findings from several clinical trials such as phase II KEYNOTE-059, phase III KEYNOTE-061 and phase III KEYNOTE-062. In a post hoc analysis conducted by Joseph Chao et al., it was revealed that pembrolizumab, either as a monotherapy or in combination with chemotherapy (CT), demonstrated durable antitumor activity compared to chemotherapy alone in patients with MSI-H GC or GEJ cancer. Importantly, this effect was observed regardless of the line of therapy in which pembrolizumab was administered [29,30,73,74]. A prespecified analysis of CheckMate 649 showed a survival advantage associated with nivolumab combined with chemotherapy in the group of patients with MSI-high tumors compared to those with microsatellite-stable tumors [13].
A recent meta-analysis with retrospective data on MSI status showed that anti-PD-1 agents with or without CT significantly and consistently improved OS, PFS and ORR compared to CT alone in the subgroup of patients with MSI-H advanced GC [75]. Based on the results of the phase II KEYNOTE-158, pembrolizumab is recommended in monotherapy as a second-line treatment for MSI-H/dMMR GC [74,76]. PD1/PDL1 expression changes with MMR status: PD1/PDL1 expression and CD8+ T cell/CD68+ macrophage densities are higher in MSI GC and can enhance the efficacy of immunotherapy [77].
The combination of CT and immunotherapy or immunotherapy alone may be alternative treatment choices even in the neoadjuvant setting. Phase II IMHOTEP was designed to evaluate the safety and effectiveness of perioperative pembrolizumab in patients with localized dMMR/MSI tumors, regardless of the tumors’ anatomical site. An interim analysis revealed a limited pathological complete response (pCR) rate to short-course neoadjuvant pembrolizumab; however, prolonging the pre-operative treatment duration might enhance the pCR rate [78]. The promising findings from the phase II NEONIPIGA trial support the safety and efficacy of neoadjuvant nivolumab plus low-dose ipilimumab followed by adjuvant nivolumab in patients with resectable locally advanced dMMR/MSI-H resectable GC/GEJ adenocarcinoma [79]. The study demonstrated a high rate of pCR without an increase in expected toxicities [79]. Given the better survival association and the potential limited benefit from CT in MSI-H tumors, neoadjuvant or definitive dual CTLA-4/PD(L)-1 inhibition may allow the omission of chemotherapy or surgery. The INFINITY II trial investigated the activity and safety of tremelimumab–durvalumab as neoadjuvant or definitive treatment for MSI, dMMR and EBV-negative resectable GAC/GEJAC. Pre-operative treatment for 3 months was safe and exhibited promising efficacy in MSI dMMR patients, suggesting the possibility of exploring non-operative management in patients with a clinical, pathological and molecular complete response after treatment [80]. Although not practice-changing, these results lay the groundwork for potential paradigm shifts in the standard of care in this patient subgroup.

2.4. Neurotrophic Tyrosine Receptor Kinases (NTRKs)

NTRKs are a family of transmembrane receptor tyrosine kinases involved in neuronal development. The three TRK family members, TRKA, TRKB and TRKC, encoded by the NTRK1, NTRK2 and NTRK3 genes, respectively, have an extracellular ligand-binding domain, a transmembrane region and an intracellular kinase domain and their activation promotes survival and proliferation in cancer cells [81]. Rearrangements of the NTRK1, NTRK2 and NTRK3 genes are the result of intra- or inter-chromosomal translocations. The 3’ end of the NTRK gene, which contains the tyrosine kinase domain, fuses with the 5’ end of a different gene, resulting in a fusion protein that is missing the ligand-binding domain, leading to ligand-independent receptor activation and the uncontrolled initiation of downstream pathways, lastly driving oncogenesis [82]. While other molecular alterations, such as activating splice variants, somatic mutations and amplifications, have been observed, there is no evidence to suggest that they function as oncogenic drivers. Additionally, these alterations do not exhibit a noteworthy response to NTRK inhibitors [82].
Targeted RNA-sequencing methods might be considered the gold standard for NTRK fusion detection, assuming that the RNA quality is optimal. Furthermore, where tissue availability is restricted, a DNA/RNA-based approach may be preferred if DNA and/or RNA has already been extracted and is accessible; IHC could serve as screening method or to confirm the presence of the fusion [83].
NTRK-altered GCs represent a rare entity: they represent < 1% of GC. GCs represent 8.5% of NTRK-mutated tumors [84]. Usually, NTRK alterations are detected in CIN neoplasms [21]. In clinical practice, the decision to conduct NTRK fusion testing involves the consideration of various factors, such as the tumor type and probability of harboring a fusion, resource accessibility and costs, tissue availability, requirements for concomitant genomic profiling and clinical evaluation. Currently, there are no guidelines indicating the routine clinical test useful for NTRK fusion detection or its treatment in GC [84].
Larotrectinib has received FDA agnostic approval for the treatment of patients with advanced solid tumors harboring an NTRK gene fusion. Given the poor prognosis of GC and GEJ cancer patients, larotrectinib might be a potential therapeutic option [85]. Recently, the TRIDENT-1 trial showed the efficacy of repotrectinib [86], obtaining FDA accelerated approval for the treatment of NTRK-positive advanced tumors [87].
In a preliminary analysis of data from three phase I/II trials (ALKA-372-001, STARTRK-1 and STARTRK-2 [88,89]), entrectinib, a potent TRKA/B/C inhibitor, demonstrated significant and durable antitumor activity and a manageable safety profile in adults with NTRK fusion-positive (NTRK-fp) gastrointestinal cancers. An updated analysis confirmed that entrectinib maintains a high level of clinical benefit for patients with NTRK-fp solid tumors and a manageable safety profile independently of cancer primitivity, reaffirming its promising therapeutic potential [90].

3. Future Perspectives

3.1. Epstein–Barr Virus

EBV is an oncovirus involved in the genesis of different cancers (e.g., nasopharyngeal carcinoma and T cell lymphoma) but also in 9% of GCs [91,92]. In the Cancer Genome Atlas (TCGA) classification, EBV-associated GCs are characterized by high tumoral immune infiltration and show molecular signs of potential sensitivity to immunotherapy such as the amplification of the chromosome 9 locus harboring the PD-L1 and PD-L2 genes [21,93]. Derks et al. found that PD-L1 expression by tumors or tumors infiltrating immune cells is a general phenomenon in EBV-associated cancers along with MSI GCs; this provides a strong rationale for testing ICIs in this patient population and for evaluating EBV status along with MSI status as key variables in immunotherapy trials [93].
The EBV test can be performed using EBV-encoded RNA (EBER) in situ hybridization (ISH) or immunostaining, but these tests have a low sensitivity. Bai et al. developed an NGS-based EBV test for the simultaneous molecular definition of GC, which could improve the diagnostic sensitivity and specificity provided by the EBER-ISH test [94].
The gene expression profile analysis of EBV-positive GC shows an increased expression of the gene involved in the immune response with the activation of infiltrating T CD8+ T cells and macrophages and a better impact on prognosis [91,95]. However, the correlation between EBV status and treatment efficacy with nivolumab has not been evaluated in the CheckMate 649 [13] and ATTRACTION-04 trials [96]. A single-center phase II trial evaluated 61 patients with metastatic GC treated with pembrolizumab in monotherapy; the patients underwent molecular profiling including an EBV test of the primary tumor with EBV DNA sequencing. Six (9.8%) patients were confirmed as EBV-positive and all of them obtained a benefit from pembrolizumab with a partial response, a median duration of response of 8.5 months and an ORR of 100% [97]. In addition, they confirmed that EBV-associated GC and MSI GC are mutually exclusive [97].
EBV infection could be a predictive biomarker of response to ICI as effective as MSI-H, but further studies are needed to establish its potential clinical role.

3.2. Claudin 18.2

Claudins (CLDNs) play an essential role in cellular adhesion and migration. The expression of CLDNs has been reported in several types of cancer, including GC, showing its potential role as a new therapeutic target [98]. Claudin 18 isoform 2 (CLDN18.2), a member of the claudin protein family, plays a crucial role in regulating tissue permeability, paracellular transport and signal transduction through tight junctions. It is primarily expressed in the gastric mucosa and remains present even during malignant transformation. Claudin 18.2 may become more exposed in malignant tissues with the disruption of tight junctions; for this reason, it represents an attractive target for cancer treatment [99,100]. CLDN18.2 expression was assessed by immunohistochemistry (IHC) and CLDN positivity was defined as moderate-to-strong expression in ≥75% of tumor cells [101]. The prevalence of CLDN positivity is reported to be 30–33% in patients with GC/GEJC and it is associated with diffuse-type [102,103] and GS GCs [21].
Zolbetuximab is a novel chimeric immunoglobulin G1 antibody highly specific for CLDN18.2. It binds to CLDN18.2 on the tumor cell to activate ADCC and complement-dependent cytotoxicity [104]. The SPOTLIGHT trial, a global double-blinded phase III trial, aimed to verify the efficacy of CT with a modified schedule of 5-fluorouracile and oxaliplatin for 6 cycles (mFOLFOX6) plus Zolbetuximab compared with mFOLFOX6 plus placebo in CLDN18.2-positive advanced GC/GEJC. Both the primary endpoint of PFS and the secondary endpoint of OS were significantly improved with the addition of zolbetuximab to the standard treatment [105]. The GLOW trial showed that zolbetuximab added to first-line chemotherapy with capecitabine and oxaliplatin added a benefit in terms of PFS and OS in CLDN18.2-positive, HER2-negative GC [106].
Kubota Y et al. examined genomic alterations and immune cell markers in relation to CLDN18.2 expression. CLDN-positive tumors (cut off by 75%) could be found among various molecular subtypes, suggesting that CLDN18.2 could be targetable regardless of the molecular subtype. In addition, there were no significant differences between CLDN-positive and CLDN-negative tumors regarding the efficacy of standard first- and second-line CT, and CLDN positivity was not a prognostic factor in advanced unresectable or metastatic GC/ GEJC. Furthermore, nearly 50% of the CLDN-positive tumors showed a concomitant PD-L1 CPS ≥ 5 [101]; for this reason, an ongoing trial is evaluating the efficacy and safety of zolbetuximab in combination with immunotherapy [107].

3.3. Tumor Mutational Burden (TMB)

In the years since the immunotherapy revolution, the tumor mutational burden (TMB) has been studied as a potential predictive biomarker of response to ICIs [108]. The TMB cut-off value described for predicting a response in GC patients treated with ICIs was 13.31 mt/Mb [109]. The TMB is represented by the total number of mutations present in a tumor specimen and it has been hypothesized that highly mutated tumors express several neoantigens that could contribute to immune cell activation [110]. MSI-H tumors are generally associated with a high TMB, since defects in DNA damage repair such as dMMR lead to the accumulation of mutations, although there are microsatellite-stable (MSS) tumors with a high TMB [110].
Different trials have tried to evaluate the association between the TMB and ICI response in GC. Kim ST et al. revealed, with a small study on GC, how MSI-H, EBV positivity and high TMB were associated with high response rates to pembrolizumab [97]. A post hoc analysis of the KEYNOTE-061 second-line trial showed a prolonged OS, higher response rate and durable responses with pembrolizumab in patients with a PD-L1 CPS ≥ 10 [36]. An exploratory analysis of KEYNOTE-061 evaluated the association of the TMB with PD-L1 and MSI-H and their relationship with the clinical outcomes, revealing a significant association between the TMB and clinical outcomes in the pembrolizumab arm. However, this analysis is weakened by the small sample size [111]. Also, in the first-line setting, a favorable association between the TMB and ICI benefit emerged: the KEYNOTE-062 trial suggested a benefit in the TMB in GC treated with the addition of pembrolizumab; however, the clinical utility of the data was attenuated by the exclusion of MSI-H tumors [112].
The TMB is a significant predictive biomarker for selecting patients who may benefit from ICIs, but its clinical application is limited by the high cost and wet laboratory resources needed for its evaluation. To reduce costs and increase the reproducibility of the data, a multimodal fusion deep learning model was tested to predict TMB levels [113].

3.4. BRAF Mutations

Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF) mutations are related to tumorigenesis since they can influence abnormal cell growth and proliferation through MEK and ERK pathway activation [114]. The most frequently detected BRAF mutation is a single amino acid substitution (V600E) in exon 15 and BRAF mutation is frequent in melanoma (67%) and colorectal cancer (10%) [115]. It is not yet clear if BRAF mutation is a driver mutation in GC: BRAF mutations were observed only in 2% of patients and most of them were uncommon mutations of BRAF (i.e., V599M) [116].
Lee S. et al. suggested that alteration of the rat sarcoma (RAS)–Rapidly Accelerated Fibrosarcoma (RAF) kinase pathway by BRAF mutation together with RAS mutation may play an important role in gastric carcinogenesis [117]. In colorectal cancer, KRAS and BRAF mutations appear to be mutually exclusive, while two reports demonstrated that GC could harbor KRAS and BRAF mutations simultaneously [117,118].
Because of the rarity of these alterations, no data are available about the efficacy of treatments available for BRAF-mutated tumors.

3.5. RET

REarranged during Transfection (RET) is a transmembrane receptor encoded by the RET proto-oncogene situated on chromosome 10q11.2 and plays an important role in the development of nervous and urogenital systems and cells derived from the neural crest [119,120]. Chromosomal rearrangements, point mutations and overexpression of the RET gene can result in the development and progression of various tumors [121]. There is no widely accepted standard method for detecting RET rearrangements. In general, reverse transcription polymerase chain reaction (RT-PCR) and FISH are both sensitive and effective approaches [122].
RET alterations, such as fusions or mutations, are present in multiple tumor types, in particular, in lung and thyroid cancers but also in non-canonical (e.g., gastrointestinal, breast, gynecological, genitourinary, histiocytic) cancers [123].
Many multikinase RET inhibitors (MKIs), such as cabozantinib and lenvatinib, have demonstrated activity against RET. The relatively limited efficacy of MKIs in treating RET-driven cancers can be partially attributed to their off-target activity [124,125,126,127]. In recent years, there has been significant progress in the development of selective RET inhibitors aimed at higher potency and lower toxicity. The next-generation small molecular inhibitors pralsetinib (BLU-667) and selpercatinib (LOXO-292) have been rapidly tested in clinical practice [128,129]. Beyond lung cancers and thyroid cancers, the clinical activity of selective RET inhibitors has been seen in patients with GI cancers (pancreatic cancer and intrahepatic bile duct carcinoma) [130,131]. In total, 3.3% of RET-altered tumors are GCs [132]. Overall, further research into the potential benefits of RET inhibitors in the treatment of GC is needed.

3.6. MET

Hepatocyte growth factor receptor (MET) is a tyrosine kinase receptor with pleiotropic effects. MET might be, together with its ligand hepatocyte growth factor/scatter factor (HGF/SF), a possible therapeutic target in GC. The MET pathway is often hyperactivated in GC by overexpression, which is detectable by IHC and/or in situ hybridization in 50–60% of tumors [133]. Since its implementation in clinical practice, MET aberrations are often detected by an NGS test. Similarly to other biomarkers, MET expression in GC is heterogenous and it has been demonstrated that MET-positive GCs are characterized by poor prognosis [134].
Despite numerous studies on anti-MET agents, few notable results have been achieved. The P-phase I/II and III trials that explored monoclonal antibodies directed against the HGF/MET pathway, such as rilotumumab, onartuzumab or emibetuzumab, failed to demonstrate activity in the treatment of GC [135,136,137,138,139]. In addition, multi-tyrosine kinase inhibitors (TKIs) (e.g., crizotinib) and selective MET TKIs (capmatinib, tivantinib, savolitinib and AMG337) were studied in GC in phase II trials conducted on small groups of patients, but they showed no significant effectiveness [140,141,142,143]. The VIKTORY umbrella trial showed the most successful response to a selective MET TKI, savolitinib, in patients affected by GC and compared to the standard of care, paclitaxel plus ramucirumab as a second-line therapy. MET amplification was determined by NGS or FISH and an ORR of 50% was reached, showing the possible potential of cMET inhibitors in GC in a well-defined population [144].
Further research is needed to establish the role of MET inhibitors in GC treatment.

3.7. FGFR2

The dysregulation of the fibroblast growth factor receptor (FGFR) pathway has been studied in many different types of tumors [145]. In GC, the FGFR2 is the most frequently mutated member of the FGFR family; the prevalence of the FGFR2 amplification varies by country, ranging from 4–7.4% [146,147]. FGFR2 amplification can be detected both using ISH or NGS [148]. So far, there is no established association between FGFR2 amplification and the anatomical site, histological subtype or stage [148]. Hur et al. demonstrated that FGFR2 gene amplification is a poor prognostic factor in patients with metastatic GC and that a high copy number (≥30) identified by NGS-based genomic profiling is significantly associated with a lower PFS and a shorter OS [149].
In the phase II FIGHT clinical trial, a novel FGFR2 inhibitor, bemarituzumab, a humanized anti-FGFR2b monoclonal IgG1 antibody, has shown interesting results in patients affected by advanced GC, with FGFR2 gene amplification in the circulating tumor DNA (ctDNA); the patients received mFOLFOX6 plus bemarituzumab as a first-line therapy versus mFOLFOX6 plus placebo. The primary endpoint, median PFS, was 9.5 months in the bemarituzumab group versus 7.4 months in the placebo group (HR: 0.68, 95% CI 0.44–1.04; p = 0.073), while median OS was not reached. This finding indicates that the higher the percentage of tumor cells expressing FGFR2b, the more effective the targeted therapy [150,151]. Based on these positive results, a phase III trial in gastric cancer overexpressing FGFR2b (FORTITUDE-101) and a phase Ib/III trial combining mFOLFOX6 plus nivolumab and bemarituzumab (FORTITUDE-102) are ongoing [152].

4. Role of Liquid Biopsy

Liquid biopsy aims to collect different biological sources such as circulating tumor cells (CTCs), cell-free nucleic acids, exosomes, circulating tumor microemboli (CTM) or tumor-educated platelets (TEPs). The detection of these circulating factors, in terms of presence or concentration, is an emerging tool that can be used in clinical practice, from early diagnosis to the prediction of the best therapeutic option [153]. Among solid tumors, the potential impact of liquid biopsy has been most studied in colorectal cancer, non-small cell lung cancer and breast cancer, but is less studied in GC [154,155,156].
CtDNA and circulating free DNA (cfDNA) have been shown in several studies to help the detection of metastatic disease. CtDNA is composed of DNA fragments originating from tumor cells, while cfDNA is defined as DNA fragments released by cells and it is composed of ctDNA, circulating cell-free mitochondrial DNA and cell-free fetal DNA [157]. Kim et al. demonstrated that mean plasma cfDNA levels were the lowest among healthy subjects and highest in patients with advanced GC [158,159]. The Japanese MONSTAR-SCREEN study performed a serial circulating tumor DNA assay involving 540 patients with advanced solid tumors, of whom 133 had gastrointestinal (GI) tumors (48 with GC). The trial showed that ctDNA levels in gastrointestinal tumors were significantly higher compared to other tumor types [160].
CfDNA and/or ctDNA detection is a more sensitive method for identifying tumor-specific genetic changes than traditional biopsies. For example, the detection rate of FGFR2 amplification was higher with ctDNA analysis despite tissue biopsy because of tumor heterogeneity, improving treatment strategies and efficacy [161]. Furthermore, cfDNA and ctDNA have been shown to be helpful in different solid malignancies for detecting new resistance mechanisms to chemotherapy and targeted therapy. Li J. et al. showed that the amount of cfDNA or ctDNA after therapy predicts patients’ outcome; in particular, during the treatment, in GC patients, the progression of the disease was associated with higher concentrations of plasma cfDNA/ctDNA over time, whereas the concentration of cfDNA/ctDNA remained stable in patients with stable disease [162]. Kim et al. showed that patients with advanced PD-L1-positive GC treated with pembrolizumab achieved an ORR of 83.3%. This was associated with a higher mutational load in ctDNA prior to treatment, while patients with a low mutational load achieved an ORR of only 7.7% [97]. In addition, it was also found that patients with reduced ctDNA six weeks after therapy had extended progression-free survival [97]. A clinical trial investigated the use of ctDNA for the detection of biomarkers of resistance to trastuzumab therapy in 39 patients with advanced GC; the authors showed a consistent correlation of clonal mutations between tumor and blood samples, identifying 32 mutations potentially related to trastuzumab resistance [163]. In summary, in the non-metastatic setting, liquid biopsy can provide biomarkers for early cancer detection, estimate tumor volume and help determine the prognosis. In the metastatic setting, the levels of cfDNA show a positive correlation with the tumor burden [158]. For this reason, the monitoring of cfDNA can help to differentiate between disease progression and radiological pseudo-progression, opening a window for immediate treatment changes in clinical practice [164,165].
CTCs can help in monitoring tumor recurrence and metastatic spread [166]. Significant correlations between CTC counts and patient survival time have been found in many studies. In a prospective trial enrolling 93 patients with respectable GC, patients with CTC levels ≥ 5/7.5 mL detected in postoperative blood samples had significantly inferior disease-free survival (DFS) and OS than those with a smaller number of CTCs; the correlation between high levels of CTCs and poor OS can be explained by CTCs’ ability to aggregate, forming CTM, facilitating cancer metastasis [167]. Further studies demonstrated that the CTC expression of specific molecules can be helpful in understanding the prognosis of patients with GC. For instance, N-cadherin is a marker related to GC relapse and N-cadherin expression on CTCs can be a predictor of a higher risk of recurrence [168]. The detection of HER2-positive CTCs in patients with advanced GCs that are HER2-negative based on tissue biopsy allowed the identification of patients who can benefit from trastuzumab treatment [169] Therefore, the number of HER2-positive CTCs can be a useful tool for monitoring the effectiveness of trastuzumab therapy [170].
Exosomes are small (30–140 nm) membrane-bound extracellular vesicles that are secreted by large multivesicular bodies and are released into the extracellular environment through fusion. Many cell types can release exosomes and they can be detected in blood, urine and cerebrospinal fluid. Non-coding RNAs (ncRNAs) can be packaged into exosomes and they can be related to angiogenesis, formation of the premetastatic niche and metastasis, indicating their role in the initiation and progression of cancer [171]. TEPs are among the newest liquid biopsy components; TEPs help cancer cells grow and escape from the immune system and they can show tumoral fingerprints. Several studies are ongoing to clearly define the role of exosomal ncRNAs and TEPs in clinical practice and to determine their prognostic value [172].

5. Conclusions

Several biomarkers have been explored and are being evaluated in GC with the aim of tailoring treatment and improving the clinical outcomes of GC patients. Table 3 sums up the most important evidence and clinical practice.
To date, only some of these findings are currently validated as predictive factors for guiding targeted treatment in the clinical practice (e.g., CPS score for ICI in combination with standard CT, HER2 for anti-HER2 agents) [13,14,47,74,76]. However, most of them still require further extensive assessment to better define their impact on the efficacy of targeted therapy strategies. The results of ongoing clinical trials are eagerly awaited, as well as further research on other potential predictive factors, to understand the real impact of the biomarkers under study in improving survival in GC, which still has a dismal prognosis despite the standard treatments.

Author Contributions

Conceptualization, G.A., C.S. and C.T.; validation, G.A. and C.T.; writing—original draft preparation, G.A., V.A., M.P. (Marianna Peroni) and F.P.; writing—review and editing, V.A., M.P. (Marianna Peroni), F.P., G.M., M.U.-N., E.L. and C.S.; visualization, G.A. and C.T.; supervision, S.B., G.M., M.U.-N., E.L., M.P. (Marco Puzzoni), F.C., N.D., M.S., C.S. and C.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors have no conflicts of interest related to this work.

References

  1. Ferlay, J.; Ervik, M.; Lam, F.; Laversanne, M.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today (Version 1.1); International Agency for Research on Cancer: Lyon, France, 2024; Available online: https://gco.iarc.who.int/today (accessed on 3 March 2024).
  2. Colquhoun, A.; Arnold, M.; Ferlay, J.; Goodman, K.J.; Forman, D.; Soerjomataram, I. Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut 2015, 64, 1881–1888. [Google Scholar] [CrossRef] [PubMed]
  3. Allum, W.; Lordick, F.; Alsina, M.; Andritsch, E.; Ba-Ssalamah, A.; Beishon, M.; Braga, M.; Caballero, C.; Carneiro, F.; Cassinello, F.; et al. ECCO essential requirements for quality cancer care: Oesophageal and gastric cancer. Crit. Rev. Oncol. Hematol. 2018, 122, 179–193. [Google Scholar] [CrossRef] [PubMed]
  4. Stomach Cancer Survival Rates and Statistics—NCI. Available online: https://www.cancer.gov/types/stomach/survival (accessed on 5 March 2024).
  5. Heidelberger, C.; Chaudhuri, N.K.; Danneberg, P.; Mooren, D.; Griesbach, L.; Duschinsky, R.; Schnitzer, R.J.; Pleven, E.; Scheiner, J. Fluorinated Pyrimidines, A New Class of Tumour-Inhibitory Compounds. Nature 1957, 179, 663–666. [Google Scholar] [CrossRef] [PubMed]
  6. Cunningham, D.; Allum, W.H.; Stenning, S.P.; Thompson, J.N.; Van de Velde, C.J.H.; Nicolson, M.; Scarffe, J.H.; Lofts, F.J.; Falk, S.J.; Iveson, T.J.; et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N. Engl. J. Med. 2006, 355, 11–20. [Google Scholar] [CrossRef] [PubMed]
  7. Ychou, M.; Boige, V.; Pignon, J.-P.; Conroy, T.; Bouché, O.; Lebreton, G.; Ducourtieux, M.; Bedenne, L.; Fabre, J.-M.; Saint-Aubert, B.; et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: An FNCLCC and FFCD multicenter phase III trial. J. Clin. Oncol. 2011, 29, 1715–1721. [Google Scholar] [CrossRef]
  8. Reynolds, J.V.; Preston, S.R.; O’Neill, B.; Lowery, M.A.; Baeksgaard, L.; Crosby, T.; Cunningham, M.; Cuffe, S.; Griffiths, G.O.; Parker, I.; et al. Trimodality therapy versus perioperative chemotherapy in the management of locally advanced adenocarcinoma of the oesophagus and oesophagogastric junction (Neo-AEGIS): An open-label, randomised, phase 3 trial. Lancet Gastroenterol. Hepatol. 2023, 8, 1015–1027. [Google Scholar] [CrossRef]
  9. Cunningham, D.; Starling, N.; Rao, S.; Iveson, T.; Nicolson, M.; Coxon, F.; Middleton, G.; Daniel, F.; Oates, J.; Norman, A.R. Capecitabine and Oxaliplatin for Advanced Esophagogastric Cancer. N. Engl. J. Med. 2008, 358, 36–46. [Google Scholar] [CrossRef]
  10. Al-Batran, S.-E.; Hartmann, J.T.; Probst, S.; Schmalenberg, H.; Hollerbach, S.; Hofheinz, R.; Rethwisch, V.; Seipelt, G.; Homann, N.; Wilhelm, G.; et al. Phase III Trial in Metastatic Gastroesophageal Adenocarcinoma with Fluorouracil, Leucovorin Plus Either Oxaliplatin or Cisplatin: A Study of the Arbeitsgemeinschaft Internistische Onkologie. J. Clin. Oncol. 2008, 26, 1435–1442. [Google Scholar] [CrossRef]
  11. Dank, M.; Zaluski, J.; Barone, C.; Valvere, V.; Yalcin, S.; Peschel, C.; Wenczl, M.; Goker, E.; Cisar, L.; Wang, K.; et al. Randomized phase III study comparing irinotecan combined with 5-fluorouracil and folinic acid to cisplatin combined with 5-fluorouracil in chemotherapy naive patients with advanced adenocarcinoma of the stomach or esophagogastric junction. Ann. Oncol. 2008, 19, 1450–1457. [Google Scholar] [CrossRef]
  12. Guimbaud, R.; Louvet, C.; Ries, P.; Ychou, M.; Maillard, E.; André, T.; Gornet, J.-M.; Aparicio, T.; Nguyen, S.; Azzedine, A.; et al. Prospective, Randomized, Multicenter, Phase III Study of Fluorouracil, Leucovorin, and Irinotecan Versus Epirubicin, Cisplatin, and Capecitabine in Advanced Gastric Adenocarcinoma: A French Intergroup (Fédération Francophone de Cancérologie Digestive, Féd. J. Clin. Oncol. 2014, 32, 3520–3526. [Google Scholar] [CrossRef]
  13. 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]
  14. Shitara, K.; Ajani, J.A.; Moehler, M.; Garrido, M.; Gallardo, C.; Shen, L.; Yamaguchi, K.; Wyrwicz, L.; Skoczylas, T.; Bragagnoli, A.C.; et al. Nivolumab plus chemotherapy or ipilimumab in gastro-oesophageal cancer. Nature 2022, 603, 942–948. [Google Scholar] [CrossRef] [PubMed]
  15. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef] [PubMed]
  16. 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] [PubMed]
  17. 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]
  18. Laurén, P. The Two Histological Main Types of Gastric Carcinoma: Diffuse and So-Called Intestinal-Type Carcinoma. Acta Pathol. Microbiol. Scand. 1965, 64, 31–49. [Google Scholar] [CrossRef] [PubMed]
  19. Fléjou, J.-F. Classification OMS 2010 des tumeurs digestives: La quatrième édition. Ann. Pathol. 2011, 31, S27–S31. [Google Scholar] [CrossRef]
  20. Mariette, C.; Carneiro, F.; Grabsch, H.I.; van der Post, R.S.; Allum, W.; de Manzoni, G.; European Chapter of International Gastric Cancer Association. Consensus on the pathological definition and classification of poorly cohesive gastric carcinoma. Gastric Cancer 2019, 22, 1–9. [Google Scholar] [CrossRef] [PubMed]
  21. The Cancer Genome Atlas Research Network; Bass, A.J.; Thorsson, V.; Shmulevich, I.; Reynolds, S.M.; Miller, M.; Bernard, B.; Hinoue, T.; Laird, P.W.; Curtis, C.; et al. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef]
  22. Sohn, B.H.; Hwang, J.E.; Jang, H.J.; Lee, H.S.; Oh, S.C.; Shim, J.J.; Lee, K.-W.; Kim, E.H.; Yim, S.Y.; Lee, S.H.; et al. Clinical Significance of Four Molecular Subtypes of Gastric Cancer Identified by The Cancer Genome Atlas Project. Clin. Cancer Res. 2017, 23, 4441–4449. [Google Scholar] [CrossRef]
  23. Nirschl, C.J.; Drake, C.G. Molecular Pathways: Coexpression of Immune Checkpoint Molecules: Signaling Pathways and Implications for Cancer Immunotherapy. Clin. Cancer Res. 2013, 19, 4917–4924. [Google Scholar] [CrossRef] [PubMed]
  24. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
  25. Kulangara, K.; Zhang, N.; Corigliano, E.; Guerrero, L.; Waldroup, S.; Jaiswal, D.; Jansson, M.; Shah, S.; Hanks, D.; Wang, J.; et al. Clinical utility of the combined positive score for programmed death ligand-1 expression and the approval of pembrolizumab for treatment of gastric cancer. Arch. Pathol. Lab. Med. 2019, 143, 330–337. [Google Scholar] [CrossRef] [PubMed]
  26. Lordick, F.; Carneiro, F.; Cascinu, S.; Fleitas, T.; Haustermans, K.; Piessen, G.; Vogel, A.; Smyth, E. Gastric cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2022, 33, 1005–1020. [Google Scholar] [CrossRef]
  27. Shah, M.A.; Kennedy, E.B.; Alarcon-Rozas, A.E.; Alcindor, T.; Bartley, A.N.; Malowany, A.B.; Bhadkamkar, N.A.; Deighton, D.C.; Janjigian, Y.; Karippot, A.; et al. Immunotherapy and Targeted Therapy for Advanced Gastroesophageal Cancer: ASCO Guideline. J. Clin. Oncol. 2023, 41, 1470–1491. [Google Scholar] [CrossRef]
  28. US Food and Drug Administration. List of Cleared or Approved Companion Diagnostic Devices (In Vitro and Imaging Tools). 2020. Available online: https://www.fda.gov/medicalDevices/productsandMedicalProcedures/InVitroDiagnostics/ucm301431.htm (accessed on 3 March 2024).
  29. 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] [PubMed]
  30. Shitara, K.; Özgüroğlu, M.; Bang, Y.J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.H.; Fornaro, L.; Olesiński, T.; Caglevic, C.; Muro, K.; et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): A randomised, open-label, controlled, phase 3 trial. Lancet 2018, 392, 123–133. [Google Scholar] [CrossRef]
  31. Ahn, S.; Kim, K.M. PD-L1 expression in gastric cancer: Interchangeability of 22C3 and 28-8 pharmDx assays for responses to immunotherapy. Mod. Pathol. 2021, 34, 1719–1727. [Google Scholar] [CrossRef]
  32. Yeong, J.; Lum, H.Y.J.; Teo, C.B.; Tan, B.K.J.; Chan, Y.H.; Tay, R.Y.K.; Choo, J.R.-E.; Jeyasekharan, A.D.; Miow, Q.H.; Loo, L.-H.; et al. Choice of PD-L1 immunohistochemistry assay influences clinical eligibility for gastric cancer immunotherapy. Gastric Cancer 2022, 25, 741–750. [Google Scholar] [CrossRef]
  33. Sundar, R.; Liu, D.H.W.; Hutchins, G.G.A.; Slaney, H.L.; Silva, A.N.S.; Oosting, J.; Hayden, J.D.; Hewitt, L.C.; Ng, C.C.; Mangalvedhekar, A.; et al. Spatial profiling of gastric cancer patient-matched primary and locoregional metastases reveals principles of tumour dissemination. Gut 2021, 70, 1823–1832. [Google Scholar] [CrossRef]
  34. Zhou, K.I.; Peterson, B.; Serritella, A.; Thomas, J.; Reizine, N.; Moya, S.; Tan, C.; Wang, Y.; Catenacci, D.V. Spatial and Temporal Heterogeneity of PD-L1 Expression and Tumor Mutational Burden in Gastroesophageal Adenocarcinoma at Baseline Diagnosis and after Chemotherapy. Clin. Cancer Res. 2020, 26, 6453–6463. [Google Scholar] [CrossRef] [PubMed]
  35. Yang, J.H.; Kim, H.; Roh, S.Y.; Lee, M.A.; Park, J.M.; Park, C.H.; Lee, H.H.; Jung, E.S.; Lee, S.H.; Lee, Y.J.; et al. Discordancy and changes in the pattern of programmed death ligand 1 expression before and after platinum-based chemotherapy in metastatic gastric cancer. Gastric Cancer 2019, 22, 147–154. [Google Scholar] [CrossRef] [PubMed]
  36. Wainberg, Z.A.; Fuchs, C.S.; Tabernero, J.; Shitara, K.; Muro, K.; Van Cutsem, E.; Bang, Y.-J.; Chung, H.C.; Yamaguchi, K.; Varga, E.; et al. Efficacy of pembrolizumab monotherapy for advanced gastric/gastroesophageal junction cancer with programmed death ligand 1 combined positive score ≥10. Clin. Cancer Res. 2021, 27, 1923–1931. [Google Scholar] [CrossRef]
  37. Ye, M.; Huang, D.; Zhang, Q.; Weng, W.; Tan, C.; Qin, G.; Jiang, W.; Sheng, W.; Wang, L. Heterogeneous programmed death-ligand 1 expression in gastric cancer: Comparison of tissue microarrays and whole sections. Cancer Cell Int. 2020, 20, 186. [Google Scholar] [CrossRef]
  38. Slamon, D.J.; Godolphin, W.; Jones, L.A.; Holt, J.A.; Wong, S.G.; Keith, D.E.; Levin, W.J.; Stuart, S.G.; Udove, J.; Ullrich, A.; et al. Studies of the HER-2/ neu Proto-Oncogene in Human Breast and Ovarian Cancer. Science 1989, 244, 707–712. [Google Scholar] [CrossRef]
  39. Kaptain, S.; Tan, L.K.; Chen, B. Her-2/ neu and Breast Cancer. Diagn. Mol. Pathol. 2001, 10, 139–152. [Google Scholar] [CrossRef] [PubMed]
  40. Tewari, M.; Kumar, A.; Mishra, R.; Kumar, M.; Shukla, H.S. HER2 Expression in Gastric and Gastroesophageal Cancer: Report from a Tertiary Care Hospital in North India. Indian J. Surg. 2015, 77, 447–451. [Google Scholar] [CrossRef]
  41. Polkowski, W.; van Sandick, J.W.; Offerhaus, G.J.A.; ten Kate, F.J.W.; Mulder, J.; Obertop, H.; van Lanschot, J.J.B. Prognostic Value of Laurén Classification and c-erbB-2 Oncogene Overexpression in Adenocarcinoma of the Esophagus and Gastroesophageal Junction. Ann. Surg. Oncol. 1999, 6, 290–297. [Google Scholar] [CrossRef] [PubMed]
  42. García, I.; Vizoso, F.; Martín, A.; Sanz, L.; Abdel-Lah, O.; Raigoso, P.; García-Muñiz, J.L. Clinical Significance of the Epidermal Growth Factor Receptor and HER2 Receptor in Resectable Gastric Cancer. Ann. Surg. Oncol. 2003, 10, 234–241. [Google Scholar] [CrossRef]
  43. Jonjić, N.; Kovac, K.; Krasević, M.; Valković, T.; Ernjak, N.; Sasso, F.; Melato, M. Epidermal growth factor-receptor expression correlates with tumor cell proliferation and prognosis in gastric cancer. Anticancer Res. 1997, 17, 3883–3888. [Google Scholar]
  44. Son, H.S.; Shin, Y.M.; Park, K.K.; Seo, K.W.; Yoon, K.Y.; Jang, H.K.; Lee, S.-H.; Yang, S.I.; Kim, J.H. Correlation between HER2 Overexpression and Clinicopathological Characteristics in Gastric Cancer Patients Who Have Undergone Curative Resection. J. Gastric Cancer 2014, 14, 180–186. [Google Scholar] [CrossRef] [PubMed]
  45. Takehana, T.; Kunitomo, K.; Kono, K.; Kitahara, F.; Iizuka, H.; Matsumoto, Y.; Fujino, M.A.; Ooi, A. Status of c- erb B-2 in gastric adenocarcinoma: A comparative study of immunohistochemistry, fluorescence in situ hybridization and enzyme-linked immuno-sorbent assay. Int. J. Cancer 2002, 98, 833–837. [Google Scholar] [CrossRef] [PubMed]
  46. Hofmann, M.; Stoss, O.; Shi, D.; Büttner, R.; Van De Vijver, M.; Kim, W.; Ochiai, A.; Rüschoff, J.; Henkel, T. Assessment of a HER2 scoring system for gastric cancer: Results from a validation study. Histopathology 2008, 52, 797–805. [Google Scholar] [CrossRef]
  47. 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]
  48. Rha, S.Y.; Lee, C.-K.; Kim, H.S.; Kang, B.; Jung, M.; Bae, W.K.; Koo, D.-H.; Shin, S.-J.; Jeung, H.-C.; Zang, D.Y.; et al. Targeting HER2 in combination with anti-PD-1 and chemotherapy confers a significant tumor shrinkage of gastric cancer: A multi-institutional phase Ib/II trial of first-line triplet regimen (pembrolizumab, trastuzumab, chemotherapy) for HER2-positive advanced gastric cancer (AGC). J. Clin. Oncol. 2020, 38, 3081. [Google Scholar] [CrossRef]
  49. 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]
  50. Janjigian, Y.Y.; Kawazoe, A.; Bai, Y.; Xu, J.; Lonardi, S.; Metges, J.P.; Yanez, P.; Wyrwicz, L.S.; Shen, L.; Ostapenko, Y.; et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: Interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial. Lancet 2023, 402, 2197–2208. [Google Scholar] [CrossRef]
  51. Elimova, E.; Ajani, J.A.; Iii, H.A.B.; Denlinger, C.S.; Iqbal, S.; Kang, Y.-K.; Kim, Y.H.H.; Lee, K.-W.; Lin, B.; Mehta, R.; et al. Zanidatamab + chemotherapy as first-line treatment for HER2-expressing metastatic gastroesophageal adenocarcinoma (mGEA). J. Clin. Oncol. 2023, 41, 347. [Google Scholar] [CrossRef]
  52. Catenacci, D.V.T.; Kang, Y.-K.; Park, H.; Uronis, H.E.; Lee, K.-W.; Ng, M.C.H.; 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]
  53. Catenacci, D.; Kang, Y.-K.; Yoon, H.; Shim, B.; Kim, S.; Oh, D.-Y.; Spira, A.; Ulahannan, S.; Avery, E.; Boland, P.; et al. Margetuximab with retifanlimab as first-line therapy in HER2+/PD-L1+ unresectable or metastatic gastroesophageal adenocarcinoma: MAHOGANY cohort A. ESMO Open 2022, 7, 100563. [Google Scholar] [CrossRef]
  54. 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] [PubMed]
  55. Tabernero, J.; Hoff, P.M.; Shen, L.; Ohtsu, A.; A Shah, M.; 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] [PubMed]
  56. Kang, Y.-K.; Shah, M.A.; Ohtsu, A.; Van Cutsem, E.; Ajani, J.A.; van der Horst, T.; Harle-Yge, M.-L.; Piao, Y.; Althaus, B.; Thuss-Patience, P.C. A randomized, open-label, multicenter, adaptive phase 2/3 study of trastuzumab emtansine (T-DM1) versus a taxane (TAX) in patients (pts) with previously treated HER2-positive locally advanced or metastatic gastric/gastroesophageal junction adenocarcinoma (LA/MGC/GEJC). J. Clin. Oncol. 2016, 34, 5. [Google Scholar] [CrossRef]
  57. Stahl, P.; Seeschaaf, C.; Lebok, P.; Kutup, A.; Bockhorn, M.; Izbicki, J.R.; Bokemeyer, C.; Simon, R.; Sauter, G.; Marx, A.H. Heterogeneity of amplification of HER2, EGFR, CCND1 and MYC in gastric cancer. BMC Gastroenterol. 2015, 15, 7. [Google Scholar] [CrossRef] [PubMed]
  58. Seo, S.; Ryu, M.-H.; Park, Y.S.; Ahn, J.Y.; Park, Y.; Park, S.R.; Ryoo, B.-Y.; Lee, G.H.; Jung, H.-Y.; Kang, Y.-K. Loss of HER2 positivity after anti-HER2 chemotherapy in HER2-positive gastric cancer patients: Results of the GASTric cancer HER2 reassessment study 3 (GASTHER3). Gastric Cancer 2019, 22, 527–535. [Google Scholar] [CrossRef]
  59. Zhang, J.; Qiu, W.; Zhang, W.; Chen, Y.; Shen, H.; Zhu, H.; Liang, X.; Shen, Z. Tracking of trastuzumab resistance in patients with HER2-positive metastatic gastric cancer by CTC liquid biopsy. Am. J. Cancer Res. 2023, 13, 5684–5697. [Google Scholar]
  60. Klein-Scory, S.; Ladigan-Badura, S.; Mika, T.; Verdoodt, B.; Tannapfel, A.; Pohl, M.; Schroers, R.; Baraniskin, A. Liquid biopsy based HER2 amplification status in gastric cancer patients indicates clinical response. Heliyon 2023, 9, e21339. [Google Scholar] [CrossRef]
  61. Makiyama, A.; Sukawa, Y.; Kashiwada, T.; Kawada, J.; Hosokawa, A.; Horie, Y.; Tsuji, A.; Moriwaki, T.; Tanioka, H.; Shinozaki, K.; et al. Randomized, Phase II Study of Trastuzumab Beyond Progression in Patients with HER2-Positive Advanced Gastric or Gastroesophageal Junction Cancer: WJOG7112G (T-ACT Study). J. Clin. Oncol. 2020, 38, 1919–1927. [Google Scholar] [CrossRef]
  62. Takegawa, N.; Nonagase, Y.; Yonesaka, K.; Sakai, K.; Maenishi, O.; Ogitani, Y.; Tamura, T.; Nishio, K.; Nakagawa, K.; Tsurutani, J. DS-8201a, a new HER2-targeting antibody–drug conjugate incorporating a novel DNA topoisomerase I inhibitor, overcomes HER2-positive gastric cancer T-DM1 resistance. Int. J. Cancer 2017, 141, 1682–1689. [Google Scholar] [CrossRef]
  63. Van Cutsem, E.; di Bartolomeo, M.; Smyth, E.; Chau, I.; Park, H.; Siena, S.; Lonardi, S.; A Wainberg, Z.; Ajani, J.; Chao, J.; et al. Trastuzumab deruxtecan in patients in the USA and Europe with HER2-positive advanced gastric or gastroesophageal junction cancer with disease progression on or after a trastuzumab-containing regimen (DESTINY-Gastric02): Primary and updated analyses from a single-arm, phase 2 study. Lancet Oncol. 2023, 24, 744–756. [Google Scholar] [CrossRef]
  64. 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] [PubMed]
  65. Shitara, K.; Seraj, J.; Franke, F.; Kawaguchi, Y.; Shen, L.; Kamio, T.; Meinhardt, G.; Tabernero, J. 1436TiP Trastuzumab deruxtecan (T-DXd) in patients (Pts) with HER2-positive gastric cancer (GC) or gastroesophageal junction (GEJ) adenocarcinoma who have progressed on or after a trastuzumab-containing regimen (DESTINY-gastric04, DG-04): A randomized phase III study. Ann. Oncol. 2021, 32, S1073. [Google Scholar] [CrossRef]
  66. Wang, Y.; Gong, J.; Wang, A.; Wei, J.; Peng, Z.; Wang, X.; Zhou, J.; Qi, C.; Liu, D.; Li, J.; et al. Disitamab vedotin (RC48) plus toripalimab for HER2-expressing advanced gastric or gastroesophageal junction and other solid tumours: A multicentre, open label, dose escalation and expansion phase 1 trial. eClinicalMedicine 2024, 68, 102415. [Google Scholar] [CrossRef]
  67. Holliday, R. A mechanism for gene conversion in fungi. Genet. Res. 2007, 89, 285–307. [Google Scholar] [CrossRef] [PubMed]
  68. Tamura, K.; Kaneda, M.; Futagawa, M.; Takeshita, M.; Kim, S.; Nakama, M.; Kawashita, N.; Tatsumi-Miyajima, J. Genetic and genomic basis of the mismatch repair system involved in Lynch syndrome. Int. J. Clin. Oncol. 2019, 24, 999–1011. [Google Scholar] [CrossRef] [PubMed]
  69. Schöniger, S.; Rüschoff, J. Mismatch Repair Deficiency and Microsatellite Instability. Encyclopedia 2022, 2, 1559–1576. [Google Scholar] [CrossRef]
  70. Bateman, A.C. DNA mismatch repair protein immunohistochemistry—An illustrated guide. Histopathology 2021, 79, 128–138. [Google Scholar] [CrossRef]
  71. Bartley, A.N.; Mills, A.M.; Konnick, E.; Overman, M.; Ventura, C.B.; Souter, L.; Colasacco, C.; Stadler, Z.K.; Kerr, S.; Howitt, B.E.; et al. Mismatch Repair and Microsatellite Instability Testing for Immune Checkpoint Inhibitor Therapy: Guideline From the College of American Pathologists in Collaboration With the Association for Molecular Pathology and Fight Colorectal Cancer. Arch. Pathol. Lab. Med. 2022, 146, 1194–1210. [Google Scholar] [CrossRef]
  72. Yu, H.Y.; Li, C.P.; Huang, Y.H.; Hsu, S.J.; Wang, Y.P.; Hsieh, Y.C.; Fang, W.L.; Huang, K.H.; Li, A.F.Y.; Lee, R.C.; et al. Microsatellite Instability, Epstein–Barr Virus, and Programmed Cell Death Ligand 1 as Predictive Markers for Immunotherapy in Gastric Cancer. Cancers 2022, 14, 218. [Google Scholar] [CrossRef]
  73. Tabernero, J.; Van Cutsem, E.; Bang, Y.-J.; Fuchs, C.S.; Wyrwicz, L.; Lee, K.W.; Kudaba, I.; Garrido, M.; Chung, H.C.; Salguero, H.R.C.; et al. Pembrolizumab with or without chemotherapy versus chemotherapy for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma: The phase III KEYNOTE-062 study. J. Clin. Oncol. 2019, 37, LBA4007. [Google Scholar] [CrossRef]
  74. Yoshino, T.; Pentheroudakis, G.; Mishima, S.; Overman, M.; Yeh, K.-H.; Baba, E.; Naito, Y.; Calvo, F.; Saxena, A.; Chen, L.-T.; et al. JSCO—ESMO—ASCO—JSMO—TOS: International expert consensus recommendations for tumour-agnostic treatments in patients with solid tumours with microsatellite instability or NTRK fusions. Ann. Oncol. 2020, 31, 861–872. [Google Scholar] [CrossRef] [PubMed]
  75. Pietrantonio, F.; Randon, G.; Di Bartolomeo, M.; Luciani, A.; Chao, J.; Smyth, E.; Petrelli, F. Predictive role of microsatellite instability for PD-1 blockade in patients with advanced gastric cancer: A meta-analysis of randomized clinical trials. ESMO Open 2021, 6, 100036. [Google Scholar] [CrossRef]
  76. Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.-P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients with Noncolorectal High Microsatellite Instability/Mismatch Repair–Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef]
  77. Wang, Y.-L.; Gong, Y.; Lv, Z.; Li, L.; Yuan, Y. Expression of PD1/PDL1 in gastric cancer at different microsatellite status and its correlation with infiltrating immune cells in the tumor microenvironment. J. Cancer 2021, 12, 1698–1707. [Google Scholar] [CrossRef]
  78. De La Fouchardiere, C.; Zaanan, A.; Cohen, R.; Le Sourd, S.; Tougeron, D.; Soularue, E.; Dubreuil, O.; Willet, N.; Samalin, E.; Piessen, G.; et al. Immunotherapy for localized dMMR/MSI tumors: First interim analysis of the IMHOTEP trial. J. Clin. Oncol. 2023, 41, 2591. [Google Scholar] [CrossRef]
  79. André, T.; Tougeron, D.; Piessen, G.; de la Fouchardière, C.; Louvet, C.; Adenis, A.; Jary, M.; Tournigand, C.; Aparicio, T.; Desrame, J.; et al. Neoadjuvant Nivolumab Plus Ipilimumab and Adjuvant Nivolumab in Localized Deficient Mismatch Repair/Microsatellite Instability–High Gastric or Esophagogastric Junction Adenocarcinoma: The GERCOR NEONIPIGA Phase II Study. J. Clin. Oncol. 2023, 41, 255–265. [Google Scholar] [CrossRef]
  80. Pietrantonio, F.; Raimondi, A.; Lonardi, S.; Murgioni, S.; Cardellino, G.G.; Tamberi, S.; Strippoli, A.; Palermo, F.; Prisciandaro, M.; Randon, G.; et al. INFINITY: A multicentre, single-arm, multi-cohort, phase II trial of tremelimumab and durvalumab as neoadjuvant treatment of patients with microsatellite instability-high (MSI) resectable gastric or gastroesophageal junction adenocarcinoma (GAC/GEJAC). J. Clin. Oncol. 2023, 41, 358. [Google Scholar] [CrossRef]
  81. Manea, C.A.; Badiu, D.C.; Ploscaru, I.C.; Zgura, A.; Bacinschi, X.; Smarandache, C.G.; Serban, D.; Popescu, C.G.; Grigorean, V.T.; Botnarciuc, V. A review of NTRK fusions in cancer. Ann. Med. Surg. 2022, 79, 103893. [Google Scholar] [CrossRef]
  82. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747. [Google Scholar] [CrossRef]
  83. Huygens, S.; Vellekoop, H.; Versteegh, M.; Santi, I.; Szilberhorn, L.; Zelei, T.; Nagy, B.; Tsiachristas, A.; Koleva-Kolarova, R.; Wordsworth, S.; et al. Cost-Effectiveness Analysis of Treating Patients With NTRK-Positive Cancer with the Histology-Independent Therapy Entrectinib. Value Health 2023, 26, 193–203. [Google Scholar] [CrossRef]
  84. Xu, Y.; Shi, X.; Wang, W.; Zhang, L.; Cheung, S.; Rudolph, M.; Brega, N.; Dong, X.; Qian, L.; Wang, L.; et al. Prevalence and clinico-genomic characteristics of patients with TRK fusion cancer in China. NPJ Precis. Oncol. 2023, 7, 75. [Google Scholar] [CrossRef] [PubMed]
  85. Drilon, A.; Laetsch, T.W.; Kummar, S.; Dubois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion–Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef] [PubMed]
  86. Solomon, B.; Drilon, A.; Lin, J.; Bazhenova, L.; Goto, K.; De Langen, J.; Kim, D.-W.; Wolf, J.; Springfeld, C.; Popat, S.; et al. 1372P Repotrectinib in patients (pts) with NTRK fusion-positive (NTRK+) advanced solid tumors, including NSCLC: Update from the phase I/II TRIDENT-1 trial. Ann. Oncol. 2023, 34, S787–S788. [Google Scholar] [CrossRef]
  87. U.S. Food & Drug. FDA Grants Accelerated Approval to Repotrectinib for Adult and Pediatric Patients with NTRK Gene Fusion-Positive Solid Tumors. 13 June 2024. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-repotrectinib-adult-and-pediatric-patients-ntrk-gene-fusion-positive (accessed on 4 July 2024).
  88. Drilon, A.; Siena, S.; Ou, S.-H.I.; Patel, M.; Ahn, M.J.; Lee, J.; Bauer, T.M.; Farago, A.F.; Wheler, J.J.; Liu, S.V.; et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017, 7, 400–409. [Google Scholar] [CrossRef]
  89. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
  90. Patel, M.; Siena, S.; Demetri, G.; Doebele, R.; Chae, Y.; Conkling, P.; Garrido-Laguna, I.; Longo, F.; Rolfo, C.; Sigal, D.; et al. O-3 Efficacy and safety of entrectinib in NTRK fusion-positive gastrointestinal cancers: Updated integrated analysis of three clinical trials (STARTRK-2, STARTRK-1 and ALKA-372-001). Ann. Oncol. 2020, 31, 232–233. [Google Scholar] [CrossRef]
  91. Sun, K.; Jia, K.; Lv, H.; Wang, S.-Q.; Wu, Y.; Lei, H.; Chen, X. EBV-Positive Gastric Cancer: Current Knowledge and Future Perspectives. Front. Oncol. 2020, 10, 583463. [Google Scholar] [CrossRef] [PubMed]
  92. Murphy, G.; Pfeiffer, R.; Camargo, M.C.; Rabkin, C.S. Meta-analysis shows that prevalence of Epstein–Barr virus-positive gastric cancer differs based on sex and anatomic location. Gastroenterology 2009, 137, 824–833. [Google Scholar] [CrossRef]
  93. Derks, S.; Liao, X.; Chiaravalli, A.M.; Xu, X.; Camargo, M.C.; Solcia, E.; Sessa, F.; Fleitas, T.; Freeman, G.J.; Rodig, S.J.; et al. Abundant PD-L1 expression in Epstein-Barr Virus-infected gastric cancers. Oncotarget 2016, 7, 32925–32932. [Google Scholar] [CrossRef]
  94. Bai, Y.; Xie, T.; Wang, Z.; Tong, S.; Zhao, X.; Zhao, F.; Cai, J.; Wei, X.; Peng, Z.; Shen, L. Efficacy and predictive biomarkers of immunotherapy in Epstein-Barr virus-associated gastric cancer. J. Immunother. Cancer 2022, 10, e004080. [Google Scholar] [CrossRef]
  95. Kim, S.Y.; Park, C.; Kim, H.-J.; Park, J.; Hwang, J.; Kim, J.-I.; Choi, M.G.; Kim, K.-M.; Kang, M.-S. Deregulation of immune response genes in patients with Epstein-Barr virus-associated gastric cancer and outcomes. Gastroenterology 2015, 148, 137–147.e9. [Google Scholar] [CrossRef] [PubMed]
  96. Kang, Y.-K.; Chen, L.-T.; Ryu, M.-H.; Oh, D.-Y.; Oh, S.C.; Chung, H.C.; Lee, K.-W.; Omori, T.; Shitara, K.; Sakuramoto, S.; et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): A randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022, 23, 234–247. [Google Scholar] [CrossRef] [PubMed]
  97. 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]
  98. Tabariès, S.; Siegel, P.M. The role of claudins in cancer metastasis. Oncogene 2016, 36, 1176–1190. [Google Scholar] [CrossRef] [PubMed]
  99. Niimi, T.; Nagashima, K.; Ward, J.M.; Minoo, P.; Zimonjic, D.B.; Popescu, N.C.; Kimura, S. claudin-18, a Novel Downstream Target Gene for the T/EBP/NKX2.1 Homeodomain Transcription Factor, Encodes Lung- and Stomach-Specific Isoforms through Alternative Splicing. Mol. Cell. Biol. 2001, 21, 7380–7390. [Google Scholar] [CrossRef]
  100. Sahin, U.; Koslowski, M.; Dhaene, K.; Usener, D.; Brandenburg, G.; Seitz, G.; Huber, C.; Türeci, 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]
  101. Kubota, Y.; Kawazoe, A.; Mishima, S.; Nakamura, Y.; Kotani, D.; Kuboki, Y.; Bando, H.; Kojima, T.; Doi, T.; Yoshino, T.; et al. Comprehensive clinical and molecular characterization of claudin 18.2 expression in advanced gastric or gastroesophageal junction cancer. ESMO Open 2023, 8, 100762. [Google Scholar] [CrossRef]
  102. Moran, D.; Maurus, D.; Rohde, C.; Arozullah, A. Prevalence of CLDN18.2, HER2 and PD-L1 in gastric cancer samples. Ann. Oncol. 2018, 29, viii32. [Google Scholar] [CrossRef]
  103. Pellino, A.; Brignola, S.; Riello, E.; Niero, M.; Murgioni, S.; Guido, M.; Nappo, F.; Businello, G.; Sbaraglia, M.; Bergamo, F.; et al. Association of CLDN18 Protein Expression with Clinicopathological Features and Prognosis in Advanced Gastric and Gastroesophageal Junction Adenocarcinomas. J. Pers. Med. 2021, 11, 1095. [Google Scholar] [CrossRef]
  104. Singh, P.; Toom, S.; Huang, Y. Anti-claudin 18.2 antibody as new targeted therapy for advanced gastric cancer. J. Hematol. Oncol. 2017, 10, 105. [Google Scholar] [CrossRef]
  105. Shitara, K.; Lordick, F.; Bang, Y.-J.; Enzinger, P.; Ilson, D.; A Shah, M.; Van Cutsem, E.; Xu, R.-H.; Aprile, G.; Xu, J.; et al. Zolbetuximab plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma (SPOTLIGHT): A multicentre, randomised, double-blind, phase 3 trial. Lancet 2023, 401, 1655–1668. [Google Scholar] [CrossRef] [PubMed]
  106. Shah, M.A.; Shitara, K.; Ajani, J.A.; Bang, Y.-J.; Enzinger, P.; Ilson, D.; Lordick, F.; Van Cutsem, E.; Plazas, J.G.; Huang, J.; et al. Zolbetuximab plus CAPOX in CLDN18.2-positive gastric or gastroesophageal junction adenocarcinoma: The randomized, phase 3 GLOW trial. Nat. Med. 2023, 29, 2133–2141. [Google Scholar] [CrossRef] [PubMed]
  107. Study Details|A Study to Assess the Antitumor Activity, Safety, Pharmacokinetics and Biomarkers of Zolbetuximab (IMAB362) in Participants with Claudin (CLDN) 18.2 Positive, Metastatic or Advanced Unresectable Gastric and Gastroesophageal Junction (GEJ) Adenocarcinoma and Locoregional Gastric or GEJ Adenocarcinoma|ClinicalTrials.gov. Available online: https://www.clinicaltrials.gov/study/NCT03505320 (accessed on 11 May 2024).
  108. Klempner, S.J.; Fabrizio, D.; Bane, S.; Reinhart, M.; Peoples, T.; Ali, S.M.; Sokol, E.S.; Frampton, G.; Schrock, A.B.; Anhorn, R.; et al. Tumor Mutational Burden as a Predictive Biomarker for Response to Immune Checkpoint Inhibitors: A Review of Current Evidence. Oncologist 2020, 25, e147–e159. [Google Scholar] [CrossRef]
  109. Jang, J.Y.; Jeon, Y.; Jeong, S.Y.; Lim, S.H.; Kang, W.K.; Lee, J.; Kim, S.T. The Optimal Tumor Mutational Burden Cutoff Value as a Novel Marker for Predicting the Efficacy of Programmed Cell Death-1 Checkpoint Inhibitors in Advanced Gastric Cancer. J. Gastric Cancer 2023, 23, 476–486. [Google Scholar] [CrossRef]
  110. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef]
  111. Shitara, K.; Özgüroğlu, M.; Bang, Y.-J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.-H.; Caglevic, C.; Chung, H.; Muro, K.; Van Cutsem, E.; et al. Molecular determinants of clinical outcomes with pembrolizumab versus paclitaxel in a randomized, open-label, phase III trial in patients with gastroesophageal adenocarcinoma. Ann. Oncol. 2021, 32, 1127–1136. [Google Scholar] [CrossRef]
  112. Lee, K.W.; Van Cutsem, E.; Bang, Y.J.; Fuchs, C.S.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; Castro, H.R.; Chao, J.; et al. Association of Tumor Mutational Burden with Efficacy of Pembrolizumab ± Chemotherapy as First-Line Therapy for Gastric Cancer in the Phase III KEYNOTE-062 Study. Clin. Cancer Res. 2022, 28, 3489–3498. [Google Scholar] [CrossRef] [PubMed]
  113. Li, J.; Liu, H.; Liu, W.; Zong, P.; Huang, K.; Li, Z.; Li, H.; Xiong, T.; Tian, G.; Li, C.; et al. Predicting gastric cancer tumor mutational burden from histopathological images using multimodal deep learning. Briefings Funct. Genom. 2023, 23, 228–238. [Google Scholar] [CrossRef]
  114. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  115. Rajagopalan, H.; Bardelli, A.; Lengauer, C.; Kinzler, K.W.; Vogelstein, B.; Velculescu, V.E. RAF/RAS oncogenes and mismatch-repair status. Nature 2002, 418, 934. [Google Scholar] [CrossRef]
  116. Choi, Y.Y.; Noh, S.H.; Cheong, J.-H. Molecular Dimensions of Gastric Cancer: Translational and Clinical Perspectives. J. Pathol. Transl. Med. 2016, 50, 1–9. [Google Scholar] [CrossRef] [PubMed]
  117. Lee, S.H.; Soung, Y.H.; Kim, H.S.; Park, W.S.; Kim, S.Y.; Lee, J.H.; Park, J.Y.; Cho, Y.G.; Kim, C.J.; Nam, S.W.; et al. BRAF and KRAS mutations in stomach cancer. Oncogene 2003, 22, 6942–6945. [Google Scholar] [CrossRef]
  118. Ali, S.M.; Sanford, E.M.; Klempner, S.J.; Rubinson, D.A.; Wang, K.; Palma, N.A.; Chmielecki, J.; Yelensky, R.; Palmer, G.A.; Morosini, D.; et al. Prospective Comprehensive Genomic Profiling of Advanced Gastric Carcinoma Cases Reveals Frequent Clinically Relevant Genomic Alterations and New Routes for Targeted Therapies. Oncologist 2015, 20, 499–507. [Google Scholar] [CrossRef]
  119. Takahashi, M.; Buma, Y.; Iwamoto, T.; Inaguma, Y.; Ikeda, H.; Hiai, H. Cloning and expression of the ret proto-oncogene encoding a tyrosine kinase with two potential transmembrane domains. Oncogene 1988, 3, 571–578. [Google Scholar] [PubMed]
  120. Mulligan, L.M. GDNF and the RET Receptor in Cancer: New Insights and Therapeutic Potential. Front. Physiol. 2019, 9, 1873. [Google Scholar] [CrossRef]
  121. Zhang, F.; Tang, J.M.; Wang, L.; Wu, P.P.; Zhang, M. Immunohistochemical detection of RET proto-oncogene product in tumoral and nontumoral mucosae of gastric cancer. Anal. Quant. Cytopathol. Histpathol. 2014, 36, 128–136. [Google Scholar] [PubMed]
  122. Belli, C.; Penault-Llorca, F.; Ladanyi, M.; Normanno, N.; Scoazec, J.-Y.; Lacroix, L.; Reis-Filho, J.; Subbiah, V.; Gainor, J.; Endris, V.; et al. ESMO recommendations on the standard methods to detect RET fusions and mutations in daily practice and clinical research. Ann. Oncol. 2021, 32, 337–350. [Google Scholar] [CrossRef]
  123. Desilets, A.; Repetto, M.; Yang, S.-R.; Sherman, E.J.; Drilon, A. RET-Altered Cancers—A Tumor-Agnostic Review of Biology, Diagnosis and Targeted Therapy Activity. Cancers 2023, 15, 4146. [Google Scholar] [CrossRef]
  124. Romei, C.; Cosci, B.; Renzini, G.; Bottici, V.; Molinaro, E.; Agate, L.; Passannanti, P.; Viola, D.; Biagini, A.; Basolo, F.; et al. RET genetic screening of sporadic medullary thyroid cancer (MTC) allows the preclinical diagnosis of unsuspected gene carriers and the identification of a relevant percentage of hidden familial MTC (FMTC). Clin. Endocrinol. 2011, 74, 241–247. [Google Scholar] [CrossRef]
  125. Yakes, F.M.; Chen, J.; Tan, J.; Yamaguchi, K.; Shi, Y.; Yu, P.; Qian, F.; Chu, F.; Bentzien, F.; Cancilla, B.; et al. Cabozantinib (XL184), a Novel MET and VEGFR2 Inhibitor, Simultaneously Suppresses Metastasis, Angiogenesis, and Tumor Growth. Mol. Cancer Ther. 2011, 10, 2298–2308. [Google Scholar] [CrossRef]
  126. Carlomagno, F.; Vitagliano, D.; Guida, T.; Ciardiello, F.; Tortora, G.; Vecchio, G.; Ryan, A.; Fontanini, G.; Fusco, A.; Santoro, M. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res. 2002, 62, 7284–7290. [Google Scholar] [PubMed]
  127. Wedge, S.R.; Ogilvie, D.J.; Dukes, M.; Kendrew, J.; Chester, R.; Jackson, J.A.; Boffey, S.J.; Valentine, P.J.; Curwen, J.O.; Musgrove, H.L.; et al. ZD6474 inhibits vascular endothelial growth factor signaling, angiogenesis, and tumor growth following oral administration. Cancer Res. 2002, 62, 4645–4655. [Google Scholar] [PubMed]
  128. Subbiah, V.; Gainor, J.F.; Rahal, R.; Brubaker, J.D.; Kim, J.L.; Maynard, M.; Hu, W.; Cao, Q.; Sheets, M.P.; Wilson, D.; et al. Precision Targeted Therapy with BLU-667 for RET-Driven Cancers. Cancer Discov. 2018, 8, 836–849. [Google Scholar] [CrossRef] [PubMed]
  129. Subbiah, V.; Velcheti, V.; Tuch, B.; Ebata, K.; Busaidy, N.; Cabanillas, M.; Wirth, L.; Stock, S.; Smith, S.; Lauriault, V.; et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann. Oncol. 2018, 29, 1869–1876. [Google Scholar] [CrossRef]
  130. Gainor, J.F.; Lee, D.H.; Curigliano, G.; Doebele, R.C.; Kim, D.-W.; Baik, C.S.; Tan, D.S.-W.; Lopes, G.; Gadgeel, S.M.; Cassier, P.A.; et al. Clinical activity and tolerability of BLU-667, a highly potent and selective RET inhibitor, in patients (pts) with advanced RET-fusion+ non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2019, 37, 9008. [Google Scholar] [CrossRef]
  131. Drilon, A.E.; Subbiah, V.; Oxnard, G.R.; Bauer, T.M.; Velcheti, V.; Lakhani, N.J.; Besse, B.; Park, K.; Patel, J.D.; Cabanillas, M.E.; et al. A phase 1 study of LOXO-292, a potent and highly selective RET inhibitor, in patients with RET-altered cancers. J. Clin. Oncol. 2018, 36, 102. [Google Scholar] [CrossRef]
  132. Andreev-Drakhlin, A.; Roszik, J.; Subbiah, V. The landscape of RET alterations from 56,970 adult patients with cancer: Clinical implications. J. Clin. Oncol. 2019, 37, 3106. [Google Scholar] [CrossRef]
  133. Röcken, C. Predictive biomarkers in gastric cancer. J. Cancer Res. Clin. Oncol. 2022, 149, 467–481. [Google Scholar] [CrossRef]
  134. Metzger, M.; Behrens, H.; Böger, C.; Haag, J.; Krüger, S.; Röcken, C. MET in gastric cancer—Discarding a 10% cutoff rule. Histopathology 2015, 68, 241–253. [Google Scholar] [CrossRef]
  135. Catenacci, D.V.T.; 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]
  136. 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]
  137. Malka, D.; François, E.; Penault-Llorca, F.; Castan, F.; Bouché, O.; Bennouna, J.; Ghiringhelli, F.; de la Fouchardière, C.; Borg, C.; Samalin, E.; et al. FOLFOX alone or combined with rilotumumab or panitumumab as first-line treatment for patients with advanced gastroesophageal adenocarcinoma (PRODIGE 17-ACCORD 20-MEGA): A randomised, open-label, three-arm phase II trial. Eur. J. Cancer 2019, 115, 97–106. [Google Scholar] [CrossRef]
  138. 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. JAMA Oncol. 2017, 3, 620–627. [Google Scholar] [CrossRef]
  139. Sakai, D.; Chung, H.C.; Oh, D.-Y.; Park, S.H.; Kadowaki, S.; Kim, Y.H.; Tsuji, A.; Komatsu, Y.; Kang, Y.-K.; Uenaka, K.; et al. A non-randomized, open-label, single-arm, Phase 2 study of emibetuzumab in Asian patients with MET diagnostic positive, advanced gastric cancer. Cancer Chemother. Pharmacol. 2017, 80, 1197–1207. [Google Scholar] [CrossRef]
  140. Kaji, M.; Yonemura, Y.; Harada, S.; Liu, X.; Terada, I.; Yamamoto, H. Participation of c-met in the progression of human gastric cancers: Anti-c-met oligonucleotides inhibit proliferation or invasiveness of gastric cancer cells. Cancer Gene Ther. 1996, 3, 393–404. [Google Scholar]
  141. Inoue, T.; Chung, Y.; Yashiro, M.; Nishimura, S.; Hasuma, T.; Otani, S.; Sowa, M. Transforming Growth Factor-β and Hepatocyte Growth Factor Produced by Gastric Fibroblasts Stimulate the Invasiveness of Scirrhous Gastric Cancer Cells. Jpn. J. Cancer Res. 1997, 88, 152–159. [Google Scholar] [CrossRef]
  142. Toiyama, Y.; Yasuda, H.; Saigusa, S.; Matushita, K.; Fujikawa, H.; Tanaka, K.; Mohri, Y.; Inoue, Y.; Goel, A.; Kusunoki, M. Co-expression of hepatocyte growth factor and c-Met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c-Met signaling in gastric cancer. Int. J. Cancer 2011, 130, 2912–2921. [Google Scholar] [CrossRef]
  143. 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]
  144. 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]
  145. Babina, I.S.; Turner, N.C. Advances and challenges in targeting FGFR signalling in cancer. Nat. Rev. Cancer 2017, 17, 318–332. [Google Scholar] [CrossRef]
  146. Schrumpf, T.; Behrens, H.-M.; Haag, J.; Krüger, S.; Röcken, C. FGFR2 overexpression and compromised survival in diffuse-type gastric cancer in a large central European cohort. PLoS ONE 2022, 17, e0264011. [Google Scholar] [CrossRef]
  147. Katoh, M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat. Rev. Clin. Oncol. 2018, 16, 105–122. [Google Scholar] [CrossRef]
  148. Ahn, S.; Lee, J.; Hong, M.; Kim, S.T.; Park, S.H.; Choi, M.G.; Lee, J.-H.; Sohn, T.S.; Bae, J.M.; Kim, S.; et al. FGFR2 in gastric cancer: Protein overexpression predicts gene amplification and high H-index predicts poor survival. Mod. Pathol. 2016, 29, 1095–1103. [Google Scholar] [CrossRef]
  149. 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]
  150. 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]
  151. A Wainberg, Z.; Enzinger, P.C.; Kang, Y.-K.; Qin, S.; Yamaguchi, K.; Kim, I.-H.; Saeed, A.; Oh, S.C.; Li, J.; Turk, H.M.; et al. Bemarituzumab in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma (FIGHT): A randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 2022, 23, 1430–1440. [Google Scholar] [CrossRef]
  152. Smyth, E.C.; Chao, J.; Muro, K.; Yen, P.; Yanes, R.E.; Zahlten-Kumeli, A.; Rha, S.Y. Trial in progress: Phase 3 study of bemarituzumab + mFOLFOX6 versus placebo + mFOLFOX6 in previously untreated advanced gastric or gastroesophageal junction (GEJ) cancer with FGFR2b overexpression (FORTITUDE-101). J. Clin. Oncol. 2022, 40, TPS4164. [Google Scholar] [CrossRef]
  153. Poulet, G.; Massias, J.; Taly, V. Liquid Biopsy: General Concepts. Acta Cytol. 2019, 63, 449–455. [Google Scholar] [CrossRef]
  154. Dasari, A.; Morris, V.K.; Allegra, C.J.; Atreya, C.; Benson, A.B., 3rd; Boland, P.; Chung, K.; Copur, M.S.; Corcoran, R.B.; Deming, D.A.; et al. ctDNA applications and integration in colorectal cancer: An NCI Colon and Rectal–Anal Task Forces whitepaper. Nat. Rev. Clin. Oncol. 2020, 17, 757–770. [Google Scholar] [CrossRef]
  155. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic—Implementation issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef]
  156. Rodríguez, J.; Avila, J.; Rolfo, C.; Ruíz-Patiño, A.; Russo, A.; Ricaurte, L.; Ordóñez-Reyes, C.; Arrieta, O.; Zatarain-Barrón, Z.L.; Recondo, G.; et al. When Tissue is an Issue the Liquid Biopsy is Nonissue: A Review. Oncol. Ther. 2021, 9, 89–110. [Google Scholar] [CrossRef]
  157. Spellman, P.T.; Gray, J.W. Detecting cancer by monitoring circulating tumor DNA. Nat. Med. 2014, 20, 474–475. [Google Scholar] [CrossRef]
  158. Kim, K.; Shin, D.G.; Park, M.K.; Baik, S.H.; Kim, T.H.; Kim, S.; Lee, S. Circulating cell-free DNA as a promising biomarker in patients with gastric cancer: Diagnostic validity and significant reduction of cfDNA after surgical resection. Ann. Surg. Treat. Res. 2014, 86, 136–142. [Google Scholar] [CrossRef]
  159. Schwarzenbach, H.; Hoon, D.S.B.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef]
  160. Nakamura, Y.; Fujisawa, T.; Kadowaki, S.; Takahashi, N.; Goto, M.; Yoshida, K.; Kawakami, T.; Esaki, T.; Oki, E.; Nishida, N.; et al. Characteristics of genomic alterations in circulating tumor DNA (ctDNA) in patients (Pts) with advanced gastrointestinal (GI) cancers in nationwide large-scale ctDNA screening:SCRUM-Japan Monstar-Screen. J. Clin. Oncol. 2021, 39, 106. [Google Scholar] [CrossRef]
  161. Jogo, T.; Nakamura, Y.; Shitara, K.; Bando, H.; Yasui, H.; Esaki, T.; Terazawa, T.; Satoh, T.; Shinozaki, E.; Nishina, T.; et al. Circulating Tumor DNA Analysis Detects FGFR2 Amplification and Concurrent Genomic Alterations Associated with FGFR Inhibitor Efficacy in Advanced Gastric Cancer. Clin. Cancer Res. 2021, 27, 5619–5627. [Google Scholar] [CrossRef]
  162. Li, J.; Jiang, W.; Wei, J.; Zhang, J.; Cai, L.; Luo, M.; Wang, Z.; Sun, W.; Wang, S.; Wang, C.; et al. Patient specific circulating tumor DNA fingerprints to monitor treatment response across multiple tumors. J. Transl. Med. 2020, 18, 293. [Google Scholar] [CrossRef]
  163. Wang, Y.; Zhao, C.; Chang, L.; Jia, R.; Liu, R.; Zhang, Y.; Gao, X.; Li, J.; Chen, R.; Xia, X.; et al. Circulating tumor DNA analyses predict progressive disease and indicate trastuzumab-resistant mechanism in advanced gastric cancer. EBioMedicine 2019, 43, 261–269. [Google Scholar] [CrossRef]
  164. Bratman, S.V.; Yang, S.Y.C.; Iafolla, M.A.J.; Liu, Z.; Hansen, A.R.; Bedard, P.L.; Lheureux, S.; Spreafico, A.; Razak, A.A.; Shchegrova, S.; et al. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat. Cancer 2020, 1, 873–881. [Google Scholar] [CrossRef]
  165. Matsusaka, S.; Chìn, K.; Ogura, M.; Suenaga, M.; Shinozaki, E.; Mishima, Y.; Terui, Y.; Mizunuma, N.; Hatake, K. Circulating tumor cells as a surrogate marker for determining response to chemotherapy in patients with advanced gastric cancer. Cancer Sci. 2010, 101, 1067–1071. [Google Scholar] [CrossRef]
  166. Pantel, K.; Alix-Panabières, C. Circulating tumour cells and cell-free DNA in gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 73–74. [Google Scholar] [CrossRef]
  167. Zhang, Q.; Shan, F.; Li, Z.; Gao, J.; Li, Y.; Shen, L.; Ji, J.; Lu, M. A prospective study on the changes and clinical significance of pre-operative and post-operative circulating tumor cells in resectable gastric cancer. J. Transl. Med. 2018, 16, 171. [Google Scholar] [CrossRef]
  168. Ishiguro, Y.; Sakihama, H.; Yoshida, T.; Ichikawa, N.; Homma, S.; Fukai, M.; Kawamura, H.; Takahashi, N.; Taketomi, A. Prognostic Significance of Circulating Tumor Cells with Mesenchymal Phenotypes in Patients with Gastric Cancer: A Prospective Study. Ann. Surg. Oncol. 2021, 28, 1178–1186. [Google Scholar] [CrossRef]
  169. Mishima, Y.; Matsusaka, S.; Chin, K.; Mikuniya, M.; Minowa, S.; Takayama, T.; Shibata, H.; Kuniyoshi, R.; Ogura, M.; Terui, Y.; et al. Detection of HER2 Amplification in Circulating Tumor Cells of HER2-Negative Gastric Cancer Patients. Target. Oncol. 2017, 12, 341–351. [Google Scholar] [CrossRef]
  170. Wang, H.; Li, B.; Liu, Z.; Gong, J.; Shao, L.; Ren, J.; Niu, Y.; Bo, S.; Li, Z.; Lai, Y.; et al. HER2 copy number of circulating tumour DNA functions as a biomarker to predict and monitor trastuzumab efficacy in advanced gastric cancer. Eur. J. Cancer 2018, 88, 92–100. [Google Scholar] [CrossRef]
  171. Xie, Y.; Dang, W.; Zhang, S.; Yue, W.; Yang, L.; Zhai, X.; Yan, Q.; Lu, J. The role of exosomal noncoding RNAs in cancer. Mol. Cancer 2019, 18, 37. [Google Scholar] [CrossRef]
  172. Varkey, J.; Nicolaides, T. Tumor-Educated Platelets: A Review of Current and Potential Applications in Solid Tumors. Cureus 2021, 13, e19189. [Google Scholar] [CrossRef]
Table 1. Trial with PD1 inhibitors in clinical practice. Abbreviations: GC: gastric cancer; CPS: PD-L1 combined positive score, FDA: US Food and Drug Administration, CT: chemotherapy, EMA: European Medicines Agency.
Table 1. Trial with PD1 inhibitors in clinical practice. Abbreviations: GC: gastric cancer; CPS: PD-L1 combined positive score, FDA: US Food and Drug Administration, CT: chemotherapy, EMA: European Medicines Agency.
Drug Trial and Phase Determination of PDL1 Level Design of the Study and ResultsRef.
Pembrolizumab KEYNOTE-059, phase IICPS ≥ 1 with Dako 22C3Monotherapy in heavily pretreated GC—FDA approved [29]
PembrolizumabKEYNOTE-061, phase III CPS ≥ 1 with Dako 22C3Pembrolizumab vs. paclitaxel as II line—no advantages [30]
NivolumabCheckMate-649, phase IIIPD-L1 CPS ≥ 5 with Dako 28-8 Nivolumab + CT vs. CT alone as first line—FDA and EMA approved [14]
Table 2. Anti-HER2 treatment in GC. Abbreviations: CT: chemotherapy; SoC: standard of care, FDA: US Food and Drug Administration, EMA: European Medicines Agency.
Table 2. Anti-HER2 treatment in GC. Abbreviations: CT: chemotherapy; SoC: standard of care, FDA: US Food and Drug Administration, EMA: European Medicines Agency.
Drug Trial and Phase Design of the StudyLevel of Development Ref.
Trastuzumab ToGA trial, phase IIITrastuzumab + CT vs. CT alone as first line SoC [47]
TrastuzumabKEYNOTE-811, phase III Trastuzumab + pembrolizumab + CT vs. standard of care as first line in CPS ≥ 1FDA and EMA approved[50]
Zanidatamab (ZW25)HERIZON-GEA-01, phase IIIZanidatamab plus standard CT with or without the PD-1 inhibitor as first lineOngoing trial [51]
MargetuximabMAHOGANY, phase II/IIIMargetuximab with antiPD1 vs. standard of careOngoing[53]
LapatinibTyTAN—A, phase III Lapatinib + paclitaxel vs. paclitaxel as second line Failed [54]
PertuzumabJACOB, phase IIIPertuzumab + standard of care vs. standard of care as first lineFailed [55]
Trastuzumab emtansine (T-DM1)GATSBY, phase II/IIIT-DM1 vs. paclitaxel as second lineFailed [56]
Trastuzumab deruxtecan (T-DXd)DESTINYGastric04, phase IIIT-DXd vs. SoC as second line Ongoing [65]
Table 3. Current biomarkers in GC. Abbreviations: CT, chemotherapy; FISH, fluorescence in situ hybridization; HER2, human epidermal growth factor Receptor 2; IHC, immunohistochemistry; MSI, microsatellite instability; NGS, next-generation sequencing; NTRK, neurotrophic tyrosine receptor kinases; PD-L1, programmed cell death protein ligand 1.
Table 3. Current biomarkers in GC. Abbreviations: CT, chemotherapy; FISH, fluorescence in situ hybridization; HER2, human epidermal growth factor Receptor 2; IHC, immunohistochemistry; MSI, microsatellite instability; NGS, next-generation sequencing; NTRK, neurotrophic tyrosine receptor kinases; PD-L1, programmed cell death protein ligand 1.
BiomarkerAssessment DrugSetting Clinical Trial
PD-L1IHCNivolumab (plus standard CT)
Pembrolizumab (plus trastuzumab and CT)
First line in PD-L1 CPS ≥ 5
First line in HER2-positive GC with PD-L1 CPS ≥ 1
CheckMate-649
KEYNOTE-811
HER2IHC/FISHTrastuzumab (plus standard CT)
Trastuzumab deruxtecan
First line
Third or later line
ToGA
DESTINY-Gastric 0
MSIIHC, PCR or NGSPembrolizumabFirst lineKEYNOTE-158
NTRKRNA-seq, IHCLarotrectinib
Entrectinib
Advanced diseaseLOXO-TRK-14001, NAVIGATE
STARTRK-1 and STARTRK-2
Claudin 18.2IHCZolbetuximab (plus standard CT)
Zolbetuximab plus Capecitabine and Oxaliplatin
First line
First line
SPOTLIGHT
GLOW
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Airò, G.; Agnetti, V.; Pratticò, F.; Peroni, M.; Bui, S.; Mura, G.; Urbanowicz-Nijaki, M.; Lai, E.; Puzzoni, M.; Contu, F.; et al. Tissue Biomarkers in Gastric Cancer Treatment: Present and Future. Int. J. Transl. Med. 2024, 4, 640-660. https://doi.org/10.3390/ijtm4040045

AMA Style

Airò G, Agnetti V, Pratticò F, Peroni M, Bui S, Mura G, Urbanowicz-Nijaki M, Lai E, Puzzoni M, Contu F, et al. Tissue Biomarkers in Gastric Cancer Treatment: Present and Future. International Journal of Translational Medicine. 2024; 4(4):640-660. https://doi.org/10.3390/ijtm4040045

Chicago/Turabian Style

Airò, Giulia, Virginia Agnetti, Fabiana Pratticò, Marianna Peroni, Simona Bui, Giovanni Mura, Maria Urbanowicz-Nijaki, Eleonora Lai, Marco Puzzoni, Fabiana Contu, and et al. 2024. "Tissue Biomarkers in Gastric Cancer Treatment: Present and Future" International Journal of Translational Medicine 4, no. 4: 640-660. https://doi.org/10.3390/ijtm4040045

APA Style

Airò, G., Agnetti, V., Pratticò, F., Peroni, M., Bui, S., Mura, G., Urbanowicz-Nijaki, M., Lai, E., Puzzoni, M., Contu, F., Denaro, N., Scartozzi, M., Solinas, C., & Tommasi, C. (2024). Tissue Biomarkers in Gastric Cancer Treatment: Present and Future. International Journal of Translational Medicine, 4(4), 640-660. https://doi.org/10.3390/ijtm4040045

Article Metrics

Back to TopTop