Skip to main content

A literature review of recent advances in gastric cancer treatment: exploring the cross-talk between targeted therapies

Abstract

Background

Gastric cancer (GC) ranks fourth in global mortality rates and fifth in prevalence, making it one of the most common cancers worldwide. Recent clinical studies have highlighted the potential of immunotherapies as a promising approach to treating GC. This study aims to shed light on the most impactful therapeutic strategies in the context of GC immunotherapy, highlighting both established and emerging approaches.

Main body

This review examines over 160 clinical studies conducted globally, focusing on the effectiveness of various immunotherapy modalities, including cancer vaccines, adoptive cell therapy, immune checkpoint inhibitors (ICIs), and monoclonal antibodies (mAbs). A comprehensive search of peer-reviewed literature was performed using databases such as Web of Science, PubMed, and Scopus. The selection criteria included peer-reviewed articles published primarily within the last 10 years, with a focus on studies that provided insights into targeted therapies and their mechanisms of action, clinical efficacy, and safety profiles. The findings indicate that these immunotherapy strategies can enhance treatment outcomes for GC, aligning with current treatment guidelines. ICIs like pembrolizumab and nivolumab have shown significant survival benefits in specific GC subgroups. Cancer vaccines and CAR-T cell therapies demonstrate potential, while mAbs targeting HER2 and VEGFR pathways enhance outcomes in combination regimens. We discuss the latest advancements and challenges in targeted therapy and immunotherapy for GC. Given the evolving nature of this field, this research emphasizes significant evidence-based therapies and those currently under evaluation rather than providing an exhaustive overview. Challenges include resistance mechanisms, immunosuppressive tumor environments, and inconsistent results from combination therapies. Biomarker-driven approaches and further research into emerging modalities like CAR-T cells and cancer vaccines are critical for optimizing treatments.

Conclusions

Immunotherapy is reshaping GC management by improving survival and quality of life. Ongoing research and clinical evaluations are crucial for refining personalized and effective therapies.

Introduction

Gastric Cancer (GC) represents a significant global health burden, ranking as the fifth most commonly diagnosed malignancy and the fourth leading cause of cancer-related mortality worldwide. According to GLOBOCAN 2022 data, GC accounts for nearly one million new cases annually, resulting in approximately 659,000 deaths [1]. Despite advancements in detection and treatment, the prognosis for GC remains poor, particularly for advanced and metastatic cases, where therapeutic options are primarily palliative. Currently, complete surgical resection with lymph node dissection offers the only curative treatment for GC. However, most patients present with advanced-stage disease at diagnosis, rendering them ineligible for surgery. Conventional treatment modalities, including radiation therapy and cytotoxic chemotherapy, have demonstrated limited efficacy due to inherent challenges such as radioresistance, chemoresistance, and the high proportion of unresectable tumors [2, 3]. These limitations underscore the urgent need for novel therapeutic strategies to improve patient outcomes.

The advent of precision medicine has significantly enhanced the understanding of GC pathogenesis and molecular heterogeneity, paving the way for the development of targeted therapies tailored to specific tumor characteristics. Agents such as trastuzumab and ramucirumab, designed for HER-2-positive and VEGFR2-positive GC subtypes, respectively, represent notable advancements in this domain. However, the modest survival benefits observed with these agents highlight the necessity for further innovation [4, 5].

In recent years, immunotherapy has emerged as a promising strategy in GC treatment, offering the potential to address the limitations of traditional approaches. Immune-based interventions, including ICIs, cancer vaccines, adoptive cell therapy, and mAbs, have demonstrated encouraging clinical outcomes. Drugs such as pembrolizumab have already been incorporated into treatment guidelines for advanced GC, and numerous other immunotherapeutic agents are currently under investigation in clinical trials [5, 6].

The evolving landscape of GC treatment necessitates a comprehensive understanding of the opportunities and challenges of novel therapeutic approaches. Although immunotherapies and targeted medicines have shown great promise, information gaps about their best use, resistance mechanisms, and suitability for various patient populations make it difficult to incorporate them into clinical practice. A deeper comprehension of the molecular composition of GC is necessary to address these issues and direct the creation of more potent solutions. In this article, we provide a detailed review of recent advances in GC treatment, focusing on updates in targeted therapy and immunotherapy. By synthesizing evidence from the latest clinical trials and preclinical studies, we aim to highlight the most promising therapies, critically evaluate their limitations, and identify areas for further research. While not exhaustive—given the rapidly evolving nature of this field—this study seeks to emphasize proven therapies and those under active evaluation. Ultimately, our goal is to contribute to developing personalized treatment paradigms that improve survival outcomes and quality of life for GC patients.

Heterogeneity of GC microenvironment

Cancer cells progress in complex tissue environments and their development is associated with desmoplastic reactions involving immune cells and different stromal cell types in the local environment, which is called tumor microenvironment (TME). Components of this environment and interactions among them ultimately determine the growth, invasion, and metastasis of tumors and more importantly predict therapeutic efficacy [7, 8]. There is growing data that the inflammatory environment within GC involves a variety of cell types, including natural killer cells (NK cells), dendritic cells (DCs), tumor-associated mast cells, tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), tumor-infiltrating neutrophils and lymphocytes and (TINs and TILs), and mesenchymal stem cells [8, 9]. It has been shown that all of the cells listed as well as their products (cytokines) together with changes in the extracellular matrix, microRNAs [10, 11], and deregulated cellular signaling contribute to a TME in GC that enables tumor cells to evade host immunity and develop resistance to multiple treatments [7]. In Fig. 1, the interactions between immune cells within TME are illustrated.

Fig. 1
figure 1

An overview of the interactions of the immune cells in TME; Different components of this environment and interactions among them which ultimately determine the growth, invasion, and metastasis of tumors is presented

Tumor-associated macrophages (TAMs)

A subset of macrophages known as TAMs or tumor-associated macrophages are vital to immune cell infiltration of tumors. TAMs originate from inflammatory monocytes in the blood that express the chemokine receptor type 2 (CCR2) [12]. In addition to accelerating tumor growth, TAMs release pro-inflammatory cytokines, proteolytic enzymes, and growth factors, that cause tumor progression and resistance to treatment [7, 8]. It is known that up to 50% of the entire mass of the tumor may be made up of infiltrating macrophages, which tends to be linked with poor prognosis regarding solid tumors [13]. There are two main categories of macrophages, classified based on their responses to the microenvironment as M1 (Classical) and M2 (Alternative). Macrophages of type M1 perform inflammatory functions and aid in pathogen clearance and anti-tumor immunity. M1 macrophages, through the creation of chemokines such as IL-1α, TNF-α, IL-6, IL-1β, and IL-12 drive the polarization and recruitment of consequently amplifying the type 1 immune response and type 1 T helper cells (Th1) [14]. Alternatively, M2 macrophages, which compose an important part of TAM accumulation, possess anti-inflammatory functions, are stimulated by type 2 T helper cell cytokines (IL-10, IL-13, and IL-4), and represent pro-tumorigenic functions, which include tumor progression [15]. This frame shows a direct relationship between tumor cells and TAMs. Numerous cytokines and growth factors, including prostaglandin E2 (PGE2), IL-6, and colony-stimulating factor 1 (CSF-1), are released by tumor cells and cause M2 polarization in TAMs. Through the production of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), and epidermal growth factor (EGF), TAMs can stimulate tumor growth directly. Disruption of these signaling pathways can convert M2 macrophages into M1 macrophages, resulting in an alteration in the immune microenvironment [12]. In a recent investigation, Che et al. showed that polarization of M2 macrophages in TME induced by IL-33 successfully impeded the evolution of the tumor and GC’s peritoneal metastasis, improving the immunotherapy’s ultimate outcomes. Hence, IL-33 injection created a unique inflammatory environment locally, which can be used in conjunction with TAM reprogramming to modify TME to improve therapy effectiveness for peritoneal metastases [16].

Cancer-associated fibroblasts (CAFs)

Cancer-associated fibroblasts (CAFs) are activated fibroblasts within the TME that are a substantial stromal component in the tumor microenvironment and are vital to the growth of the tumor [7]. Bone marrow-derived stem cells, fibroblasts in healthy tissues, and certain cells surrounding arteries, such as pericytes, can provide these cells [17]. Studies carried out in vivo and in vitro have shown that CAFs promote tumor growth and metastasis. Platelet-derived growth factor (PDGF), TGF-β, VEGF, FGF, and interleukins are among the many chemicals secreted by activated CAFs, which are identified by α-smooth muscle actin (α-SMA). These substances regulate the formation of tumors and increase the rate at which CAFs infiltrate relative to normal fibroblasts [9]. Aside from CAF, stromal-derived factor 1 (SDR1), CXCL12 (C-X-C motif chemokine ligand 12), and interleukin 11 have also been identified as CAF-derived factors that promote GC migration and invasion [17]. Furthermore, research has demonstrated that CAFs overexpress several proteins, including FGF, IL-1β, TNF-α, TGF-β, and IL-6, which can promote the epithelial-mesenchymal transition (EMT) and hasten the invasion and metastasis of tumor cells [7, 17]. As a result of a complex reprogramming process known as EMT, epithelial cells become mesenchymal and lose their differentiation. This phenomenon occurs during embryonic development, tissue formation, wound healing, and carcinogenesis [18, 19]. There is a close relationship between the EMT phenomenon and GC invasion. In their study on GC subtypes, Cristescu et al. used exact molecular analysis to show a strong correlation between the poor prognosis of primary GC and the expression of the EMT gene signature [20]. Additionally, by activating the JAK2/STAT3 pathway in these cells, GC-derived CAFs can release high quantities of IL-6, which may speed up the migration of GC cells and enhance the EMT process [17]. It is also important to note that CAFs play an important role in maintaining the immune suppressive function of the TME. The IL-6 that CAFs produce inhibits the development of dendritic cells and instructs monocytes to become macrophages [21]. Additionally, CAFs are responsible for attracting macrophages to the TME as well as promoting their polarization towards the M2 type [17].

Tumor-infiltrating neutrophils (TINs)

TINs are neutrophils recruited to the tumor site, where they can exhibit either pro-tumor (N2) or anti-tumor (N1) phenotypes [22]. There is controversy regarding the role of neutrophils in tumor progression in the GC microenvironment. However, it is evident that this cell type contributes considerably to TME and has been related in multiple studies to poorer GC consequences [23]. High neutrophil proliferation seems to be detrimental to 5-year survival [9].

Additionally, TINs are reported to promote the migration and invasion of GC cells by activating the ERK pathway and inducing EMT, indicating they may strongly contribute to GC metastasis [24]. Neutrophils are widely recognized as a major source of IL-17a during inflammation. As a member of the IL-17 family, this cytokine plays an important role in inflammation and is prevalent in many cancers, including GC, where it promotes tumor growth and metastasis [25]. Recent research has found that IL-17a produced from TINs also activates the JAK2/STAT3 pathway, which promotes GC cell EMT [26]. Zhang et al. reported in a recent study that TOB1 expression within TME is essential to the GC’s potential for targeted immunotherapy. They reported that TOB1 may increase the survival rate of GC patients by causing neutrophils to become anti-tumor polarised, inhibiting their apoptosis, and improving the patient’s response to immunotherapy [27]. Lastly, the TIN population has been proposed as a valuable independent prognostic indicator and appears to be associated with the outcomes of GC.

Tumor-infiltrating lymphocytes (TILs)

The immune response to tumors is mediated by lymphocytes that infiltrate the tumor, including B cells, NK cells, and T cells, with T cells being indispensable for anti-tumor immunity [7, 28]. The T cell population includes several different cell types, such as helper cells (CD4+), cytotoxic T cells (CD8+), regulatory T cells (FOXP3+), and memory T cells [7]. A recent meta-analysis showed that among TILs, high levels of CD8+ lymphocytes were the most significant predictor of enhanced survival [29]. These lymphocytes can infiltrate the microenvironment of tumor cells and modulate the immune response against them, although their function can be suppressed by a variety of mechanisms such as abnormal expression of stress-inducible NKG2D (Natural Killer Group 2D) ligands, Inhibition of T cell function by gamma/delta T cells, which are immunosuppressive, decreased perforin, cytotoxins like TGF-β, and significantly, Cytotoxic T cells are affected by the activation of immune inhibitory pathways through programmed cell death protein 1 and its ligand (PD1/PDL-1) [30]. One of the most distinctive Treg markers is the transcription FOXP3, which is typified by the CD4+CD25+FOXP3+ phenotype. It is an essential intracellular molecule for Treg formation and activity. Due to Treg suppression of antitumor cytotoxic T cells in the tumor microenvironment and self-immune tolerance, high Treg infiltration in tumor tissues is associated with poor prognoses due to immune escape by tumor cells [29]. In GC tissue, Excessive FOXP3 expression was associated with lymph node metastases and a diminished survival rate in a prior investigation involving 100 GC specimens [31]. In addition, CTLA4 (cytotoxic T-lymphocyte-associated antigen4) has been recognized as a significant inhibitory mediator, especially in CD4 + Treg cells [30, 32]. Future studies and examining novel animal models will be required to pinpoint the precise pathways underlying Tumor immunity mediated by TIL in GCs.

Dendritic cells (DCs)

In addition to their morphological and functional similarities, dendritic cells are a diverse group of cells characterized by considerable flexibility. DCs are antigen-presenting cells responsible for initiating and regulating adaptive immune responses. They are primarily divided into two main populations: plasmacytoid dendritic cells (PDCs) and myeloid dendritic cells (MDCs) [33]. Human MDCs, not PDCs, are the main IL-12 producers, and this fact seems to show the suppressor role of MDCs for tumor neovascularization and therefore suppress tumor growth. However, research has revealed that angiogenesis-stimulatory DCs, including plasmacytoid DCs, are common in the tumor environment while angiogenesis-inhibitory myeloid DCs seem missing [34]. This may be due to CXCL12, a chemokine produced by tumors that attracts and preserves plasmacytoid DCs in the TME. In response, these cells stimulate vascularization by releasing IL-8 and TNF-α [35]. Separate studies have shown that PDCs contribute to tumor vascularization and ICOS+ regulatory T cells (Treg) in the peripheral blood of GC patients, creating an immunosuppressive tumor microenvironment that assists cancer in evading the immune system [36].

According to our mentions above, some of the cells involved in the TME of GC are significant oncogenic drivers and are consequently linked to a poor prognosis; nevertheless, other cells exhibit anti-tumor action. Targeting the pro-tumorigenic stroma and promoting an anti-tumorigenic microenvironment may be practical and successful cancer treatment approaches.

Targeting small molecules in GC

Recent studies highlight the expanding potential of small molecule-targeted treatments in GC, with a particular focus on natural small molecules derived from natural sources. These molecules have shown promise in slowing cancer progression by targeting specific molecular pathways involved in oncogenesis, offering a more effective and less toxic alternative to traditional therapies. The growing interest in small compounds stems from their ability to improve therapeutic efficacy while minimizing adverse effects. Numerous strategies and substances have demonstrated potential in combating GC, underscoring the need for innovative drug development methods. Ultimately, the therapeutic potential of natural small molecules in GI cancers emphasizes their role in overcoming the limitations of current treatments and offering better treatment alternatives [37].

Tyrosine kinases, Fibroblast growth factors, PIM kinases, YAP/TAZ, and mTOR, among other small molecules that control the development, and invasion of GC might be suitable sites for targeted therapies [38,39,40,41]. Because of the intricacy of downstream signaling and the difficulty of inhibiting specific molecular interactions, not all GC-related small molecules can be effectively interfered with. Next, we want to discuss a few small molecules that have been proven to be appropriate targets for GC therapy in preclinical and clinical studies. Figure 2. shows an overview of regulatory cascades of small molecules involved in GC cell survival and proliferation.

Fig. 2
figure 2

An overview of regulatory cascades of small molecules involved in GC cell survival and proliferation; Tyrosine kinases, Fibroblast growth factors, PIM kinases, YAP/TAZ, and mTOR, among other small molecules that control the development, and invasion of GC cells and interaction between potential inhibitor agents are presented

Tyrosine kinase inhibitors (TKIs)

The human genome encodes a significant class of protein kinases called tyrosine kinases (TKs), which are vital for a variety of cellular functions such as cell division, differentiation, and death. Both normal cellular function and the onset of many illnesses, especially cancer and neurological disorders, depend on them. There are 90 different tyrosine kinase genes in the human genome, which are separated into 10 subfamilies of non-receptor tyrosine kinases (non-RTKs) and 20 subfamilies of receptor-type tyrosine kinases (RTKs). These enzymes have distinct structural traits that are necessary for their regulatory activities, such as a propensity to assume inactive conformations. Their significance in both physiological and pathological circumstances is highlighted by their varied roles in cellular signaling networks [42, 43].

RTKs relay the process of extracellular signal transduction into the cells by catalyzing phosphate transfer from ATP to tyrosine residues of intracellular proteins and non-RTKs function in intracellular signal transmission. Receptors of many growth factor families, such as Fibroblast growth factor (FGF), Platelet-derived growth factor (PDGF), Epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF), function through RTKs. Also, RTKs have important roles in metastasis and tumor growth, hence they are among the most frequently studied targeted therapies for different kinds of cancer [44, 45]. Upregulation of several known RTKs is observed in GC, which could make them excellent therapeutic targets [38]. So, small molecule receptor tyrosine kinase inhibitors (RTKIs), could be of significance in the process of GC treatment.

Apatinib is an RTKI that suppresses tumor angiogenesis by selectively targeting VEGFR-2. By interacting with the intracellular ATP binding portion of VEGFR-2, this orally bioavailable agent prevents phosphate transfer and the continuation of the subsequent signaling pathways. Also, Apatinib reduces angiogenesis by lowering the expression of VEGF through the suppression of PKM2, which is important for tumor development and metastasis [46, 47]. Apatinib, through suppressing angiogenesis, prevented the growth and metastasis of tumors, and several studies showed promising results of Apatinib in GC treatment [46]. Following preclinical in vivo and in vitro research that validated Apatinib’s effectiveness in treating GC [48, 49], many clinical studies have been done to investigate its antitumor potential in GC patients. In combination with SOX chemotherapy, Lin et al. demonstrated that Apatinib was an effective and protective neoadjuvant for locally advanced GC [50]. According to Su et al., apatinib targets c-kit signaling and its downstream pathways to reverse pyrotinib resistance in HER2+ GC [51]. Additionally, as a 1st line treatment, apatinib plus paclitaxel may prolong survival [52]. Compared to a placebo, apatinib significantly increased overall survival and progression-free survival in patients with advanced GC who did not respond to treatment [53]. Despite its promising safety profile, side effects like proteinuria, hand-foot syndrome, and hypertension, have been observed in patients participating in clinical studies [46].

Foretinib is another novel small molecule RTKI which has multiple targets including VEGFR2, RON, AXL, TIE-2, and MET. Foretinib hinders cell proliferation of cells amplified in MET and FGFR2 expression in multiple GC cell lines in preclinical studies [54]. Shah et al.‘s clinical study revealed that single-agent foretinib did not significantly improve tumor regression, even though it inhibited MET. Hence, Shah et al. suggest that Foretinib may be more effective against GC, as a combination therapy with other targeted therapy agents or chemotherapeutics [55]. Grojean et al. and Awasthi et al. then assessed the anti-tumor effects of foretinib in combination with albumin-bound paclitaxel or oxaliplatin and nanoparticle paclitaxel in two different investigations. Their results revealed that Foretinib’s simultaneous hinders of VEGFR2 and c-Met significantly enhanced the therapeutic effects of these chemotherapeutics in preclinical GC models and this therapeutic combination may improve GC patients’ survival [56, 57]. Additionally, Sohn et al. discovered that foretinib targets CD44 signaling to limit GC stemness in addition to hindering GC cell proliferation by targeting c-MET [58].

Crizotinib (PF-02341066) is also an RTKI agent that exerts antitumor action against GC by targeting MET. In vivo and in vitro, Okamoto et al. revealed that crizotinib upregulates proapoptotic Bcl2 family members and suppresses GC cell growth by targeting MET [59]. In addition, Lapatinib, an RTKI that targets EGFR/HER-2, was found to have anti-tumor effects in GC patients. The combination of Lapatinib with commonly used chemotherapeutic agents like cisplatin, paclitaxel, oxaliplatin, SN-38 (Irinotecan’s active metabolite) also, and 5-FU appears to be synergistic [60, 61]. Hence, Lapatinib could serve as an excellent adjuvant to currently used GC chemotherapeutic regimens. SAR125844 is another RTKI that targets MET. Egile et al. showed that SAR125844 in MET-amplified GC xenograft models caused tumor regression in a dose-dependent manner without adverse consequences of treatment [62].

In addition to the mentioned RTKIs, many other agents in this drug category are currently under investigation to evaluate their therapeutic efficacy against GC. A number of these agents are listed in Table 1. Overall, RTKIs have shown promising therapeutic potential in GC recently. More clinical studies in this field could establish RTKIs as a novel targeted therapy in treatment guidelines for GC.

Table 1 TKI agents under clinical investigation for the treatment of GC

Fibroblast growth factor receptors inhibitors

As part of the fibroblast growth factor (FGF) family, fibroblast growth factor receptors (FGFRs) bind the FGF molecules. There are 5 main types of FGFRs -FGFR1-5, and 23 FGF ligands that bind to these receptors. Activation of each FGFR by different types of FGFs results in triggering various downstream signaling pathways [63]. FGF/FGFR signaling could be relayed in the forms of endocrine, paracrine, or autocrine signaling. FGFs by binding to and dimerizing FGFRs, are involved in various physiological activities in the process of embryogenesis, regulation of metabolism, and maintaining homeostasis [64]. Furthermore, disruption of the FGF/FGFR signaling system may be linked to oncogenesis due to the critical roles this signaling route plays in controlling cellular migration, mitosis, and apoptosis. Two significant processes that may result in disruption of FGF/FGFR signaling throughout the oncogenesis process are the secretion of FGF ligands from tumor cells, which unhinderedly activates FGFRs, or mutations in FGFR genes, which activate this pathway ligand-independently. In addition, the overexpression of FGFR in certain cancers, including GC, is implicated in the pathogenesis of those diseases [39]. The most notable components of this signaling pathway that are dysregulated in GC are FGFR2 and FGF18. Due to FGFR2’s role in maintaining healthy stomach tissue, its interruption could lead to GC progression [65]. Amplification of FGFR2, which has been seen in 3–10% of primary GCs, is associated with poor survival rates [66]. Overexpression of FGF18 is the most prominent among the FGF family and could be used as a prognostic and diagnostic biomarker. Consequently, many therapeutic agents have attempted to target the FGF/FGFR signaling pathway in GC pathogenesis. mAbs and small molecule inhibitors targeting FGF/FGFR signaling are two main categories of drugs that could be used for this purpose. Small molecule inhibitors include non-specific RTKIs and selective RTKIs which specifically target FGFR [65]. Non-specific RTKIs were discussed accordingly in the previous section of the article. Here, we review the main selective RTKIs targeting FGFR which have the advantage of fewer off-target effects.

FGFR1-3 is the target of the selective small-molecule FGFR inhibitor AZD4547. AZD4547 caused apoptosis in GC cell lines SNU-16 and KATO-III, which overexpressed the FGFR2 gene, according to in vivo and in vitro investigations. Inhibition of GC tumor growth induced by AZD4547 was also more significant in cell lines that harbor FGFR2 amplification compared to nonamplified models. Also in vivo studies confirmed that AZD4547 enhanced the cytotoxicity of chemotherapeutic agents against GC cells [67]. However, the phase II ‘SHINE’ clinical study showed that AZD4547 did not considerably enhance progression-free survival compared to paclitaxel in individuals with FGFR2-amplified GC and maybe another biomarker should be chosen to select patients who may show a better therapeutic response [68]. Schmidt et al. carried out research to determine the effectiveness of BGJ398, a pan-FGFR inhibitor, on three GC cell lines. Their findings demonstrated that the expression levels of FGFR1 and FGFR2IIIc in GC cells determine the cytotoxicity of BGJ398. FGFR inhibition was most effective in inhibiting the invasion and growth of GC cells in KKLS cells, which express high levels of FGFR [69]. PRN1371 is an FGFR1-4 inhibitor that is effective against various solid tumors. In vitro studies confirmed an antiproliferative role of PRN1371 against the SNU-16 GC cell line [70]. Tsimafeyeu et al. designed Alofanib (RPT835), a selective FGFR2 inhibitor, using a computational molecular modeling protocol. Then, KATO-III, a FGFR2-amplified GC cell line, was significantly suppressed by Alofanib in subsequent in vitro experiments [71]. Another specific FGFR inhibitor, ARQ087, has the ability to cause FGFR2-amplified GC cell lines to undergo apoptosis and G1 phase cell cycle arrest [72]. LY2874455 is another FGFR inhibitor that targets FGFR1-4. The in vivo and in vitro antiproliferative effects of LY2874455 were observed in KATO-III and SNU-16 cell lines in a study by Zhao et al. [73]. Studies conducted in clinical settings, the effective half-life of LY2874455 was shown to be about 12 h. A total of 12 patients with stable disease and one patient with partial response were evaluated in this study out of 15 evaluable GC patients [74]. However, a case of acquired resistance to LY2874455 has been reported in a patient who had shown a long-term response to this drug through a potential mechanism of FGFR2-ACSL5 fusion protein [75]. Additionally, preclinical research on the mechanism of GC cell lines’ resistance to FGFR inhibitors showed that drug resistance may be caused by the quick reactivation of the mitogen-activated protein kinase (MEK) pathway in response to FGFR inhibition. And combinational use of inhibitors of the MEK pathway with FGFR inhibitors could be more effective in FGFR-amplified GC [76]. Table 2 lists additional FGFR inhibitors that are presently being studied clinically for the treatment of GC.

Table 2 FGFR inhibitors under clinical investigation for the treatment of GC

PIM kinase inhibitors

The class of serine/threonine kinases known as the Proviral Integration site for Moloney murine leukemia virus (PIM) kinases was named after the chromosomal loci where they were initially discovered. The PIM-3, PIM-1, PIM-2, and isoforms are the three main members of this family. Cell proliferation, growth, survival, and migration are all regulated by PIM kinases in different cell types. The expression of PIM kinases is virtually undetectable in normal tissues. However, numerous forms of human malignancies, including GC, have shown different expression patterns [77, 78].

A lower prognosis is linked to elevated expression of PIM-1 and PIM-3 in GC. There has been a correlation between lymph node metastases and GC patients who overexpress PIM-1. Overexpression of PIM-1 causes oncogenesis primarily through three mechanisms: apoptosis inhibition, cell proliferation promotion, and genomic instability promotion. Due to its oncogenic properties and overexpression in numerous cancers, PIM-1 may be a potential therapeutic target for the development of anticancer medications. The creation of PIM inhibitors that are selective against other kinases has advanced significantly in recent years [77, 79].

To assess the therapeutic effectiveness of targeting PIM-1 in GC, Yan et al. carried out a study. A comparison between normal gastric epithelial cells and GC samples showed that PIM-1 was aberrantly expressed. Also, the ratio of cytoplasmic to nuclear expression of PIM-1, which is correlated with poor survival rate and tumor invasion, is significantly higher in GC tissue samples. It was also demonstrated that K00135, a small molecule inhibitor targeting PIM-1, has cytotoxic effects against GC cell lines harboring PIM-1 overexpression. Altogether, PIM-1 as a possible therapeutic target in the treatment of GC was validated by this investigation [80]. AZD1208 is a PIM kinase inhibitor that targets PIM-1-3. This pan-PIM kinase inhibitor through induction of autophagy inhibits tumor growth and proliferative characteristics of tumor cells. Additionally, because PIM kinases play a part in the DNA damage repair pathway, AZD1208 causes tumor cells to accumulate DNA damage by blocking PIM-1-3. As a consequence, resistance to AZD1208 may result from the reactivation of DNA damage repair. Moreover, AZD1208 showed synergistic effects when used in combination with Akt inhibitors which could reverse drug resistance. Interestingly, expression levels of different PIM isoforms in GC cells are not correlated with response to AZD1208 treatment [78].

Resveratrol is a phytochemical first extracted from the Veratrum grandiflorum plant. Kim et al. demonstrated that resveratrol could function as a PIM kinase inhibitor by directly binding to PIM-1. Through induction of apoptosis, Resveratrol prevents SNU-601 GC cells from growing and proliferating. Resveratrol administration decreases PIM-1’s catalytic activity in a dose-dependent manner even though it does not affect PIM-1 expression levels. Resveratrol is also an effective inhibitor of PIM-2, but its effectiveness against PIM-3 is much weaker [81]. Overall, though most of the studies involving PIM kinase inhibitors assessed the efficacy of these agents against hematologic malignancies, recently some preclinical studies have shown the effectiveness of PIM kinase inhibitors against GC. Further clinical studies are needed to evaluate their effectiveness in GC patients as potential anticancer drugs.

YAP/TAZ inhibitors

The primary mediators of the Hippo signaling pathway in human cells are transcriptional coactivators with PDZ-binding motif (TAZ) and yes-associated protein (YAP) [82]. The Hippo pathway, which is conserved from Drosophila to humans, controls organ growth, tissue homeostasis, and carcinogenesis [83]. In response to activation of the Hippo pathway, phosphorylation of several serine residues results in the degradation of YAP and TAZ by the proteasome. Dephosphorylated YAP/TAZ reaches the nucleus and increases the expression of invasion and proliferation mediators [84]. Aside from its role as a downstream effector of the Hippo pathway, YAP has a role in several important carcinogenic pathways, including the mTOR and WNT pathways, which enhances its influence on cancer development [40, 85]. YAP/TAZ expression is typically upregulated in several cancers, such as the lung, breast, liver, and GC. Only minor YAP expression could be seen in the normal proliferating cells of the gastric mucosa in adults, but YAP expression is elevated in both metastatic and primary GC [40].

YAP and TAZ are identified as attractive therapeutic targets for the treatment of cancer due to their elevated expression in malignant cells relative to normal tissues and their crucial function in several stages of cancer cell survival and drug resistance [86, 87]. Since lymphatic invasion and a significantly lower overall survival are strongly associated with overexpression of YAP, it is possible that YAP could be used as a prognostic biomarker for GC. In addition, suppressing YAP inhibits proliferation, invasion, and metastatic spread in several GC models, suggesting it could be used as a therapeutic target in the treatment of this disease [88, 89].

Many studies have been done in an attempt to treat GC targeting YAP/TAZ. Giraud et al. showed that Verteporfin, a benzoporphyrin derivative, reduced the proliferative properties of GC stem cells by regulating Hippo/YAP signaling. Verteporfin caused anti-GC effects by inhibiting YAP expression, binding to YAP selectively and changing its structure, and interfering with YAP interactions with other molecules [90]. VGLL4 is a peptide that competes with YAP in binding to its target TEAD. Jiao et al. synthesized a VGLL4 mimetic named Super-TDU which reduced YAP-TEAD interaction by competitively binding to TEAD. Super-TDU inhibited GC cell growth and proliferation by this mechanism both in vivo and in vitro [91]. Sitagliptin which is an oral anti-diabetic agent has recently shown some anti-cancer effects. Wang et al. showed that Sitagliptin inhibited YAP by inducing its phosphorylation and inhibiting its translocation to the nucleus. Additionally, via controlling YAP, sitagliptin suppressed the production of tumor-testis antigen Melanoma-associated antigen-A3. Furthermore, sitagliptin generally inhibits GC by controlling AMPK/YAP/melanoma-associated antigen-A3 signaling [92].

In addition to agents directly targeting YAP, upstream regulators of Hippo/YAP signaling by inhibiting YAP expression could be important targets in GC treatment. Tang et al. revealed that an STRN3-containing PP2A complex dephosphorylates MST1/2 kinases, activating YAP and blocking the Hippo pathway. They synthesized a peptide called STRN3-derived Hippo-activating peptide (SHAP) that suppresses YAP activation by blocking the STRN3-PP2A interaction. SHAP has shown anti-GC effects and inhibited GC progression in vivo [93]. Song et al. evaluated the efficacy of tertiary amide derivatives incorporating benzothiazole moiety against GC. Some of their developed compounds which were named F1-F14 have shown antiproliferative effects against GC cells. In particular, compound F10 suppressed YAP via activating the Hippo signaling pathway and also induced apoptosis, and inhibited GC cell proliferation in vitro [94]. According to Seeneevassen et al., the Hippo signaling pathway reduces YAP/TAZ accumulation in GC stem cells when leukemia inhibitory factor (LIF) is stimulated. Also LIF by inhibiting YAP/TAZ mitigated chemoresistance and proliferative characteristics of GC stem cells [95]. Also, Ursolic Acid (UA) has shown anti-GC effects via indirectly targeting YAP/TAZ. Kim et al. showed that UA by inducing RASSF1 expression, activated the Hippo signaling pathway and inhibited YAP accumulation in GC cells, and mitigated proliferative and invasive characteristics of GC cells [96]. Essential oils extracted from the Pinus Koraiensis (EOP) plant have also shown anti-GC effects by targeting YAP. Zhang et al. demonstrated that EOP by inhibiting YAP expression mitigated proliferative and invasive characteristics of GC cells. Apoptosis has been demonstrated to occur when EOP is applied to GC cells in vitro, as well as cell cycle arrest in the G2/M phase when EOP is applied to these cells in vitro [97]. In another similar study, Ye et al. reported that CL-6, a derivative of commonly used phytochemical curcumin, suppressed proliferation and invasion and induced apoptosis in GC cells. In vitro analysis showed that regulation of Hippo/YAP signaling could be involved in these effects [98]. Overall, YAP inhibitors offer a variety range of therapeutic applications, although further studies on their effects and toxicity in GC are required.

mTOR inhibitors

The primary effector of the mTOR/PI3K/AKT signaling pathway is the mammalian target of rapamycin (mTOR) [99]. Since a range of upstream molecules can activate it and act as a junction for multiple signaling pathways, mTOR is one of the most autonomous elements of this axis [100]. mTOR regulates angiogenesis, cell survival, cell metabolism and by enhancing the expression of anti-apoptotic proteins, nutrient transporter proteins, and HIF-1/HIF-2 [101].

The regulation of GC cells’ development, uncontrolled proliferation, and differentiation may be influenced by overactive mTOR [41]. In 60–80% of stomach adenocarcinomas, inappropriate mTOR activation has been discovered at various stages of the disease [102, 103]. Mutations in upstream regulators including PTEN, PI3K, and EGFR are primarily responsible for activating mTOR and associated signaling pathways in GC [100]; However, nothing is known about the inherent alterations of this protein kinase in GC. Genetic mutations or other effectors may be responsible for mTOR’s activation in GC patients, but regardless, mTOR phosphorylation is linked to several clinical and prognostic aspects [104, 105]. Based on the results of a cohort study on GC patients, it appears that tumor growth and 5-year survival are closely related to the phosphorylation of this protein kinase [105]. Thus, several selective inhibitors of mTOR may be effective in treating GC due to its fundamental role in GC pathogenesis.

The multi-protein complexes mTORC1 and mTORC2 are identified as two unique subtypes of mTOR. According to this classification, 2 generations of mTOR inhibitors have been developed; The first generations of mTOR inhibitors have only The capability to suppress mTORC1 and include agents like rapamycin, temsirolimus, and everolimus. However, the second-generation mTOR inhibitors are effective against both mTORC1 and mTORC2. For instance, AZD8055 and AZD2014 are two second-generation mTOR inhibitors that have shown promising results against HER-positive and TSC1/2 mutated or null GC cells, respectively [99].

Rapamycin was first used as an antifungal agent. Numerous studies have been conducted to assess Rapamycin’s effectiveness against GC due to its anticancer properties. Matsuzaki et al. showed that Rapamycin and Temsirolimus (CCI-779) could act as a chemosensitizer of GC cells to 5-FU treatment. These mTOR inhibitors by decreasing IC50 of 5-FU against GC cells increased the apoptotic rate after 5-FU treatment. So, because of the synergistic effects of these agents, mTOR inhibitors could be used as an adjuvant to 5-FU in chemotherapeutic regimens of GC patients [106]. Rapamycin also by inhibition of mTOR inhibits lymphangiogenesis of gastric tumors. Chen et al. showed that GC cells’ Akt/mTOR inhibition dramatically reduced the expression levels of VEGF-C and VEGF-D. And inhibition of the Akt/mTOR-VEGF-C/VEGF-D axis by Rapamycin could be of significance in the treatment of GC [107].

A Food and Drug Administration (FDA)-approved derivative of rapamycin, everolimus (RAD001) is used to treat a variety of tumors, including breast cancer and renal cell carcinoma [108]. Despite preclinical in vitro studies confirming some anti-GC roles of Everolimus, a phase II clinical trial failed to show an Important response of patients with advanced GC to this agent as an adjuvant to the chemotherapeutic regimen [109]. In a different study, Fukamachi et al. demonstrate that everolimus and another mTOR inhibitor, temsirolimus, are both effective only against a special subset of diffuse-type GC which harbors PIK3CA mutation and may be developed from intestinal-type GCs [110]. Additionally, Park et al. reported a successful experience using everolimus as salvage treatment in a patient with metastatic GC who had PIK3CA and pS6 abnormalities. This case study also highlighted how crucial it is to use these biomarkers to forecast how well GC patients will respond to everolimus treatment [111]. Against GC, BEZ235, a dual PI3K/mTOR inhibitor, has demonstrated greater therapeutic success than everolimus. Also, the effectiveness of BEZ235 against paclitaxel-resistance GC could be important as a part of a strategy to treat drug-resistant GC targeting the PI3K/Akt/mTOR pathway [112].

Some medications that are not commonly classified as anti-cancer agents and some natural products by exerting inhibitory effects against mTOR and upstream regulators of mTOR have shown anti-GC properties. Mefloquine, an FDA-approved antimalarial drug, has shown synergistic effects with paclitaxel against GC cells by inhibiting phosphorylation of mTOR and other components of the PI3K/mTOR/Akt pathway. Mefloquine and Paclitaxel together increase apoptosis and suppress the proliferation of GC cells [113]. According to Tang et al., Tigecycline, a glycylcycline class antibiotic, inhibits mTOR phosphorylation through activation of the AMPK pathway, thereby inducing autophagy in GC cells and inhibiting cell growth [114]. Salidroside is a phytochemical extracted from the Rhodiola rosea plant. Rong et al. reported that Salidroside through inhibition of expression of mTOR induced autophagic changes in AGS GC cells. In vivo and in vitro, Salidroside also induced apoptosis and inhibited cell growth in GC cells [115]. N-butylidenephthalide (NBP) is a bioactive compound extracted from the Radix Angelica Sinensis plant. In a case-control study, Liao et al. found that GC patients taking herbal medications containing NBP were more likely to survive and had a lower mortality rate. Then, the in vitro studies that have been undergone to clarify the molecular mechanism of anti-GC effects of NBP showed that NBP by enhancing the expression of REDD1 inhibited the mTOR signaling, induced apoptosis, and inhibited proliferation of GC cells [116].

Immunotherapy-based immune checkpoint inhibitors (ICIs)

GC patients now have a more comprehensive treatment landscape because of newly developed treatments like immunotherapy with ICIs [117]. Since immunotherapy’s unique and long-lasting anti-cancer effects have been extensively researched, it has been widely accepted as a successful cancer treatment approach. Immunotherapy through ICI as a promising strategy in cancer treatment can prevent the recurrence of tumors [118]. Various methods to identify immunosuppressive components that thwart anti-tumor immune responses are necessary for significant advancement in cancer immunotherapy. To prevent immunological damage, immuno-inhibitory molecules often suppress immune responses by adversely altering immune cell communication pathways. Nevertheless, Cancer immunity is suppressed during tumor formation when inhibitory immunological checkpoints are activated within immune cells [119]. Programmed cell death protein-1 (PD-1)/programmed death ligand-1 (PD-L1) and Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) are considered as two main co-inhibitory immune checkpoints. The inhibition of the binding of ligands to checkpoint receptors is a factor contributing to the reactivation of immunity in cells [51, 120, 121]. Hence, In the sections that follow, we will concentrate on the clinical advancements of ICIs and their combination with targeted therapy for GC.

Targeting immune checkpoint in GC

To date, the use of mAbs as ICIs has received remarkable regard for cancer therapy. T-cell activation is suppressed via the inhibitory signals by different immune checkpoint molecules in interaction with their cognate receptors such as PD-L1/PD-1 axis, CD28 pathway, and CTLA-4 or cytotoxic T-lymphocyte antigen 4 [122]. Antigen-presenting cells (APCs) attach CTLA-4 to B7 ligands, preventing B7 interaction with CD28 receptors on TCD4s. As a result, CTLA-4 blocks the CD28 stimulatory signal from reaching T cells in particular, anti-CTLA-4 antibodies bind to CTLA-4 and inhibit T-cell suppression. Ipilimumab and tremelimumab are the two main antibodies used as anti-CTLA-4 antibodies [122, 123]. Ipilimumab, the first anti-CTLA-4 human monoclonal antibody (IgG1), was authorized by the FDA in early 2011 to treat patients with metastatic melanoma [124]. Additionally, By blocking the interaction between CTLA4 and its ligand, tremelimumab, a monoclonal antibody that is completely human, increases cellular immunity [125]. A phase Ib/II trial study found that 12 patients with GC and gastroesophageal junction (GEJ) cancer who received tremelimumab as a second-line treatment after chemotherapy had a median overall survival (OS) of 7.7 months and a median progression-free survival (PFS) of 1.7 months [126]. Ralph et al. carried out another trial on the use of tremelimumab as a second-line treatment for patients with metastatic gastric and esophageal adenocarcinomas in order to block CTLA-4. The results demonstrate that one patient experienced a remarkably long-lasting benefit for this poor prognosis condition, despite tremelimumab’s low response rate. Increased proliferative responses to pertinent tumor-associated antigens are also indicated by the in vitro results. In conclusion, their study showed that tremelimumab can be effective when combined with antigen-targeted therapy and warrants further investigation [127]. According to the evidence, further investigations are needed to identify the benefit of tremelimumab as monotherapy or combination therapy in GC treatment.

As a negative costimulatory immunological checkpoint, the surface receptor PD-1 contributes to the suppression of T cell anti-tumor activity and helps tumor cells evade the immune response [122]. As a PD-1 ligand, PD-L1 is expressed on APCs as well as on different types of tumor cells. PD-1 and PD-L1 interact to induce immunosuppressive signaling pathways, suppress T-cell activity, and escape tumor cells [51]. Inhibiting anti-tumor immunity and increasing chemoresistance are effects of elevated PD-L1 expression in human malignancies [118]. Noteworthy, According to the findings, patients with GC bear a significantly high level of PD-L1 expression [128]. The PD-1 antibody pembrolizumab showed promise in treating patients with advanced GC in 2016, according to the phase Ib (KEYNOTE-012) trial. According to this study, GC patients with positive PD-L1 expression can experience anti-tumor immune responses from pembrolizumab [129]. The FDA originally authorized nivolumab, an anti-PD-1, in 2014 for the treatment of melanoma patients [130]. In patients with advanced GC who have received two or more chemotherapy regimens, nivolumab may improve overall survival (ONO-4538-12, ATTRACTION-2). According to the phase III trial, the nivolumab group’s median overall survival was 5.26 months, whereas the placebo group’s was 4.14 months. These results suggest that nivolumab may be a novel therapeutic option for patients with advanced GC who did not respond to earlier chemotherapy therapies [131]. Combining anti-PD-1 antibodies and anti-CTLA-4 considerably reduced invasion, inhibited migration, epithelial-mesenchymal transition (EMT), cell proliferation, and induced apoptosis in MGC-803 and MKN-45 cells. Moreover, the activation of signaling pathways such as MAPK, β-catenin, and PI3K/AKT was inhibited by CTLA-4 and PD-1-blocking antibodies. Consequently, anti-CTLA-4 and anti-PD-1 antibody combination therapy produced encouraging outcomes in GC patients [132]. Additionally, a phase I/II CheckMate 032 study showed encouraging clinical outcomes for patients with advanced (adv)/metastatic chemotherapy-refractory (CTx-R) GEJ cancer or gastric (G), esophageal (E) cancer when nivolumab (as anti-PD-1) was used alone or in combination with ipilimumab (as anti-CTLA4) [133]. Furthermore, ICI therapy may improve some but not all survival endpoints for patients with advanced or metastatic G/GEJ cancer, suggesting a small benefit with fewer side effects, according to a systematic review and meta-analysis study. Patients with PD-L1 positivity, MSI-H, EBV positivity, or a high tumor mutational burden have been found to benefit more from anti-PD-1/PD-L1 therapy [134].

A few individuals with cancer may experience negative effects from using blocking antibodies to target immunological checkpoint members, which could limit their treatment options. Adverse effects are one of the challenges and restrictions in front of ICI therapies [118, 122]. IrAEs or immune-related adverse events are immune-related diseases such as endocrinopathies, pneumonitis, colitis, hepatitis, and rash. All things considered, the immune system’s capacity to fight malignancies like GC can be enhanced by antibodies that target immunological checkpoint proteins. As a result, irAEs must be managed and treated in order to decrease, and using immunosuppressive medications is a good way to do so [118].

Combination approaches of ICIs and targeting therapy in GC

Although immune checkpoint therapy generally has satisfactory results, due to limitations in tumor immunity some patients do not achieve the desired outcomes. Therefore, combining techniques designed for different targets increases the development of various mechanisms, which enhances response rates [119]. Recent strategies to use targeted therapy plus mAbs have attracted considerable regard in several cancer treatments. The primary candidates for GC therapy in conjunction with ICIs are VEGF/VEGFR inhibitors and anti-HER2 mAbs [51]. Before this research, trastuzumab, an anti-HER2-monoclonal antibody, was the first-line treatment of choice for advanced GC patients with HER2-positive tumors [51]. A study by Rha et al. at 2020 proposed that combining chemotherapy with anti-PD-1 and HER2 targeting results in a considerable reduction of GC tumor size. For patients with advanced GC who tested positive for HER2, pembrolizumab, trastuzumab, and chemotherapy were used as the 1st line triple therapy [135]. Additionally, another study looked at the efficacy and safety of pembrolizumab in combination with chemotherapy and trastuzumab for patients with HER2-positive 1st line metastatic oesophagogastric (GEJ, or G, E) cancer. According to this trial, pembrolizumab is safe when combined with trastuzumab and chemotherapy, and it shows promise in treating HER2-positive metastatic EC [136].

In addition, a phase III study (KEYNOTE-811) evaluated the combination of trastuzumab and chemotherapy for patients with unresectable or metastatic GC or GEJC who were HER2-positive (NCT03615326). The results of the trial showed that pembrolizumab plus trastuzumab and chemotherapy greatly increases the objective response rate, significantly diminishes tumor size, and causes full responses in certain patients [137]. The tolerability, safety, and anticancer efficacy of margetuximab with pembrolizumab were evaluated in a single-arm, phase Ib–II trial (CP-MGAH22–05) in patients with HER2-positive gastro-oesophageal adenocarcinoma who had received prior treatment. The results of this study demonstrated that margetuximab and pembrolizumab had synergistic anticancer efficacy [138].

The completely humanized monoclonal antibody ramucirumab blocks VEGFR-2. As a second-line treatment for GC or GEJ adenocarcinoma, ramucirumab is approved using either alone or with paclitaxel. There are many current studies examining the Ramucirumab function when combined with new agents in both metastatic and perioperative situations. In particular, the combination of ramucirumab with ICIs is being studied in several active clinical trials [139]. GC/GEJC, hepatocellular carcinoma (HCC), and non-small-cell lung cancer (NSCLC) patients were studied in an open-label, phase Ia/b study using ramucirumab (an anti-VEGFR2 IgG1) combined with durvalumab (an anti–PD-L1 IgG1). The safety of durvalumab and ramucirumab was in line with the established safety profiles of individual treatments. The combination had an antitumor effect in all cohorts, especially in patients with elevated PD-L1 expression exhibited the strongest effects. These findings suggest that VEGF/VEGFR inhibitor combinations may be potent treatment options for advanced GC [140].

CAR T cell engineering

An additional type of immunotherapy known as CAR T Cell (chimeric antigen receptor) therapy, has recently attracted considerable interest among oncologists and researchers. While the FDA has approved mainly CAR T cell treatments for hematologic cancers, including lymphomas, leukemias, and multiple myomas, they appear to be among the most effective treatments for solid tumors, and a rising number of clinical trials are now emphasizing solid tumors like GC [141]. The use of animal xenotransplantation has shown that CAR T-cells have antitumor effectiveness and prolonged activity against GC in vivo and in vitro [142].

This immunotherapy approach involves isolating the patient’s peripheral blood T cells and genetically modifying them to express chimeric antigen receptor (CAR) genes by plasmid transfection, mRNA, or viral vectors; The T cells can now identify tumor-associated antigens because of this alteration [143]. A CAR consists of three components: an antigen-binding domain located extracellularly, a transmembrane spacer, and an intracellular signaling/activation domain [144]. A recombinant polypeptide called a single-chain variable fragment (scFv) is created when the variable light (VL) and variable heavy (VH) chains of mAbs are connected by a flexible linker to produce the extracellular domain. Without the limitations of MHC restriction, this fragment enables direct binding to tumor-associated surface antigens. The transmembrane domain, which is mostly generated from immunoglobulin G4 or CD8 molecules, is essential for strengthening the stability of the chimeric antigen receptor (CAR) and creating a structural connection between the endodomain and ectodomain. In turn, the intracellular domain mediates intracellular signal transduction required for cellular activation and guarantees strong membrane attachment of the CAR [143, 144]. The Fc receptor motifs or the CD3 chain mediate the first intracellular signals for T-cell activation. Furthermore, co-stimulatory signaling domains like CD28, CD134 (OX40), CD137 (4-1BB), or DNAX-activating protein 10 (DAP10) are frequently included in the endodomain. These co-stimulatory factors boost effector functions, cytokine generation, and T-cell proliferation. Numerous generations of chimeric antigen receptors (CARs) have had their integration thoroughly studied, leading to notable improvements in CAR performance and design [145]. From its initial design, which only contained the CD3 signaling domain (referred to as a “first-generation CAR”), CARs have undergone structural changes, evolving into more sophisticated variants that include co-stimulatory endodomains. Second-generation CARs, which combine CD3 with either the 4-1BB or CD28 signaling domains, and third-generation CARs, which combine CD3 with both the 4-1BB and CD28 signaling domains, have been developed as a result of this progression and exhibit enhanced T cell proliferation and persistence [146]. An outline of CAR-T cell therapy’s development and clinical application procedure is shown in Fig. 3.

Fig. 3
figure 3

A summary of CAR-T cell therapy construction and the clinical application process. (1) Obtaining Blood from the patient, (2) CAR gene insertion, (3) Antibody receptor expression, (4) CAR-T Cell Expansion, (5) Pharmaceutical interventions, (6) Injections of CAR-T cells

An overview of ongoing CAR T cell trials in GC

As previously noted, CAR-T cell therapy has received approval for the treatment of specific hematological malignancies; however, its application in the management of solid tumors remains unapproved. Target selection is one of the most critical challenges and obstacles to implementing Therapy with CAR-T cells for solid tumors. The majority of solid tumors are of epithelial origin, resulting in tumor antigens often being expressed on the surface of healthy epithelial cells. Ideally, tumor-associated antigens (TAAs) should exhibit selective expression on tumor cells to minimize the risk of off-target effects. Failure to achieve this specificity can lead to suboptimal therapeutic outcomes and potentially severe or fatal off-target toxicities [145]. For example, mesothelin, one of the antigen targets, is found not only on the surface of mesothelioma cells but also on pericardial and pleural surfaces [147]. The development of CAR T-cell immunotherapy for GC is a major problem since tumor cells can evade host immune surveillance due to the incredibly varied surface antigen expression in GC [148].

The high death rate of GC and the development of drug resistance in existing treatments highlight the need to develop CAR T-cell immunotherapy as a possible treatment option for specific patients. There is mounting evidence linking GC patients who overexpress human epidermal growth factor receptor 2 (HER2) to a poorer prognosis and a more severe disease profile. Research has demonstrated that CAR-T cells, which target HER2 antigens in an MHC-independent way and exhibit strong and specific cytotoxic activity, can successfully eliminate patient-derived HER2-positive GC cells [142, 149]. Not only HER-2 antigen but also in a preclinical investigation, CAR T cells that were directed against claudin18.2 (CLDN18.2) demonstrated promise efficacy against GC [150]. Additionally, The surprising effectiveness of autologous Claudin18.2-targeted CAR T cells (CT041) in treating a patient with metastatic GC that had previously advanced following four cycles of systemic chemotherapy and immunotherapy was reported in a recent case study by Botta et al. The patient demonstrated significant CAR T-cell expansion, a radiologic response, and marked reductions in tumor-informed circulating tumor DNA (ctDNA). These results underline the therapeutic potential of using CAR T-cell treatment to target Claudin18.2-positive GC and demonstrate the usefulness of ctDNA as a biomarker in CAR T-cell therapy [151]. Moreover, according to studies on CEA-specific CAR-T cell treatments, employing CAR T-CEA cells enables mice with advanced GC to survive longer and suppress tumor growth, especially recombinant therapy with IL-12 can improve CAR-T cells’ anti-cancer function [6].

Therefore, a variety of clinical trials have identified the following as critical markers for the diagnosis and management of GC: HER2, claudin 18.2 (CLDN 18.2), mesothelin (MSLN), folate receptor 1 (FOLR1), carcinoembryonic antigen (CEA) and other markers which are listed in Table 3 of ongoing clinical trials [150, 152, 153].

Table 3 ClinicalTrials.gov-registered CAR T cell treatment trials for GC

Challenges of CAR T cell therapy

Incidence of toxicity is the major obstacle to the development and progress of CAR T-cell therapy for GC patients; the most frequent and serious side effect linked to CAR T-cell therapy is cytokine release syndrome (CRS), a form of non-antigen-specific toxicity that can result in respiratory distress syndrome and multiple organ failure [145]. This toxicity arises as a result of the rapid and improper activation of several cytokines, including TNF α, IL 1, IL 8, IL 6, IFN β, and IFN γ [154]. Additional adverse effects that have been documented include cytopenia, tumor lysis syndrome, off-target/on-target toxicities that can cause multi-organ damage, and neurotoxicity, commonly referred to as CAR T-cell–related encephalopathy syndrome (CRES) [155]. These adverse events can become serious if not treated promptly, but they are manageable and reversible with the right treatment options like anti–IL–6 receptor antagonists and corticosteroids [155]. Therefore, a full awareness of the related adverse events particularly their grading and management is vital for advanced practitioners responsible for the care of patients receiving these therapies.

Antibody-drug conjugates (ADCs)

Monoclonal antibody-drug conjugates (ADCs) are biological agents composed of mAbs attached to cytotoxic payloads by chemical linkers [156]. According to the mechanism, the ADC enters tumor cells and delivers a cytotoxic payload that kills the cells. On the surface of cancer cells, specific receptors such as mesothelin, HER2, and guanyl cyclase C (GCC) are overexpressed, which leads to this effect [157]. Because of ADC’s tumor selectivity, the maximum tolerated dosage may be increased while the therapeutic dose is lowered, hence improving the therapeutic window and minimizing off-target damage [158]. While most ADCs have been developed to treat hematological malignancies, new ADCs targeting HER2, such as trastuzumab deruxtecan and trastuzumab emtansine, are showing promising results in solid tumors as well [157].

The GATSBY study was the first phase II/III study to assess an ADC’s efficacy in terms of GC. Patients with metastatic GC who had received prior treatment were evaluated by GATSBY for the effectiveness of trastuzumab emtansine. The trial’s findings indicate that trastuzumab emtansine does not provide more benefits than taxane therapy for patients with HER2-positive GC who have already had treatment [159]. An ADC consisting of a cytotoxic topoisomerase I inhibitor, a linker, and an anti-HER2 antibody, called trastuzumab deruxtecan (DS-8201), is known as a HER2-targeted ADC. Shitara et al. investigated the efficacy of this ADC in individuals with advanced GC who tested positive for HER2. When compared to patients receiving standard regimens, patients with HER2-positive GC who underwent trastuzumab deruxtecan therapy showed considerable improvements in response and long-term survival. The two most noteworthy harmful consequences were interstitial lung disease and myelosuppression [160].

Advanced gastrointestinal malignancies have also been studied in relation to ADCs that target proteins other than HER2, such as guanylyl-cyclase-c (GCC), mesothelin, and carcinoembryonic antigen associated CEACAM5 or cell adhesion molecule 5 [161, 162]. ADCs targeting targets other than HER2, however, have not yet demonstrated clinically significant efficacy and need more investigation. Table 4 lists a number of ADCs that are under active investigation for GC.

Table 4 ADCs under clinical investigation for the treatment of GC

Conclusion and future perspective

Overall, the manuscript highlights the heterogeneity of the GC microenvironment and the potential for targeted therapies and immunotherapies to improve treatment outcomes. Preclinical and clinical research has indicated promise in targeting particular molecules implicated in angiogenesis, immune evasion, and tumor-stroma interactions. Our article highlights that immunotherapy has greatly expanded treatment options for GC (GC), particularly through the use of ICIs like pembrolizumab and nivolumab. These therapies show promise in patients with high PD-L1 expression and specific biomarkers, and combination treatments with anti-PD-1 and anti-CTLA-4 antibodies have also demonstrated encouraging results. Moreover, targeted therapies addressing key molecular pathways, including RTKs, FGFR signaling, PIM kinases, and YAP/TAZ inhibitors, are emerging as additional options for GC treatment. Ongoing research is exploring other molecular targets, including the EGFR, MET, and more features of FGFR pathways, to further expand the repertoire of targeted therapies for GC. While several of these agents have shown preclinical promise, clinical trials have highlighted challenges such as limited survival benefits and the development of drug resistance, emphasizing the need for ongoing research to optimize their use.

Additionally, CAR-T cell therapy represents a promising strategy, although its application in GC faces challenges related to target selection and the risk of off-target effects. CAR-T cells targeting antigens like HER2 and claudin18.2 have shown potential in preclinical studies and clinical trials, with markers like HER2 and CEA emerging as key targets for treatment. Antibody-drug conjugates (ADCs) have also garnered attention in GC treatment. These agents combine mAbs with cytotoxic drugs to target overexpressed tumor cell receptors such as HER2, mesothelin, and guanylyl cyclase C (GCC), thereby enhancing treatment specificity. While new HER2-targeted ADCs like trastuzumab deruxtecan have shown promising results in HER2-positive GC, others targeting different markers such as GCC and mesothelin have shown limited clinical success and need further investigation. The future perspective for GC treatment lies in personalized medicine approaches that take into account the heterogeneity of the GC microenvironment and the molecular characteristics of individual tumors. Optimizing the sequencing and combination of targeted therapies and immunotherapies will be essential to maximize treatment efficacy.

The narrative nature of our review allows for a subjective selection of studies, which may lead to potential biases in the representation of the literature. Due to the broad scope of recent advances in GC treatment, it is possible that not all relevant studies were included, which may limit the comprehensiveness of our findings. The included studies vary in methodology and quality, which may affect the reliability of the conclusions drawn from the synthesized information. Unlike systematic reviews, our narrative approach does not provide quantitative assessments or meta-analyses, which could have offered more robust insights into the effectiveness of targeted therapies.

In conclusion, this manuscript offers a detailed synthesis of the current knowledge regarding the GC microenvironment, emphasizing the promise of targeted therapies and immunotherapies in enhancing treatment efficacy. Continued investigation, coupled with well-designed clinical trials, will be essential to advancing personalized therapeutic strategies and improving prognostic outcomes for GC patients.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

APCs:

Antigen-presenting cells

CAFs:

Cancer-associated fibroblasts

CAR:

Chimeric antigen receptor

CEA:

Carcinoembryonic antigen

CLDN 18.2:

Claudin 18.2

CRES:

CAR T-cell–related encephalopathy syndrome

CTLA-4:

Cytotoxic T lymphocyte-associated antigen 4

CXCL12:

C-X-C motif chemokine ligand 12

DCs:

Dendritic cells

EC:

Esophageal Cancer

EGF:

Epidermal growth factor

EMT:

Epithelial-Mesenchymal Transition

FDA:

Food and Drug Administration

FGF:

Fibroblast growth factor

FOLR1:

Folate receptor 1

GC:

Gastric cancer

GEJ:

Gastroesophageal junction

GEJC:

Gastroesophageal junction cancer

HCC:

Hepatocellular carcinoma

HGF:

Hepatocyte growth factor

ICIs:

Immune checkpoint inhibitors

IrAEs:

Immune-related adverse events

mAbs:

Monoclonal antibodies

MDCs:

Myeloid dendritic cells

MSLN:

Mesothelin

NKG2D:

Natural Killer Group 2D

NSCLC:

Non-small-cell lung cancer

OS:

Overall Survival

PD-1:

Programmed cell death protein-1

PDCs:

Plasmacytoid dendritic cells

PDGF:

Platelet-derived growth factor

PD-L1:

Programmed death ligand-1

PFS:

Progression-Free Survival

RTKs:

Receptor tyrosine kinases

SDR1:

Stromal derived factor 1

TAMs:

Tumor-associated macrophages

TILs:

Tumor-infiltrating lymphocytes

TINs:

Tumor-infiltrating neutrophils

TKIs:

Tyrosine kinase inhibitors

TME:

Tumor microenvironment

VEGF:

Vascular endothelial growth factor

References

  1. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians; 2024.

    Google Scholar 

  2. Chi H-C, Tsai C-Y, Wang C-S, Yang H-Y, Lo C-H, Wang W-J, et al. DOCK6 promotes chemo-and radioresistance of gastric cancer by modulating WNT/β-catenin signaling and cancer stem cell traits. Oncogene. 2020;39(37):5933–49.

    Article  CAS  PubMed  Google Scholar 

  3. Sibertin-Blanc C, Ciccolini J, Norguet E, Lacarelle B, Dahan L, Seitz J-F. Monoclonal antibodies for treating gastric cancer: promises and pitfalls. Expert Opin Biol Ther. 2016;16(6):759–69.

    Article  CAS  PubMed  Google Scholar 

  4. Liu X, Meltzer SJ. Gastric cancer in the era of precision medicine. Cell Mol Gastroenterol Hepatol. 2017;3(3):348–58.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Patel TH, Cecchini M. Targeted therapies in advanced gastric cancer. Curr Treat Options Oncol. 2020;21(9):1–14.

    Article  Google Scholar 

  6. Yang L, Wang Y, Wang H. Use of immunotherapy in the treatment of gastric cancer. Oncol Lett. 2019;18(6):5681–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Lee K, Hwang H, Nam KT. Immune response and the tumor microenvironment: how they communicate to regulate gastric cancer. Gut Liver. 2014;8(2):131–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rihawi K, Ricci AD, Rizzo A, Brocchi S, Marasco G, Pastore LV, et al. Tumor-Associated macrophages and Inflammatory Microenvironment in Gastric Cancer: Novel Translational implications. Int J Mol Sci. 2021;22(8).

  9. Baj J, Brzozowska K, Forma A, Maani A, Sitarz E, Portincasa P. Immunological aspects of the Tumor Microenvironment and epithelial-mesenchymal transition in gastric carcinogenesis. Int J Mol Sci. 2020;21(7).

  10. Łukaszewicz-Zając M, Szmitkowski M, Litman-Zawadzka A, Mroczko B. Matrix metalloproteinases and their tissue inhibitors in comparison to other inflammatory proteins in gastric Cancer (GC). Cancer Invest. 2016;34(7):305–12.

    Article  PubMed  Google Scholar 

  11. Panahizadeh R, Vatankhah MA, Safari A, Danesh H, Nazmi N, Gholizadeh P, et al. The interplay between microRNAs and Nrf2 signaling in human cancers. Cancer Cell Int. 2024;24(1):234.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gambardella V, Castillo J, Tarazona N, Gimeno-Valiente F, Martínez-Ciarpaglini C, Cabeza-Segura M, et al. The role of tumor-associated macrophages in gastric cancer development and their potential as a therapeutic target. Cancer Treat Rev. 2020;86:102015.

    Article  CAS  PubMed  Google Scholar 

  13. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel). 2014;6(3):1670–90.

    Article  PubMed  Google Scholar 

  14. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11(10):889–96.

    Article  CAS  PubMed  Google Scholar 

  15. Veremeyko T, Yung AWY, Anthony DC, Strekalova T, Ponomarev ED, Corrigendum. Early growth response Gene-2 is essential for M1 and M2 macrophage activation and plasticity by modulation of the transcription factor CEBPβ. Front Immunol. 2018;9:2923.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Che K, Luo Y, Song X, Yang Z, Wang H, Shi T, et al. Macrophages reprogramming improves immunotherapy of IL-33 in peritoneal metastasis of gastric cancer. EMBO Mol Med. 2024;16(2):251–66.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Ham IH, Lee D, Hur H. Role of Cancer-Associated Fibroblast in Gastric Cancer Progression and Resistance to treatments. J Oncol. 2019;2019:6270784.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Huang L, Wu RL, Xu AM. Epithelial-mesenchymal transition in gastric cancer. Am J Transl Res. 2015;7(11):2141–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Peng Z, Wang CX, Fang EH, Wang GB, Tong Q. Role of epithelial-mesenchymal transition in gastric cancer initiation and progression. World J Gastroenterol. 2014;20(18):5403–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cristescu R, Lee J, Nebozhyn M, Kim K-M, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21(5):449–56.

    Article  CAS  PubMed  Google Scholar 

  21. Park SJ, Nakagawa T, Kitamura H, Atsumi T, Kamon H, Sawa S, et al. IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J Immunol. 2004;173(6):3844–54.

    Article  CAS  PubMed  Google Scholar 

  22. Wang T-t, Zhao Y-l, Peng L-s, Chen N, Chen W, Lv Y-p, et al. Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF-PD-L1 pathway. Gut. 2017;66(11):1900–11.

    Article  CAS  PubMed  Google Scholar 

  23. Liu K, Yang K, Wu B, Chen H, Chen X, Chen X, et al. Tumor-infiltrating Immune cells are Associated with prognosis of gastric Cancer. Med (Baltim). 2015;94(39):e1631.

    Article  CAS  Google Scholar 

  24. Zhang W, Gu J, Chen J, Zhang P, Ji R, Qian H, et al. Interaction with neutrophils promotes gastric cancer cell migration and invasion by inducing epithelial-mesenchymal transition. Oncol Rep. 2017;38(5):2959–66.

    Article  CAS  PubMed  Google Scholar 

  25. Iida T, Iwahashi M, Katsuda M, Ishida K, Nakamori M, Nakamura M, et al. Tumor-infiltrating CD4 + Th17 cells produce IL-17 in tumor microenvironment and promote tumor progression in human gastric cancer. Oncol Rep. 2011;25(5):1271–7.

    CAS  PubMed  Google Scholar 

  26. Li S, Cong X, Gao H, Lan X, Li Z, Wang W, et al. Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. J Exp Clin Cancer Res. 2019;38(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Zhang J, Li Y, Chen J, Huang T, Lin J, Pi Y, et al. TOB1 modulates neutrophil phenotypes to influence gastric cancer progression and immunotherapy efficacy. Front Immunol. 2024;15:1369087.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang F, Lau JKC, Yu J. The role of natural killer cell in gastrointestinal cancer: killer or helper. Oncogene. 2021;40(4):717–30.

    Article  PubMed  Google Scholar 

  29. Lee JS, Won HS, Sun S, Hong JH, Ko YH. Prognostic role of tumor-infiltrating lymphocytes in gastric cancer: a systematic review and meta-analysis. Med (Baltim). 2018;97(32):e11769.

    Article  CAS  Google Scholar 

  30. Yang L, Lin PC. Mechanisms that drive inflammatory tumor microenvironment, tumor heterogeneity, and metastatic progression. Semin Cancer Biol. 2017;47:185–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Geng Y, Wang H, Lu C, Li Q, Xu B, Jiang J, et al. Expression of costimulatory molecules B7-H1, B7-H4 and Foxp3 + Tregs in gastric cancer and its clinical significance. Int J Clin Oncol. 2015;20(2):273–81.

    Article  CAS  PubMed  Google Scholar 

  32. Speiser DE, Ho PC, Verdeil G. Regulatory circuits of T cell function in cancer. Nat Rev Immunol. 2016;16(10):599–611.

    Article  CAS  PubMed  Google Scholar 

  33. Yang J, Liu X, Cheng Y, Zhang J, Ji F, Ling Z. Roles of plasmacytoid dendritic cells in gastric cancer. Front Oncol. 2022;12:818314.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fricke I, Gabrilovich DI. Dendritic cells and tumor microenvironment: a dangerous liaison. Immunol Invest. 2006;35(3–4):459–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan M, et al. Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res. 2004;64(16):5535–8.

    Article  CAS  PubMed  Google Scholar 

  36. Huang XM, Liu XS, Lin XK, Yu H, Sun JY, Liu XK, et al. Role of plasmacytoid dendritic cells and inducible costimulator-positive regulatory T cells in the immunosuppression microenvironment of gastric cancer. Cancer Sci. 2014;105(2):150–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Islam F, Mitra S, Emran TB, Khan Z, Nath N, Das R, et al. Natural small molecules in gastrointestinal tract and associated cancers: molecular insights and targeted therapies. Molecules. 2022;27(17):5686.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Becker J, Müller-Tidow C, Serve H, Domschke W, Pohle T. Role of receptor tyrosine kinases in gastric cancer: new targets for a selective therapy. World J Gastroenterology: WJG. 2006;12(21):3297.

    Article  CAS  PubMed Central  Google Scholar 

  39. Kommalapati A, Tella SH, Borad M, Javle M, Mahipal A. FGFR inhibitors in Oncology: insight on the management of toxicities in clinical practice. Cancers. 2021;13(12):2968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Qiao Y, Li T, Zheng S, Wang H. The Hippo pathway as a drug target in gastric cancer. Cancer Lett. 2018;420:14–25.

    Article  CAS  PubMed  Google Scholar 

  41. Tapia O, Riquelme I, Leal P, Sandoval A, Aedo S, Weber H, et al. The PI3K/AKT/mTOR pathway is activated in gastric cancer with potential prognostic and predictive significance. Virchows Arch. 2014;465(1):25–33.

    Article  CAS  PubMed  Google Scholar 

  42. Kumar D, Imtaiyaz Hassan M. Chapter 6 - regulation of receptor tyrosine kinase gene expression. In: Mansour HM, Khattab MM, El-khatib AS, editors. Receptor tyrosine Kinases in Neurodegenerative and Psychiatric. Disorders: Academic; 2023. pp. 245–76.

    Chapter  Google Scholar 

  43. Gizzio J, Thakur A, Haldane A, Post CB, Levy RM. Evolutionary sequence and structural basis for the distinct conformational landscapes of Tyr and Ser/Thr kinases. bioRxiv. 2024.

  44. Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, et al. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Signal Transduct Target Therapy. 2021;6(1):1–48.

    Article  Google Scholar 

  45. Qin S, Li A, Yi M, Yu S, Zhang M, Wu K. Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. J Hematol Oncol. 2019;12(1):1–11.

    Article  CAS  Google Scholar 

  46. Roviello G, Ravelli A, Polom K, Petrioli R, Marano L, Marrelli D, et al. Apatinib: a novel receptor tyrosine kinase inhibitor for the treatment of gastric cancer. Cancer Lett. 2016;372(2):187–91.

    Article  CAS  PubMed  Google Scholar 

  47. Yang X, Li W, Han X, Wang J, Dai J, Ye X, et al. Apatinib weakens proliferation, migration, invasion, and angiogenesis of thyroid cancer cells through downregulating pyruvate kinase M2. Sci Rep. 2024;14(1):879.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Tian S, Quan H, Xie C, Guo H, Lü F, Xu Y, et al. YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci. 2011;102(7):1374–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen P, Iruela-Arispe L, Lou L, Sun P, Yuan K. VEGFr inhibitor YN968D1 xenograft dose response studies against human colon cancer Ls174t and HT29. AACR; 2006.

  50. Lin J-X, Xu Y-C, Lin W, Xue F-Q, Ye J-X, Zang W-D, et al. Effectiveness and safety of Apatinib Plus Chemotherapy as Neoadjuvant Treatment for locally advanced gastric Cancer: a Nonrandomized Controlled Trial. JAMA Netw Open. 2021;4(7):e2116240–e.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Su B, Huang T, Jin Y, Yin H, Qiu H, Yuan X. Apatinib exhibits synergistic effect with pyrotinib and reverses acquired pyrotinib resistance in HER2-positive gastric cancer via stem cell factor/c-kit signaling and its downstream pathways. Gastric Cancer. 2021;24(2):352–67.

    Article  CAS  PubMed  Google Scholar 

  52. Peng W, Zhang F, Wang Z, Li D, He Y, Ning Z, et al. Large scale, multicenter, prospective study of apatinib in advanced gastric cancer: a real-world study from China. Cancer Manage Res. 2020;12:6977.

    Article  CAS  Google Scholar 

  53. Scott LJ. Apatinib: a review in advanced gastric cancer and other advanced cancers. Drugs. 2018;78(7):747–58.

    Article  CAS  PubMed  Google Scholar 

  54. Kataoka Y, Mukohara T, Tomioka H, Funakoshi Y, Kiyota N, Fujiwara Y, et al. Foretinib (GSK1363089), a multi-kinase inhibitor of MET and VEGFRs, inhibits growth of gastric cancer cell lines by blocking inter-receptor tyrosine kinase networks. Investig New Drugs. 2012;30(4):1352–60.

    Article  CAS  Google Scholar 

  55. Shah MA, Wainberg ZA, Catenacci DV, Hochster HS, Ford J, Kunz P, et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS ONE. 2013;8(3):e54014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Grojean M, Schwarz MA, Schwarz JR, Hassan S, von Holzen U, Zhang C, et al. Targeted dual inhibition of c-Met/VEGFR2 signalling by foretinib improves antitumour effects of nanoparticle paclitaxel in gastric cancer models. J Cell Mol Med. 2021;25(11):4950–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Awasthi N, Grojean M, Monahan S, Hassan S, von Holzen U, Schwarz MA, et al. The dual c-Met/VEGFR2 inhibitor foretinib augments chemotherapy response in preclinical models of gastric cancer. AACR; 2018.

  58. Sohn S-H, Kim B, Sul HJ, Choi BY, Kim HS, Zang DY. Foretinib inhibits cancer stemness and gastric cancer cell proliferation by decreasing CD44 and c-MET signaling. OncoTargets Therapy. 2020;13:1027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Okamoto W, Okamoto I, Arao T, Kuwata K, Hatashita E, Yamaguchi H, et al. Antitumor action of the MET tyrosine kinase inhibitor crizotinib (PF-02341066) in gastric cancer positive for MET amplification. Mol Cancer Ther. 2012;11(7):1557–64.

    Article  CAS  PubMed  Google Scholar 

  60. Kim JW, Kim H-P, Im S-A, Kang S, Hur HS, Yoon Y-K, et al. The growth inhibitory effect of lapatinib, a dual inhibitor of EGFR and HER2 tyrosine kinase, in gastric cancer cell lines. Cancer Lett. 2008;272(2):296–306.

    Article  CAS  PubMed  Google Scholar 

  61. LaBonte MJ, Manegold PC, Wilson PM, Fazzone W, Louie SG, Lenz HJ, et al. The dual EGFR/HER-2 tyrosine kinase inhibitor lapatinib sensitizes colon and gastric cancer cells to the irinotecan active metabolite SN‐38. Int J Cancer. 2009;125(12):2957–69.

    Article  CAS  PubMed  Google Scholar 

  62. Egile C, Kenigsberg M, Delaisi C, Bégassat F, Do-Vale V, Mestadier J, et al. The selective intravenous inhibitor of the MET tyrosine kinase SAR125844 inhibits tumor growth in MET-amplified cancer. Mol Cancer Ther. 2015;14(2):384–94.

    Article  CAS  PubMed  Google Scholar 

  63. Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol. 2001;2(3):1–12.

    Article  Google Scholar 

  64. Belov AA, Mohammadi M. Molecular mechanisms of fibroblast growth factor signaling in physiology and pathology. Cold Spring Harb Perspect Biol. 2013;5(6):a015958.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Zhang J, Tang PM, Zhou Y, Cheng AS, Yu J, Kang W, et al. Targeting the oncogenic FGF-FGFR axis in gastric carcinogenesis. Cells. 2019;8(6):637.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Grygielewicz P, Dymek B, Bujak A, Gunerka P, Stanczak A, Lamparska-Przybysz M, et al. Epithelial–mesenchymal transition confers resistance to selective FGFR inhibitors in SNU-16 gastric cancer cells. Gastric Cancer. 2016;19(1):53–62.

    Article  CAS  PubMed  Google Scholar 

  67. Xie L, Su X, Zhang L, Yin X, Tang L, Zhang X, et al. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin Cancer Res. 2013;19(9):2572–83.

    Article  CAS  PubMed  Google Scholar 

  68. Bang Y-J, Van Cutsem E, Mansoor W, Petty RD, Chao Y, Cunningham D, et al. A randomized, open-label phase II study of AZD4547 (AZD) versus paclitaxel (P) in previously treated patients with advanced gastric cancer (AGC) with fibroblast growth factor receptor 2 (FGFR2) polysomy or gene amplification (amp): SHINE study. American Society of Clinical Oncology; 2015.

  69. Schmidt K, Moser C, Hellerbrand C, Zieker D, Wagner C, Redekopf J, et al. Targeting fibroblast growth factor receptor (FGFR) with BGJ398 in a gastric cancer model. Anticancer Res. 2015;35(12):6655–65.

    CAS  PubMed  Google Scholar 

  70. Brameld KA, Owens TD, Verner E, Venetsanakos E, Bradshaw JM, Phan VT, et al. Discovery of the irreversible covalent FGFR inhibitor 8-(3-(4-Acryloylpiperazin-1-yl) propyl)-6-(2, 6-dichloro-3, 5-dimethoxyphenyl)-2-(methylamino) pyrido [2, 3-d] pyrimidin-7 (8 H)-one (PRN1371) for the treatment of solid tumors. ACS; 2017.

  71. Tsimafeyeu I, Daeyaert F, Joos J-B, Aken KV, Ludes-Meyers J, Byakhov M, et al. Molecular modeling, de novo design and synthesis of a novel, extracellular binding fibroblast growth factor receptor 2 inhibitor alofanib (RPT835). Med Chem. 2016;12(4):303–17.

    Article  CAS  PubMed  Google Scholar 

  72. Hall TG, Yu Y, Eathiraj S, Wang Y, Savage RE, Lapierre J-M, et al. Preclinical activity of ARQ 087, a novel inhibitor targeting FGFR dysregulation. PLoS ONE. 2016;11(9):e0162594.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Zhao G, Li W-y, Chen D, Henry JR, Li H-Y, Chen Z, et al. A novel, selective inhibitor of fibroblast growth factor receptors that shows a potent broad spectrum of antitumor activity in several tumor xenograft models. Mol Cancer Ther. 2011;10(11):2200–10.

    Article  CAS  PubMed  Google Scholar 

  74. Michael M, Bang Y-J, Park YS, Kang Y-K, Kim TM, Hamid O, et al. A phase 1 study of LY2874455, an oral selective pan-FGFR inhibitor, in patients with advanced cancer. Target Oncol. 2017;12(4):463–74.

    Article  PubMed  Google Scholar 

  75. Kim SY, Ahn T, Bang H, Ham JS, Kim J, Kim ST, et al. Acquired resistance to LY2874455 in FGFR2-amplified gastric cancer through an emergence of novel FGFR2-ACSL5 fusion. Oncotarget. 2017;8(9):15014.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Lau DK, Luk IY, Jenkins LJ, Martin A, Williams DS, Schoffer KL, et al. Rapid resistance of FGFR-driven gastric cancers to regorafenib and targeted FGFR inhibitors can be overcome by parallel inhibition of MEK. Mol Cancer Ther. 2021;20(4):704–15.

    Article  CAS  PubMed  Google Scholar 

  77. Arrouchi H, Lakhlili W, Ibrahimi A. A review on PIM kinases in tumors. Bioinformation. 2019;15(1):40.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Lee M, Lee K-H, Min A, Kim J, Kim S, Jang H, et al. Pan-pim kinase inhibitor AZD1208 suppresses tumor growth and synergistically interacts with akt inhibition in gastric cancer cells. Cancer Res Treatment: Official J Korean Cancer Association. 2019;51(2):451.

    Article  CAS  Google Scholar 

  79. Warnecke-Eberz U, Bollschweiler E, Drebber U, Metzger R, Baldus SE, Hoelscher AH, et al. Prognostic impact of protein overexpression of the proto-oncogene PIM-1 in gastric cancer. Anticancer Res. 2009;29(11):4451–5.

    PubMed  Google Scholar 

  80. Yan B, Yau EX, Samanta S, Ong CW, Yong KJ, Ng LK, et al. Clinical and therapeutic relevance of PIM1 kinase in gastric cancer. Gastric Cancer. 2012;15(2):188–97.

    Article  CAS  PubMed  Google Scholar 

  81. Kim S, Kim W, Kim D-H, Jang J-H, Kim S-J, Park S-A, et al. Resveratrol suppresses gastric cancer cell proliferation and survival through inhibition of PIM-1 kinase activity. Arch Biochem Biophys. 2020;689:108413.

    Article  CAS  PubMed  Google Scholar 

  82. Zhao C, Zeng C, Ye S, Dai X, He Q, Yang B, et al. Yes-associated protein (YAP) and transcriptional coactivator with a PDZ-binding motif (TAZ): a nexus between hypoxia and cancer. Acta Pharm Sinica B. 2020;10(6):947–60.

    Article  CAS  Google Scholar 

  83. Zhao B, Li L, Lei Q, Guan K-L. The Hippo–YAP pathway in organ size control and tumorigenesis: an updated version. Genes Dev. 2010;24(9):862–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Zhao B, Wei X, Li W, Udan RS, Yang Q, Kim J, et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 2007;21(21):2747–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Azzolin L, Panciera T, Soligo S, Enzo E, Bicciato S, Dupont S, et al. YAP/TAZ incorporation in the β-catenin destruction complex orchestrates the wnt response. Cell. 2014;158(1):157–70.

    Article  CAS  PubMed  Google Scholar 

  86. Harvey KF, Zhang X, Thomas DM. The Hippo pathway and human cancer. Nat Rev Cancer. 2013;13(4):246–57.

    Article  CAS  PubMed  Google Scholar 

  87. Yoshikawa K, Noguchi K, Nakano Y, Yamamura M, Takaoka K, Hashimoto-Tamaoki T, et al. The Hippo pathway transcriptional co-activator, YAP, confers resistance to cisplatin in human oral squamous cell carcinoma. Int J Oncol. 2015;46(6):2364–70.

    Article  CAS  PubMed  Google Scholar 

  88. Yu L, Gao C, Feng B, Wang L, Tian X, Wang H, et al. Distinct prognostic values of YAP1 in gastric cancer. Tumor Biology. 2017;39(4):1010428317695926.

    Article  PubMed  Google Scholar 

  89. Zhou Z, Zhu JS, Gao CP, Li LP, Zhou C, Wang H, et al. siRNA targeting YAP gene inhibits gastric carcinoma growth and tumor metastasis in SCID mice. Oncol Lett. 2016;11(4):2806–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Giraud J, Molina-Castro S, Seeneevassen L, Sifré E, Izotte J, Tiffon C, et al. Verteporfin targeting YAP1/TAZ‐TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Int J Cancer. 2020;146(8):2255–67.

    Article  CAS  PubMed  Google Scholar 

  91. Jiao S, Wang H, Shi Z, Dong A, Zhang W, Song X, et al. A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell. 2014;25(2):166–80.

    Article  CAS  PubMed  Google Scholar 

  92. Wang Q, Lu P, Wang T, Zheng Q, Li Y, Leng SX, et al. Sitagliptin affects gastric cancer cells proliferation by suppressing Melanoma-associated antigen‐A3 expression through yes‐associated protein inactivation. Cancer Med. 2020;9(11):3816–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Tang Y, Fang G, Guo F, Zhang H, Chen X, An L, et al. Selective inhibition of STRN3-containing PP2A phosphatase restores hippo tumor-suppressor activity in gastric cancer. Cancer Cell. 2020;38(1):115–28. e9.

    Article  CAS  PubMed  Google Scholar 

  94. Song J, Gao Q-L, Wu B-W, Zhu T, Cui X-X, Jin C-J, et al. Discovery of tertiary amide derivatives incorporating benzothiazole moiety as anti-gastric cancer agents in vitro via inhibiting tubulin polymerization and activating the Hippo signaling pathway. Eur J Med Chem. 2020;203:112618.

    Article  CAS  PubMed  Google Scholar 

  95. Seeneevassen L, Giraud J, Molina-Castro S, Sifré E, Tiffon C, Beauvoit C, et al. Leukaemia inhibitory factor (LIF) inhibits cancer stem cells tumorigenic properties through hippo kinases activation in gastric cancer. Cancers. 2020;12(8):2011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Kim S-H, Jin H, Meng RY, Kim D-Y, Liu YC, Chai OH, et al. Activating hippo pathway via Rassf1 by ursolic acid suppresses the tumorigenesis of gastric cancer. Int J Mol Sci. 2019;20(19):4709.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Zhang Y, Xin C, Qiu J, Wang Z. Essential oil from pinus koraiensis pinecones inhibits gastric cancer cells via the HIPPO/YAP signaling pathway. Molecules. 2019;24(21):3851.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ye C, Wang W, Xia G, Yu C, Yi Y, Hua C, et al. A novel curcumin derivative cl-6 exerts antitumor effect in human gastric cancer cells by inducing apoptosis through hippo–YaP signaling pathway. OncoTargets Therapy. 2019;12:2259.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Fattahi S, Amjadi-Moheb F, Tabaripour R, Ashrafi GH, Akhavan-Niaki H. PI3K/AKT/mTOR signaling in gastric cancer: epigenetics and beyond. Life Sci. 2020;262:118513.

    Article  CAS  PubMed  Google Scholar 

  100. Khorasani ABS, Pourbagheri-Sigaroodi A, Pirsalehi A, Safaroghli-Azar A, Zali MR, Bashash D. The PI3K/Akt/mTOR signaling pathway in gastric cancer; from oncogenic variations to the possibilities for pharmacologic interventions. Eur J Pharmacol. 2021;898:173983.

    Article  Google Scholar 

  101. Beyens M, Vandamme T, Peeters M, Van Camp G, de Beeck KO. Resistance to targeted treatment of gastroenteropancreatic neuroendocrine tumors. Endocrine-related Cancer. 2019;26(3):R109–30.

    Article  CAS  PubMed  Google Scholar 

  102. Feng W, Brown RE, Trung CD, Li W, Wang L, Khoury T, et al. Morphoproteomic profile of mTOR, Ras/Raf kinase/ERK, and NF-κB pathways in human gastric adenocarcinoma. Annals Clin Lab Sci. 2008;38(3):195–209.

    CAS  Google Scholar 

  103. Lang SA, Gaumann A, Koehl GE, Seidel U, Bataille F, Klein D, et al. Mammalian target of rapamycin is activated in human gastric cancer and serves as a target for therapy in an experimental model. Int J Cancer. 2007;120(8):1803–10.

    Article  CAS  PubMed  Google Scholar 

  104. Ying J, Xu Q, Liu B, Zhang G, Chen L, Pan H. The expression of the PI3K/AKT/mTOR pathway in gastric cancer and its role in gastric cancer prognosis. OncoTargets Therapy. 2015;8:2427.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Yu G, Wang J, Chen Y, Wang X, Pan J, Li G, et al. Overexpression of phosphorylated mammalian target of rapamycin predicts lymph node metastasis and prognosis of Chinese patients with gastric cancer. Clin Cancer Res. 2009;15(5):1821–9.

    Article  CAS  PubMed  Google Scholar 

  106. Matsuzaki T, Yashiro M, Kaizaki R, Yasuda K, Doi Y, Sawada T, et al. Synergistic antiproliferative effect of mTOR inhibitors in combination with 5-fluorouracil in scirrhous gastric cancer. Cancer Sci. 2009;100(12):2402–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Chen H, Guan R, Lei Y, Chen J, Ge Q, Zhang X, et al. Lymphangiogenesis in gastric cancer regulated through Akt/mTOR-VEGF-C/VEGF-D axis. BMC Cancer. 2015;15(1):1–7.

    Article  Google Scholar 

  108. Cao Y, Luo Y, Zou J, Ouyang J, Cai Z, Zeng X, et al. Autophagy and its role in gastric cancer. Clin Chim Acta. 2019;489:10–20.

    Article  CAS  PubMed  Google Scholar 

  109. Shen Y-C, Li C-P, Yen C-J, Hsu C, Lin Y-L, Lin Z-Z, et al. Phase II multicentered study of low-dose everolimus plus cisplatin and weekly 24-hour infusion of high-dose 5-fluorouracil and leucovorin as first-line treatment for patients with advanced gastric cancer. Oncology. 2014;87(2):104–13.

    Article  CAS  PubMed  Google Scholar 

  110. Fukamachi H, Kim S-K, Koh J, Lee HS, Sasaki Y, Yamashita K, et al. A subset of diffuse-type gastric cancer is susceptible to mTOR inhibitors and checkpoint inhibitors. J Experimental Clin Cancer Res. 2019;38(1):1–23.

    Article  Google Scholar 

  111. Park JH, Ryu M-H, Park YS, Park SR, Na Y-S, Rhoo B-Y, et al. Successful control of heavily pretreated metastatic gastric cancer with the mTOR inhibitor everolimus (RAD001) in a patient with PIK3CA mutation and pS6 overexpression. BMC Cancer. 2015;15(1):1–6.

    Article  Google Scholar 

  112. Chen D, Lin X, Zhang C, Liu Z, Chen Z, Li Z, et al. Dual PI3K/mTOR inhibitor BEZ235 as a promising therapeutic strategy against paclitaxel-resistant gastric cancer via targeting PI3K/Akt/mTOR pathway. Cell Death Dis. 2018;9(2):1–11.

    Article  Google Scholar 

  113. Liu Y, Chen S, Xue R, Zhao J, Di M. Mefloquine effectively targets gastric cancer cells through phosphatase-dependent inhibition of PI3K/Akt/mTOR signaling pathway. Biochem Biophys Res Commun. 2016;470(2):350–5.

    Article  CAS  PubMed  Google Scholar 

  114. Tang C, Yang L, Jiang X, Xu C, Wang M, Wang Q, et al. Antibiotic drug tigecycline inhibited cell proliferation and induced autophagy in gastric cancer cells. Biochem Biophys Res Commun. 2014;446(1):105–12.

    Article  CAS  PubMed  Google Scholar 

  115. Rong L, Li Z, Leng X, Li H, Ma Y, Chen Y, et al. Salidroside induces apoptosis and protective autophagy in human gastric cancer AGS cells through the PI3K/Akt/mTOR pathway. Biomed Pharmacother. 2020;122:109726.

    Article  CAS  PubMed  Google Scholar 

  116. Liao K-F, Chiu T-L, Huang S-Y, Hsieh T-F, Chang S-F, Ruan J-W, et al. Anti-cancer effects of Radix Angelica Sinensis (Danggui) and N-Butylidenephthalide on gastric Cancer: implications for REDD1 activation and mTOR inhibition. Cell Physiol Biochem. 2018;48(6):2231–46.

    Article  CAS  PubMed  Google Scholar 

  117. Zhang P, Zhang C, Li X, Chang C, Gan C, Ye T, et al. Immunotherapy for gastric cancer: advances and challenges. MedComm–Oncology. 2024;3(4):e92.

    Article  Google Scholar 

  118. Bolandi N, Derakhshani A, Hemmat N, Baghbanzadeh A, Asadzadeh Z, Afrashteh Nour M, et al. The positive and negative Immunoregulatory Role of B7 Family: promising novel targets in gastric Cancer treatment. Int J Mol Sci. 2021;22(19):10719.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Alemohammad H, Najafzadeh B, Asadzadeh Z, Baghbanzadeh A, Ghorbaninezhad F, Najafzadeh A, et al. The importance of immune checkpoints in immune monitoring: a future paradigm shift in the treatment of cancer. Biomed Pharmacother. 2022;146:112516.

    Article  CAS  PubMed  Google Scholar 

  120. Sun S, Ma J, Zuo T, Shi J, Sun L, Meng C, et al. Inhibition of PCSK9: a promising enhancer for anti-PD-1/PD-L1 immunotherapy. Research. 2024;7:0488.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Sun S, Yang Z, Yao H, Zhang Z. A new enhancer for anti-PD-1/PD-L1 immunotherapy: PCSK9 inhibition. Trends Cancer. 2024;24:S2405-8033

  122. Makaremi S, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Sgambato A, Ghorbaninezhad F, et al. Immune checkpoint inhibitors in colorectal cancer: challenges and future prospects. Biomedicines. 2021;9(9):1075.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Sznol M, Melero I. Revisiting anti-CTLA-4 antibodies in combination with PD-1 blockade for cancer immunotherapy. Ann Oncol. 2021;32(3):295–7.

    Article  CAS  PubMed  Google Scholar 

  124. Zhao Y, Yang W, Huang Y, Cui R, Li X, Li B. Evolving roles for targeting CTLA-4 in cancer immunotherapy. Cell Physiol Biochem. 2018;47(2):721–34.

    Article  CAS  PubMed  Google Scholar 

  125. Camacho LH. Novel therapies targeting the immune system: CTLA4 blockade with tremelimumab (CP-675,206), a fully human monoclonal antibody. Expert Opin Investig Drugs. 2008;17(3):371–85.

    Article  CAS  PubMed  Google Scholar 

  126. Kelly RJ, Lee J, Bang Y-J, Almhanna K, Blum-Murphy M, Catenacci DV, et al. Safety and efficacy of durvalumab and tremelimumab alone or in combination in patients with advanced gastric and gastroesophageal junction adenocarcinoma. Clin Cancer Res. 2020;26(4):846–54.

    Article  CAS  PubMed  Google Scholar 

  127. Ralph C, Elkord E, Burt DJ, O’Dwyer JF, Austin EB, Stern PL, et al. Modulation of lymphocyte regulation for Cancer Therapy: a phase II trial of Tremelimumab in Advanced gastric and esophageal AdenocarcinomaAnti-CTLA4 in gastric and esophageal adenocarcinoma. Clin Cancer Res. 2010;16(5):1662–72.

    Article  CAS  PubMed  Google Scholar 

  128. Zhang L, Qiu M, Jin Y, Ji J, Li B, Wang X, et al. Programmed cell death ligand 1 (PD-L1) expression on gastric cancer and its relationship with clinicopathologic factors. Int J Clin Exp Pathol. 2015;8(9):11084.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Muro K, Chung HC, Shankaran V, Geva R, Catenacci D, Gupta S, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol. 2016;17(6):717–26.

    Article  CAS  PubMed  Google Scholar 

  130. Kiasari BA, Abbasi A, Darestani NG, Adabi N, Moradian A, Yazdani Y, et al. Combination therapy with nivolumab (anti-PD-1 monoclonal antibody): a new era in tumor immunotherapy. Int Immunopharmacol. 2022;113:109365.

    Article  Google Scholar 

  131. Kang Y-K, Boku N, Satoh T, Ryu M-H, Chao Y, Kato K, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71.

    Article  CAS  PubMed  Google Scholar 

  132. Wang B, Qin L, Ren M, Sun H. Effects of combination of anti-CTLA-4 and anti-PD-1 on gastric cancer cells proliferation, apoptosis and metastasis. Cell Physiol Biochem. 2018;49(1):260–70.

    Article  CAS  PubMed  Google Scholar 

  133. Janjigian YY, Ott PA, Calvo E, Kim JW, Ascierto PA, Sharma P, et al. Nivolumab ± Ipilimumab in pts with advanced (adv)/metastatic chemotherapy-refractory (CTx-R) gastric (G), esophageal (E), or gastroesophageal junction (GEJ) cancer: CheckMate 032 study. American Society of Clinical Oncology; 2017.

  134. Chen C, Zhang F, Zhou N, Gu Y-M, Zhang Y-T, He Y-D, et al. Efficacy and safety of immune checkpoint inhibitors in advanced gastric or gastroesophageal junction cancer: a systematic review and meta-analysis. Oncoimmunology. 2019;8(5):e1581547.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Rha SY, Lee C-K, Kim HS, Kang B, Jung M, Bae WK, 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). American Society of Clinical Oncology; 2020.

  136. Janjigian YY, Maron SB, Chatila WK, Millang B, Chavan SS, Alterman C, et al. First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial. Lancet Oncol. 2020;21(6):821–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Janjigian YY, Kawazoe A, Yañez P, Li N, Lonardi S, Kolesnik O, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature. 2021;600(7890):727–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Catenacci DV, Kang Y-K, Park H, Uronis HE, Lee K-W, Ng MC, 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(8):1066–76.

    Article  CAS  PubMed  Google Scholar 

  139. Khan U, Shah MA. Ramucirumab for the treatment of gastric or gastro-esophageal junction cancer. Expert Opin Biol Ther. 2019;19(11):1135–41.

    Article  CAS  PubMed  Google Scholar 

  140. Bang Y-J, Golan T, Dahan L, Fu S, Moreno V, Park K, et al. Ramucirumab and durvalumab for previously treated, advanced non–small-cell lung cancer, gastric/gastro-oesophageal junction adenocarcinoma, or hepatocellular carcinoma: an open-label, phase Ia/b study (JVDJ). Eur J Cancer. 2020;137:272–84.

    Article  CAS  PubMed  Google Scholar 

  141. Newick K, O’Brien S, Moon E, Albelda SM. CAR T cell therapy for solid tumors. Annu Rev Med. 2017;68:139–52.

    Article  CAS  PubMed  Google Scholar 

  142. Han Y, Liu C, Li G, Li J, Lv X, Shi H, et al. Antitumor effects and persistence of a novel HER2 CAR T cells directed to gastric cancer in preclinical models. Am J Cancer Res. 2018;8(1):106–19.

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Bębnowska D, Grywalska E, Niedźwiedzka-Rystwej P, Sosnowska-Pasiarska B, Smok-Kalwat J, Pasiarski M et al. CAR-T cell Therapy-An overview of targets in gastric Cancer. J Clin Med. 2020;9(6).

  144. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4):69.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Long B, Qin L, Zhang B, Li Q, Wang L, Jiang X, et al. CAR T–cell therapy for gastric cancer: potential and perspective (review). Int J Oncol. 2020;56(4):889–99.

    CAS  PubMed  Google Scholar 

  146. Ma S, Li X, Wang X, Cheng L, Li Z, Zhang C, et al. Current progress in CAR-T cell therapy for solid tumors. Int J Biol Sci. 2019;15(12):2548–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Morello A, Sadelain M, Adusumilli PS. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 2016;6(2):133–46.

    Article  CAS  PubMed  Google Scholar 

  148. Fousek K, Ahmed N. The evolution of T-cell therapies for solid malignancies. Clin Cancer Res. 2015;21(15):3384–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Song Y, Tong C, Wang Y, Gao Y, Dai H, Guo Y, et al. Effective and persistent antitumor activity of HER2-directed CAR-T cells against gastric cancer cells in vitro and xenotransplanted tumors in vivo. Protein Cell. 2018;9(10):867–78.

    Article  CAS  PubMed  Google Scholar 

  150. Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022;28(6):1189–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Botta GP, Chao J, Ma H, Hahn M, Sierra G, Jia J, et al. Metastatic gastric cancer target lesion complete response with Claudin18. 2-CAR T cells. J Immunother Cancer. 2024;12(2).

  152. Kim M, Pyo S, Kang CH, Lee CO, Lee HK, Choi SU, et al. Folate receptor 1 (FOLR1) targeted chimeric antigen receptor (CAR) T cells for the treatment of gastric cancer. PLoS ONE. 2018;13(6):e0198347.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Lv J, Zhao R, Wu D, Zheng D, Wu Z, Shi J, et al. Mesothelin is a target of chimeric antigen receptor T cells for treating gastric cancer. J Hematol Oncol. 2019;12(1):18.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Frey NV, Porter DL. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Hematol Am Soc Hematol Educ Program. 2016;2016(1):567–72.

    Article  Google Scholar 

  155. Adkins S. CAR T-Cell therapy: adverse events and management. J Adv Pract Oncol. 2019;10(Suppl 3):21–8.

    PubMed  PubMed Central  Google Scholar 

  156. Beck A, Goetsch L, Dumontet C, Corvaïa N. Strategies and challenges for the next generation of antibody–drug conjugates. Nat Rev Drug Discovery. 2017;16(5):315–37.

    Article  CAS  PubMed  Google Scholar 

  157. Koganemaru S, Shitara K. Antibody–drug conjugates to treat gastric cancer. Expert Opin Biol Ther. 2021;21(7):923–30.

    Article  CAS  PubMed  Google Scholar 

  158. Singh D, Dheer D, Samykutty A, Shankar R. Antibody drug conjugates in gastrointestinal cancer: from lab to clinical development. J Controlled Release. 2021;340:1–34.

    Article  CAS  Google Scholar 

  159. Thuss-Patience PC, Shah MA, Ohtsu A, Van Cutsem E, Ajani JA, Castro H, et al. Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): an international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol. 2017;18(5):640–53.

    Article  CAS  PubMed  Google Scholar 

  160. Shitara K, Bang Y-J, Iwasa S, Sugimoto N, Ryu M-H, Sakai D, et al. Trastuzumab Deruxtecan in previously treated HER2-positive gastric cancer. N Engl J Med. 2020;382(25):2419–30.

    Article  CAS  PubMed  Google Scholar 

  161. Hsu H-J, Tung C-P, Yu C-M, Chen C-Y, Chen H-S, Huang Y-C, et al. Eradicating mesothelin-positive human gastric and pancreatic tumors in xenograft models with optimized anti-mesothelin antibody–drug conjugates from synthetic antibody libraries. Sci Rep. 2021;11(1):15430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Zhu X-y, Li Q-x, Kong Y, Huang K-k, Wang G, Wang Y-j, et al. A novel human single-domain antibody-drug conjugate targeting CEACAM5 exhibits potent in vitro and in vivo antitumor activity. Acta Pharmacol Sin. 2024;45(3):609–18.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Elham Safarzadeh put forward the content of the paper. Reza Panahizadeh, Padideh Panahi, Shima Makaremi, and Ghasem Noorkhajavi wrote the manuscript. Vahid Asghariazar and Reza Panahizadeh prepared figures. Elham Safarzadeh revised and edited the final version of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Elham Safarzadeh.

Ethics declarations

Ethical approval

The study was approved by the Research Ethics Committee of Ardabil University of Medical Sciences (IR.ARUMS.REC.1403.094).

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Panahizadeh, R., Panahi, P., Asghariazar, V. et al. A literature review of recent advances in gastric cancer treatment: exploring the cross-talk between targeted therapies. Cancer Cell Int 25, 23 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03655-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03655-8

Keywords