Review Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jun 15, 2024; 16(6): 2318-2334
Published online Jun 15, 2024. doi: 10.4251/wjgo.v16.i6.2318
Advances in targeted therapy for human epidermal growth factor receptor 2 positive in advanced gastric cancer
Ya-Kun Jiang, Meng Yue, Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
Wei Li, Health Management Center, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
Ying-Yang Qiu, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
ORCID number: Meng Yue (0009-0003-7827-5928).
Author contributions: Jiang YK wrote the manuscript; Li W and Qiu YY collected the data; Yue M guided the manuscript; all authors reviewed, edited, and approved the final manuscript and revised it critically for important intellectual content, gave final approval of the version to be published, and agreed to be accountable for all aspects of the work.
Conflict-of-interest statement: All the authors declare no conflict of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Meng Yue, MM, Doctor, Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, No. 105 Jiefang Road, Jinan 250013, Shandong Province, China. mengtiger78@163.com
Received: December 11, 2023
Revised: April 4, 2024
Accepted: April 18, 2024
Published online: June 15, 2024

Abstract

Emerging therapeutic methods represented by targeted therapy are effective supplements to traditional first-line chemoradiotherapy resistance. Human epidermal growth factor receptor 2 (HER2) is one of the most important targets in targeted therapy for gastric cancer. Trastuzumab combined with chemotherapy has been used as the first-line treatment for advanced gastric cancer. The safety and efficacy of pertuzumab and margetuximab in the treatment of gastric cancer have been verified. However, monoclonal antibodies, due to their large molecular weight, inability to penetrate the blood-brain barrier, and drug resistance, lead to decreased therapeutic efficacy, so it is necessary to explore the efficacy of other HER2-targeting therapies in gastric cancer. Small-molecule tyrosine kinase inhibitors, such as lapatinib and pyrrotinib, have the advantages of small molecular weight, penetrating the blood-brain barrier and high oral bioavailability, and are expected to become the drugs of choice for perioperative treatment and neoadjuvant therapy of gastric cancer after validation by large-scale clinical trials in the future. Antibo-drug conjugate, such as T-DM1 and T-DXd, can overcome the resistance of monoclonal antibodies despite their different mechanisms of tumor killing, and are a supplement for the treatment of patients who have failed the treatment of monoclonal antibodies such as trastuzumab. Therefore, after more detailed stratification of gastric cancer patients, various gastric cancer drugs targeting HER2 are expected to play a more significant role.

Key Words: Human epidermal growth factor receptor 2, Gastric cancer, Targeted therapy, Review

Core Tip: Human epidermal growth factor receptor 2 (HER2) is one of the most important targets in targeted therapy for gastric cancer. Trastuzumab combined with chemotherapy has been used as the first-line treatment for advanced gastric cancer. The safety and efficacy of pertuzumab and magtuximab in the treatment of gastric cancer have been verified. However, monoclonal antibodies, due to their large molecular weight, inability to penetrate the blood-brain barrier, and drug resistance, lead to decreased therapeutic efficacy, so it is necessary to explore the efficacy of other HER2-targeting therapies in gastric cancer.



INTRODUCTION

Stomach cancer is currently the fifth most common cancer worldwide, with the fourth highest case fatality rate[1-4]. Surgical resection and multidisciplinary treatment combined with chemoradiotherapy, targeted therapy and immunotherapy are the main methods in the treatment of gastric cancer[5]. Because there are no obvious specific symptoms in the early stage, most patients are not diagnosed until the tumor has developed to an advanced stage, resulting in some patients losing the opportunity for surgical treatment[6-8]. The 5-year survival rate of early gastric cancer is 70%, that of advanced gastric cancer is 32%, and that of distant metastatic gastric cancer is only 6%[9]. Therefore, there is an urgent need to find more new and effective therapeutic targets[10-12]. With the development of precision medicine, the score of cancer. Sub-targeted therapy has become the main treatment method for gastric cancer. Human epidermal growth factor receptor 2 (HER2) is an important therapeutic target of the epidermal growth factor receptor (EGFR) family, with an average positive rate of 17.9% in gastric cancer. In 2010, the ToGA trial was the first to demonstrate that trastuzumab combined with chemotherapy extended survival[13]. At present, in addition to trastuzumab, there are many other HER2-targeting therapies still under study, such as monoclonal antibodies (trastuzumab, pertuzumab and magtuximab, etc.), antibo-drug conjugate (ADC), such as T-DM1, T-DXd, etc., tyrosine kinase inhibitors (TKI) such as lapatinib, pyrrotinib, etc.[14-17].

Gastric cancer is a heterogeneous and highly aggressive malignancy with the 5th highest incidence and 3rd highest mortality worldwide[18]. Most patients are in the advanced stage when diagnosed, and the prognosis is very poor. Systemic therapy is currently the most important treatment for advanced gastric cancer, and HER2 is an important therapeutic target for HER2-positive gastric cancer[19-22]. With the continuous optimization of HER2-targeting drugs and therapies, some gastric cancer patients have benefited from it. However, the high incidence of drug resistance and serious adverse reactions are still the bottlenecks limiting the application of HER2-targeted drugs[23]. Therefore, the development of novel anti-tumor drugs is of great significance for improving the long-term survival of patients with HER2-positive gastric cancer. ADCs are a new class of highly effective anti-tumor drugs, which are composed of specific targeted monoclonal antibodies, chemical junctions and small cytotoxic payloads[24]. Their main advantages are powerful therapeutic effect and moderate tissue toxicity[25]. In recent years, ADC has made great progress in the field of targeted therapy for HER2-positive advanced gastric cancer[26-30]. First of all, after years of development, a variety of ADCs, including DS-8201, RC48, etc., have been used for second-line and postline treatment of gastric cancer. Secondly, with the progress of ADC bioengineering technology, including high ratio of drug antibodies, cleavable linkers, toxic payloads that can cause bystander effect, etc., new ADCs can play a more significant therapeutic effect in the treatment of specific tumor targets. Some of these ADCs also have multi-targeting and can play an anti-cancer effect against multiple specific targets[31-33]. At the same time, the development of ADCs has reached the third stage, and the new generation of ADCs through site-specific coupling technology has higher homogeneity, more efficient cytotoxic payload, higher precision, and lower non-targeted toxic effects[34-36]. In addition, "target-free combination" therapy consisting of ADC and immune checkpoint inhibitors (ICI) may be a promising treatment strategy for advanced gastric cancer.

This reviews the progress and advantages of HER2-targeted therapies in the treatment of gastric cancer in recent years, and discusses the existing problems and future development directions.

TARGETED THERAPY FOR PATIENTS WITH HER2 GENE POSITIVE GASTRIC CANCER

Many studies[37-40] have shown that the HER2 gene is overexpressed in different tumors such as breast, ovarian, prostate, colorectal, and lung cancers. The HER2 gene (also known as Neu or ErbB2) is located on human chromosome 17 (17q21) and encodes the transmembrane glycoprotein p185. Studies[41-45] have shown that the positive rate of HER2 in gastric cancer is 4.4% to 53.4%, with an average of 17.9%[46-50]. The HER2 protein has four extracellular domains with unique structural and functional features that interact with ligands and initiate signal transduction, domains Ⅰ and Ⅲ have inherent ligand binding capabilities, domain Ⅱ can form homologous or heterodimers with other HER molecules, and domain Ⅳ at the proximal end of the membrane[51-54]. Different ligands are involved in the proliferation and apoptosis of tumor cells by activating Ras/MAPK and PI3K-Akt by binding to different domains of HER2 receptor[55]. In addition, HER2 overexpression is closely related to the formation of neovascularization in gastric cancer[56-60]. At present, the recommended methods for detecting HER2 expression are mainly immunohistochemistry, which can be combined with fluorescence in situ hybridization if necessary. Second-generation sequencing, although expensive, is also gaining interest as it helps to find mutations in HER genes. Liquid biopsy techniques can also obtain tumor lesion information by detecting circulating tumor cells or circulating tumor DNA. In gastric cancer, Helicobacter pylori infection was positively correlated with the positive rate of HER2 in gastric cancer[61-65]. The difference in the positive rate of HER2 in gastric cancer was statistically significant between the killed and non-killed groups, suggesting that the detection of Helicobacter pylori and HER2 is of great significance for the detection of early gastric cancer and the evaluation of tumor malignancy[66]. At present, the correlation between HER2 overexpression and clinical outcome of gastric cancer is uncertain. Some studies[67-70] have shown that HER2 positivity is associated with a significantly worse prognosis. Other studies[71-75] have found no correlation between HER2 status and prognosis, so the prognostic value of HER2 in gastric cancer remains uncertain and needs further study (Figure 1).

Figure 1
Figure 1 Human epidermal growth factor receptor 2 gene mutation site diagram. Created with BioRender.com (see Supplementary material).

HER2 is currently the only clear therapeutic target for gastric cancer, and the ToGA trial is the first large-scale phase III clinical trial in the world to prove that targeted therapy for gastric cancer can prolong survival, making trastuzumab combined with first-line chemotherapy agents the standard first-line treatment for HER2-positive advanced gastroesophageal adenocarcinoma[76-80]. However, targeting HER2 has achieved significant efficacy in the treatment of breast cancer, such as trastuzumab combined with paclitaxel or pertuzumab, the combination therapy of lapatinib and the combination therapy of the antibody coupling drug T-DM1, etc., but these therapies have not achieved positive results in the targeted therapy of HER2-positive gastric cancer (Figure 2). At present, the combination of trastuzumab, pertuzumab and magtuximab with immunotherapy drugs is the main direction of clinical trials of targeted therapy for gastric cancer[81]. Due to the emergence of drug resistance, the efficacy of trastuzumab is not good in some patients, so TKI and ADC are the choice of targeted therapy for this part of patients[82-85].

Figure 2
Figure 2 Activation pathway of human epidermal growth factor receptor 2 gene in gastric cancer immunotherapy. Created with BioRender.com (see Supplementary material).
MONOCLONAL ANTIBODY TARGETING HER2

Monoclonal antibodies targeting HER2 recognize the HER2 antigen through the Fab segment, and the Fc segment binds to immune cells to exert anti-tumor function through antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis, and complement-dependent cytotoxicity[86-88]. Compared with other targeted drugs, HER2-targeting monoclonal antibodies have stronger anti-tumor specificity, but because of their relatively large molecular weight, they cannot cross the blood-brain barrier, so the treatment of patients with brain metastases has limitations (Figure 3).

Figure 3
Figure 3 Mechanism of action of anti-human epidermal growth factor receptor 2 therapies in gastric cancer. Created with BioRender.com (see Supplementary material).
TRASTUZUMAB

Trastuzumab is the only monoclonal antibody approved by the US Food and Drug Administration (FDA) for the first-line treatment of HER2-positive advanced gastric cancer[89-93]. It can bind to the extracellular domain Ⅳ of HER2, block the HER2 signaling pathway and inhibit the proliferation of tumor cells[94]. The ToGA trial showed that overall survival (OS) in the trastuzumab plus chemotherapy group was superior to chemotherapy alone [median OS (mOS) 13.8 months vs 11.1 months] compared with the traditional standard chemotherapy regimens for gastric cancer (capecitabine/cisplatin or fluorouracil/cisplatin)[95]. The ToGA trial also yielded consistent results in subgroup analyses in Japan[96-98]. In addition to survival, patients' recovery adjustment time for health-related quality of life and absence of disease or toxic symptoms was also longer than that for chemotherapy alone[99]. In addition, in the HERXO trial, trastuzumab combined with capecitabine and oxaliplatin were significantly effective in the treatment of advanced gastric cancer patients (mOS was 13.8 months vs 7.1 months), the 18-month disease-free survival (DFS) rate was 71% in perioperative treatment of gastric cancer[100-104]. The main results of trastuzumab combined with capecitabine and oxaliplatin in the treatment of advanced gastric cancer were consistent with those of the CGOG1001 trial[105-108]. The above results further established the basic position of trastuzumab in the first-line treatment of gastric cancer.

In the second-line chemotherapy regimen for gastric cancer, 5-FU, calcium folinate, oxaliplatin, irinotecan, docetaxel and paclitaxel are the main chemotherapy drugs at present[109-112]. A clinical trial of trastuzumab combined with 5-FU, leucovorin, oxaliplatin, and docetaxel for perioperative treatment of gastric cancer (median DFS was 42.5 months) demonstrated the efficacy of trastuzumab in second-line chemotherapy for gastric cancer[113-115]. However, the combination of trastuzumab and paclitaxel in the treatment of advanced gastric cancer did not effectively prolong the survival of patients in the WJOG7112G trial and T-CORE1203 trial, which is consistent with the current mainstream view that trastuzumab is not a recommended choice in second-line chemotherapy for gastric cancer[116-118]. Therefore, the efficacy of trastuzumab in second-line chemotherapy for gastric cancer still needs to be further explored[119]. The drug resistance of trastuzumab is often the main reason limiting its efficacy[120]. The use of other drugs in combination with trastuzumab is still the main research direction of clinical trials.

PERTUZUMAB

Pertuzumab is a recombinant humanized monoclonal antibody that binds to the extracellular dimerization domain Ⅱ of HER2, prevents the formation of ligand-induced HER2 heterodimer, and can play a synergistic role with trastuzumab to inhibit tumor growth[121-126]. The JACOB trial applied pertuzumab and trastuzumab combined chemotherapy in the treatment of gastric cancer. The mOS of the pertuzumab group was 17.5 months, while that of the control group was 14.2 months.

mOS was extended by 3.3 months, but there was no statistical difference between the two, nor was it confirmed that the addition of pertuzumab effectively improved survival time[127-130]. Another clinical trial evaluating the dose and safety of pertuzumab in combination with trastuzumab, capecitabine, and cisplatin in patients with HER2-positive advanced gastric cancer showed that patients receiving a valley concentration (62.7 μg/mL) of pertuzumab (840 mg) every 3 wk had a higher response rate than those receiving the first week of pertuzumab loading dose (840 mg). Partial response rates (reduction in tumor volume) were 86% and 55%, respectively, in patients receiving weekly valley concentrations of 40.0 μg/mL of pertuzumab (420 mg)[131-134]. The INNOVATION trial applied pertuzumab combined with trastuzumab and chemotherapy drugs in perioperative chemotherapy for gastric cancer, which will further clarify the significance of pertuzumab in the treatment of gastric cancer[135].

MAGTUXIMAB

Magtuximab is a monoclonal antibody with an engineered Fc domain that targets the HER2 receptor[136]. The optimized Fc domain improves binding to CD16A, a low-affinity stimulatory receptor found on macrophages and natural killer cells. The optimized Fc domain imparts enhanced ADCC action to the antibody[137-140]. A Phase I clinical trial evaluating magtuximab in patients with HER2-positive solid tumors, including gastric cancer, showed that 12% of patients were assessed as having a partial response and 50% were assessed as having stable disease. In addition, magtuximab combined with PD-1 inhibitors inhibits tumor growth by enhancing innate immunity. The CP-MGAH22-05 trial evaluated the response of magtuximab in combination with pabolizumab in HER2-positive gastroesophageal adenocarcinoma patients with an objective response rate (ORR) of 18.48% and was safe and well tolerated[141-144]. The combination of an FC-optimized anti-HER2 drug (Magtuximab) with an anti-PD-1 checkpoint blocker (pembrolizumab) synergistically enhances the activity of these anti-tumor agents[145]. The MAHOGANY trial combined PD-L1 inhibitor riflezumab with magtuximab in the first-line treatment of gastric cancer, and the results showed that ORR was 53%, the median duration of response was 10.3 months, and the disease control rate was 73%[146-148]. The above results suggest that the combination of dual-targeted drugs is effective in the treatment of gastric cancer.

TKI

HER2 is a transmembrane tyrosine kinase receptor, and TKI represented by Lapatinib blocks HER2 signaling by competing with ATP in the catalytic domain of the HER2 molecule, thereby preventing autophosphorylation and subsequent downstream signal transduction events[149]. In theory, TKI has an advantage over monoclonal antibodies because TKI binds to the intracellular domain and thus avoids monoclonal antibody resistance caused by the truncated form (P95) of HER2 (Figure 4). Monoclonal antibody has a large molecular weight and cannot effectively cross the blood-brain barrier, which has certain limitations in the treatment of patients with brain metastases[150]. In contrast, TKI has a small molecular weight and can play a role across the blood-brain barrier. At the same time, TKI has good oral bioavailability and is an ideal therapeutic drug (Figure 5).

Figure 4
Figure 4 Strategies to overcome mutation-based TKI resistance. A: PDB: 5XFF; B: PDB: 4V01; C: PDB: 4R6V; D: PDB: 6JPJ. Created with BioRender.com (see Supplementary material).
Figure 5
Figure 5 Schematic diagram of TKI-FGFR pathway activation in gastric cancer cells. Created with BioRender.com (see Supplementary material).
LAPATINIB

Lapatinib is an oral TKI that blocks HER1 and HER2 tyrosine kinase activity by binding to the ATP-binding site of the intracellular domain of the recipient cell, thereby inhibiting tumor cell growth[151,152]. In the LOGiC trial, lapatinib was combined with first-line chemotherapy capecitabine plus oxaliplatin in the treatment of HER2-positive gastric cancer. mOS in the lapatinib group and placebo group was 12.2 months and 10.5 months, respectively[153-155]. Although the OS in the lapatinib group was longer, the difference was not statistically significant. In addition, the response rate was 53% in the lapatinib group compared with 39% in the placebo group, and the OS was prolonged in the Asian population and younger patients, suggesting that lapatinib may still play a large role in the treatment of gastric cancer. In the EORTC 40071 trial, mPFS was 8.0 months in the lapatinib group and 5.9 months in the placebo group, suggesting that lapatinib combined with epirubicin, cisplatin, 5-FU, and capecitabine can significantly improve PFS in gastric cancer. Lapatinib did not improve OS in chemotherapy for second-line gastric cancer (mOS 11 months vs 8.9 months for lapatinib and paclitaxel alone)[156-160]. In the treatment of metastatic gastric cancer, a trial found that PFS and OS were comparable when treated with lapatinib alone vs lapatinib combined with capecitabine after platinum therapy failed. In another trial, lapatinib was used as a neoadjuvant therapy for gastric cancer, but the complete response rate was only 8%[161-164]. Although the above trial results did not reach a consistent positive conclusion, it can suggest that lapatinib can still play a significant role in the treatment of gastric cancer after selecting the right target population.

PYROTINIB

Pyrrotinib is a novel and irreversible TKI with extensive anti-EGFR (HER1, HER2 and HER4) activities[165]. By covalently binding to intracellular ATP binding sites, pyrrotinib inhibits the formation of homologous/heterodimer of HER family and self-phosphorylation, thereby blocking tumor cell cycle and limiting tumor development. mPFS of gastric cancer patients treated with pyrrotinib were 2.9 months, mOS was 5.9 months, disease control rate was 100%, and ORR was 50%, indicating that the treatment of pyrrotinib has a good effect on HER2-positive advanced gastric cancer patients, and its toxic effects can be well controlled[166-170]. In another retrospective study, gastric cancer patients treated with pyrrotinib had PFS of 142 d and mOS of 179 d, which also revealed that pyrrotinib has significant antitumor activity and good safety profile (Figure 6). However, the sample sizes collected in these trials are small, and the role of pyrrotinib in gastric cancer remains to be demonstrated by further larger clinical studies[171-173].

Figure 6
Figure 6 Mechanisms of alternative signaling activation induced FGFR-TKI resistance in gastric cancer. Created with BioRender.com (see Supplementary material).
ADC

ADCs consist of recombinant monoclonal antibodies that are covalently bound to cytotoxic drugs. Antibodies can be internalized after binding with specific antigens on the surface of tumor cells, so that cytotoxic drugs can be released inside cells. Another ADC is designed to promote the release of drugs from target cells to extracellular space, or release cytotoxic drugs from antibodies after binding with internalized preantigens, which can achieve the purpose of killing target cells and surrounding cells. High levels of HER2 expression are not necessarily required[174-176]. This may be beneficial for the treatment of stomach cancer. At present, the clinical ADCs for HER2-positive gastric cancer mainly include T-DM1 and T-DXd.

T-DM1

T-DM1 consists of trastuzumab and the small molecule microtubule inhibitor DM1 (maidensin)[177]. In the treatment of breast cancer, T-DM1 has achieved good efficacy and has been approved by the US FDA as a second-line treatment for HER2-positive advanced breast cancer. Based on this, many trials[178-180] have investigated its efficacy in the treatment of gastric cancer. The GATSBY trial evaluated the efficacy of T-DM1 and taxanes in HER2-positive advanced gastric or gastroesophageal junction carcinoma patients who had previously received chemotherapy and failed targeted therapy. The results showed that T-DM1 was not superior to taxanes in HER2-positive advanced gastric cancer patients who had previously received chemotherapy and failed targeted therapy (mOS was 7.9 months vs 8.6 months), and the results of the secondary efficacy endpoint PFS analysis did not reach a positive conclusion, which may be related to the HER2-positive patients with HER2 negative conversion[181]. Therefore, there is still a need to explore other therapeutic drugs for these patients.

T-DXd

T-DXd is a novel ADC composed of trastuzumab, cleavable tetrapeptidase splitter and human DNA topoisomerase I inhibitor[182]. In the Phase I dose-escalation study of T-DXd in patients with advanced HER2-positive gastric cancer, the recommended dose of 5.4 mg/kg or 6.4 mg/kgT-DXd was administered intravenously every 3 wk, and 43.2% of patients responded objectively in the dose-expansion trial[183-185]. When T-DXd was used in HER2-positive gastric cancer patients who had previously failed treatment with two or more fluoropyrimidine, platinum-based drugs, and trastuzumab (or an approved biosimilar), the difference in OS was statistically significant (12.5 months vs 8.4 months)[186]. It provides a new and effective drug for HER2-positive gastric cancer patients who have failed chemotherapy and targeted therapy.

RESEARCH PROGRESS OF ADC AND ICI IN HER2 ADVANCED GASTRIC CANCER

Immunotherapy is a new tumor treatment model. In advanced gastric cancer, the strategies of immunotherapy mainly include adoptive cell therapy, tumor vaccine and immune checkpoint regulation[187]. In recent years, the field of immunotherapy for advanced gastric cancer, especially targeted immune checkpoint therapy, has developed rapidly. The CheckMate 649 study established the important position of ICI in the immunotherapy of advanced gastric cancer. The latest research results were updated at the European Society of Medical Oncology Congress in 2022, which further confirmed the effect of nebuliuzumab combined with chemotherapy in the whole population, and the ORR of all randomized patients increased from 46% to 58%[188-190]. Both PFS and OS are extended and secure. These follow-up results further support the use of nabuliumab in combination with chemotherapy as the first-line standard of care for patients with advanced gastric cancer. In the Phase III KEYNOTE 062 trial, pembrolizumab, a PD-1 inhibitor, was no less effective than chemotherapy as a first-line treatment for advanced gastric cancer, either alone or in combination with chemotherapy, and had fewer aes. PD-1 inhibitors are also beneficial for the treatment of HER2-positive gastric cancer. In the Phase III KEYNOTE-811 study, adding pembrolizumab to trastuzumab plus chemotherapy for HER2-positive gastric cancer or gastroesophageal junction carcinoma, trastuzumab plus chemotherapy combined with pembrolizumab significantly reduced tumor size. The ORR of the patients was significantly improved. This study is another major breakthrough after the ToGA study, rewriting the first-line treatment guidelines for HER2-positive gastric cancer, and opening a new era of "immunotherapy + targeted therapy + chemotherapy" treatment for HER2-positive gastric cancer[191]. On this basis, a series of explorations have been carried out on new anti-HER2 drugs and their combination methods. ZW25, a bi-specific antibody targeting the ECD4 and ECD2 domains of HER2, also achieved an ORR and a median PFS of 10.9 months similar to KEYMATe-811 in HER2-positive gastric cancer patients treated in combination with PD-1 inhibitors and chemotherapy[192]. In addition, cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is another important checkpoint (Figure 7).

Figure 7
Figure 7 Schematic diagram of the effect of PD-1/CTLA-4 signaling on the immune microenvironment of gastric cancer. ICB: Immune checkpoint blockade; ADR: Adverse drug reaction; MHC: Major histocompatibility complex. Created with BioRender.com (see Supplementary material).

However, CTLA-4 checkpoint inhibitors have not been successful in gastric cancer. A PD-1/CTLA-4 dual-target inhibitor currently under development, cadonilimab (AK104) is the first PD-1/CTLA-4 bisspecific antibody. In a Phase I b/ II study of AK104 in combination with chemotherapy for the first-line treatment of gastric or gastroesophageal borderline cancer (NCT03852251), AK104 showed good anticancer activity and a manageable safety profile[193]. A Phase III study of AK104 combined with chemotherapy as first-line treatment for gastric or gastroesophageal borderline cancer (NCT05008783) is ongoing[194].

ADC can improve the degree of T lymphocyte infiltration in gastric cancer tissue, enhance the anti-tumor immune response, and thus improve the therapeutic effect of PD-1 inhibitors. Therefore, a promising therapeutic strategy is ADC combined with ICI. ADC enhances anti-tumor immune response in the following three aspects: First, ADC can induce anti-tumor immune activity by releasing cytotoxic payloads within cancer cells; Secondly, ADC can significantly improve the immune response by directly killing cancer cells or non-targeted cancer cells through enrichment on the tumor surface; Third, cytotoxic payloads can induce antigen-presenting dendritic cells to mature to enhance anti-tumor immune responses. At present, some preclinical studies have confirmed the potential of ADC combined with ICI in the treatment of gastric cancer. Sato et al[195] found that XMT-1522 can induce immune cell death in vitro and enhance CD8+T cell infiltration and PD-1 expression level of CD8+ cells after treating multiple gastric cancer cell lines. Yuan et al[196] found that T-DXd could increase the number of tumor infiltrating dendritic cells and up-regulate their CD86 expression. In addition, T-DXD also increased the number of tumor-infiltrating CD8+T cells and enhanced the expression levels of PD-L1 and major histocompatibility complex class I molecules on tumor cells. When combined with ICI, both XMT-1522 and T-DXd showed synergistic antitumor activity. Therefore, the combination therapy composed of ADC and ICI shows an anticancer effect of "1 + 1 > 2", which is expected to become a promising treatment strategy for HER2-positive gastric cancer, but it needs to be confirmed by large randomized controlled clinical studies.

CHALLENGES IN THE APPLICATION AND DEVELOPMENT OF ADCS

As the fastest growing HER2-targeting drug at present, ADC faces three major challenges in the application and development process: First, how to improve the uptake of ADC by tumor cells is the main difficulty in its development[197]. ADC mainly relies on the high expression of target antigen on the surface of cancer cells to ensure its effective endocytosis and release cytotoxic payloads. The killing effect of HER2 ADC is closely related to the expression level of HER2 on the surface of gastric cancer cells, and requires high expression of HER2 on the surface of gastric cancer cells. Gastric cancer limits the therapeutic effectiveness of existing ADCs. Therefore, improving the cancer cell uptake of ADC can significantly improve its anticancer activity, especially for HER2-low expressing gastric cancer. Second, non-targeted toxicity is one of the main factors leading to the failure of ADC clinical trials. Non-targeted toxicity is associated with a variety of factors, including mAb, cytotoxic payloads, linkers and target antigens, as well as the internalization of ADC by cancer cells, non-specific binding of antibodies to Fc receptors, premature hydrolysis of linkers to release payloads, and the "bystander" effect of overstrong ADCC action on normal cells. Thirdly, low expression of target antigen is another important factor in off-target toxicity of ADC[198]. ADC resistance is another problem that needs to be overcome. The results suggest that ineffective internalization of ADC and lysosomal degradation may be the main mechanism of T-DM1 inactivation. It is also possible that cancer cells develop resistance to T-DM1 through upregulation of the drug efflux pump or by alternating the use of tubulin/microtubule-associated proteins. In addition, trastuzumab related changes may also contribute to the development of T-DM1 resistance, including reduced HER2 expression levels, the emergence of a truncated form of HER2, or mutations in the ERBB2 gene. At the same time, some key issues should also be addressed in the future, including optimizing each ADC component to improve its anti-cancer activity; Accelerate the exploration of other targets in addition to HER2, such as ADCs against Trop-2, C-MET, claudin 18.2, and guanylate cyclase C.

CONCLUSION

Since the ToGA trial started the treatment of HER2-positive gastric cancer with trastuzumab, trastuzumab has played a huge role in the first-line treatment of gastric cancer, but in the second-line treatment, the efficacy of trastuzumab is not significant due to the emergence of drug resistance. At the same time, both pertuzumab and magtuximab have shown certain clinical application value in the treatment of gastric cancer. Small molecule TKI drugs lapatinib and pyrrotinib have shown certain efficacy in some clinical trials, but when combined with chemotherapy drugs, they do not significantly improve the survival rate of all patients. However, they still show irreplaceable therapeutic effects in some gastric cancer patients. In addition, the ADC drug T-DM1 did not significantly extend survival in patients with gastric cancer who had failed chemotherapy or targeted therapy, but T-DXd may be the treatment of choice for this subset of patients. Therefore, more detailed classification of HER2-positive gastric cancer patients is needed, which will help the use of various targeted drugs to better exert anti-tumor effects. At present, many drugs are still under study, such as monoclonal antibody initumab, TKI drugs lenatinib, Tucatinib, and ADC SYD985, which may play a good anti-tumor role in the treatment of gastric cancer in the future. At the same time, it is also expected that new and more effective anti-tumor drugs will be developed against the target HER2.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade C, Grade C

Novelty: Grade A, Grade B, Grade C

Creativity or Innovation: Grade A, Grade B, Grade C

Scientific Significance: Grade A, Grade B, Grade C

P-Reviewer: Ding J, China; Jin C, China; Maslennikov R, Russia S-Editor: Lin C L-Editor: A P-Editor: Zhao S

References
1.  Zeng Y, Jin RU. Molecular pathogenesis, targeted therapies, and future perspectives for gastric cancer. Semin Cancer Biol. 2022;86:566-582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 33]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
2.  Patel TH, Cecchini M. Targeted Therapies in Advanced Gastric Cancer. Curr Treat Options Oncol. 2020;21:70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 110]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
3.  Meric-Bernstam F, Johnson AM, Dumbrava EEI, Raghav K, Balaji K, Bhatt M, Murthy RK, Rodon J, Piha-Paul SA. Advances in HER2-Targeted Therapy: Novel Agents and Opportunities Beyond Breast and Gastric Cancer. Clin Cancer Res. 2019;25:2033-2041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 201]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
4.  Chen Z, Li Y, Tan B, Zhao Q, Fan L, Li F, Zhao X. Progress and current status of molecule-targeted therapy and drug resistance in gastric cancer. Drugs Today (Barc). 2020;56:469-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
5.  Narita Y, Muro K. Challenges in molecular targeted therapy for gastric cancer: considerations for efficacy and safety. Expert Opin Drug Saf. 2017;16:319-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
6.  Yang W, Raufi A, Klempner SJ. Targeted therapy for gastric cancer: molecular pathways and ongoing investigations. Biochim Biophys Acta. 2014;1846:232-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
7.  Zeng XQ, Wang J, Chen SY. Methylation modification in gastric cancer and approaches to targeted epigenetic therapy (Review). Int J Oncol. 2017;50:1921-1933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
8.  Liu L, Cao L, Gong B, Yu J. Novel biomarkers for the identification and targeted therapy of gastric cancer. Expert Rev Gastroenterol Hepatol. 2015;9:1217-1226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
9.  Gambardella V, Castillo J, Tarazona N, Gimeno-Valiente F, Martínez-Ciarpaglini C, Cabeza-Segura M, Roselló S, Roda D, Huerta M, Cervantes A, Fleitas T. The role of tumor-associated macrophages in gastric cancer development and their potential as a therapeutic target. Cancer Treat Rev. 2020;86:102015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 159]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
10.  Cann C, Ciombor KK. Systemic therapy for gastric cancer: Perioperative strategies and beyond. J Surg Oncol. 2022;125:1151-1160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
11.  Zhao TT, Xu H, Xu HM, Wang ZN, Xu YY, Song YX, Yin SC, Liu XY, Miao ZF. The efficacy and safety of targeted therapy with or without chemotherapy in advanced gastric cancer treatment: a network meta-analysis of well-designed randomized controlled trials. Gastric Cancer. 2018;21:361-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 43]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
12.  Jiang L, Gong X, Liao W, Lv N, Yan R. Molecular targeted treatment and drug delivery system for gastric cancer. J Cancer Res Clin Oncol. 2021;147:973-986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
13.  Catanese S, Lordick F. Targeted and immunotherapy in the era of personalised gastric cancer treatment. Best Pract Res Clin Gastroenterol. 2021;50-51:101738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
14.  Rao X, Zhang C, Luo H, Zhang J, Zhuang Z, Liang Z, Wu X. Targeting Gastric Cancer Stem Cells to Enhance Treatment Response. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 19]  [Reference Citation Analysis (0)]
15.  Rosenbaum MW, Gonzalez RS. Targeted therapy for upper gastrointestinal tract cancer: current and future prospects. Histopathology. 2021;78:148-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
16.  Ucaryilmaz Metin C, Ozcan G. Comprehensive bioinformatic analysis reveals a cancer-associated fibroblast gene signature as a poor prognostic factor and potential therapeutic target in gastric cancer. BMC Cancer. 2022;22:692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
17.  Garrido M, Fonseca PJ, Vieitez JM, Frunza M, Lacave AJ. Challenges in first line chemotherapy and targeted therapy in advanced gastric cancer. Expert Rev Anticancer Ther. 2014;14:887-900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
18.  Xu W, Yang Z, Lu N. Molecular targeted therapy for the treatment of gastric cancer. J Exp Clin Cancer Res. 2016;35:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 122]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
19.  Cetin B, Gumusay O, Cengiz M, Ozet A. Advances of Molecular Targeted Therapy in Gastric Cancer. J Gastrointest Cancer. 2016;47:125-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
20.  Yazici O, Sendur MA, Ozdemir N, Aksoy S. Targeted therapies in gastric cancer and future perspectives. World J Gastroenterol. 2016;22:471-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
21.  Al-Batran SE, Werner D. Recent advances and future trends in the targeted therapy of metastatic gastric cancer. Expert Rev Gastroenterol Hepatol. 2014;8:555-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
22.  Tomasello G, Ghidini M, Liguigli W, Ratti M, Toppo L, Passalacqua R. Targeted therapies in gastric cancer treatment: where we are and where we are going. Invest New Drugs. 2016;34:378-393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
23.  Pellino A, Riello E, Nappo F, Brignola S, Murgioni S, Djaballah SA, Lonardi S, Zagonel V, Rugge M, Loupakis F, Fassan M. Targeted therapies in metastatic gastric cancer: Current knowledge and future perspectives. World J Gastroenterol. 2019;25:5773-5788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 57]  [Cited by in F6Publishing: 82]  [Article Influence: 16.4]  [Reference Citation Analysis (1)]
24.  Pectasides E. Genomic Alterations and Targeted Therapy in Gastric and Esophageal Adenocarcinoma. Clin Ther. 2016;38:1589-1599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
25.  Lim SM, Lim JY, Cho JY. Targeted therapy in gastric cancer: personalizing cancer treatment based on patient genome. World J Gastroenterol. 2014;20:2042-2050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
26.  Yoo C, Park YS. Companion diagnostics for the targeted therapy of gastric cancer. World J Gastroenterol. 2015;21:10948-10955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 11]  [Cited by in F6Publishing: 9]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
27.  Aichler M, Luber B, Lordick F, Walch A. Proteomic and metabolic prediction of response to therapy in gastric cancer. World J Gastroenterol. 2014;20:13648-13657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
28.  Pazo Cid RA, Antón A. Advanced HER2-positive gastric cancer: current and future targeted therapies. Crit Rev Oncol Hematol. 2013;85:350-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
29.  Wang S, Yuan L. Predictive biomarkers for targeted and cytotoxic agents in gastric cancer for personalized medicine. Biosci Trends. 2016;10:171-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
30.  Feng W, Zhu X. Efficacy prediction of targeted therapy for gastric cancer: The current status (Review). Mol Med Rep. 2018;18:1238-1246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
31.  Kim JG. Molecular targeted therapy for advanced gastric cancer. Korean J Intern Med. 2013;28:149-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
32.  Harada K, Yamashita K, Iwatsuki M, Baba H, Ajani JA. Intraperitoneal therapy for gastric cancer peritoneal carcinomatosis. Expert Rev Clin Pharmacol. 2022;15:43-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
33.  Luis M, Tavares A, Carvalho LS, Lara-Santos L, Araújo A, de Mello RA. Personalizing therapies for gastric cancer: molecular mechanisms and novel targeted therapies. World J Gastroenterol. 2013;19:6383-6397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 23]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
34.  Aoyama T, Yoshikawa T. Adjuvant therapy for locally advanced gastric cancer. Surg Today. 2017;47:1295-1302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
35.  Huang Y, Wei J, Liu BR. [Research advances of K-ras mutation in the prognosis and targeted therapy of gastric cancer]. Zhonghua Zhong Liu Za Zhi. 2016;38:81-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
36.  Farran B, Müller S, Montenegro RC. Gastric cancer management: Kinases as a target therapy. Clin Exp Pharmacol Physiol. 2017;44:613-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
37.  Wu L, Zhong Y, Yu X, Wu D, Xu P, Lv L, Ruan X, Liu Q, Feng Y, Liu J, Li X. Selective poly adenylation predicts the efficacy of immunotherapy in patients with lung adenocarcinoma by multiple omics research. Anticancer Drugs. 2022;33:943-959.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
38.  Ding J, Kang X, Feng M, Tan J, Feng Q, Wang X, Wang J, Liu J, Li Z, Guan W, Qiao T. A novel active mitochondrion-selective fluorescent probe for the NIR fluorescence imaging and targeted photodynamic therapy of gastric cancer. Biomater Sci. 2022;10:4756-4763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
39.  Ang YL, Yong WP, Tan P. Translating gastric cancer genomics into targeted therapies. Crit Rev Oncol Hematol. 2016;100:141-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
40.  Murayama Y, Oritani K, Tsutsui S. Novel CD9-targeted therapies in gastric cancer. World J Gastroenterol. 2015;21:3206-3213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
41.  Tabernero J, Macarulla T, Ramos FJ, Baselga J. Novel targeted therapies in the treatment of gastric and esophageal cancer. Ann Oncol. 2005;16:1740-1748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 59]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
42.  Zheng T, Jin J, Zhang Y, Zhou L. Efficacy and safety of paclitaxel with or without targeted therapy as second-line therapy in advanced gastric cancer: A meta-analysis. Medicine (Baltimore). 2020;99:e20734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
43.  Lyons TG, Ku GY. Systemic therapy for esophagogastric cancer: targeted therapies. Chin Clin Oncol. 2017;6:48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
44.  Cappetta A, Lonardi S, Pastorelli D, Bergamo F, Lombardi G, Zagonel V. Advanced gastric cancer (GC) and cancer of the gastro-oesophageal junction (GEJ): focus on targeted therapies. Crit Rev Oncol Hematol. 2012;81:38-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
45.  Zhang J, Zhao T, Han F, Hu Y, Li Y. Photothermal and gene therapy combined with immunotherapy to gastric cancer by the gold nanoshell-based system. J Nanobiotechnology. 2019;17:80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
46.  Ye M, Xiu LJ, Ji QQ, Zhang YC, Sun YW, Zhao Y, Wang D, Li YJ, Wang XW, Yue XQ, Sun DZ. Research progress in targeted therapies for gastric cancer. Int J Clin Pharmacol Ther. 2022;60:509-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
47.  Zhou H, Yuan M, Yu Q, Zhou X, Min W, Gao D. Autophagy regulation and its role in gastric cancer and colorectal cancer. Cancer Biomark. 2016;17:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 61]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
48.  Jou E, Rajdev L. Current and emerging therapies in unresectable and recurrent gastric cancer. World J Gastroenterol. 2016;22:4812-4823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
49.  Bekaii-Saab T, El-Rayes B. Identifying and targeting cancer stem cells in the treatment of gastric cancer. Cancer. 2017;123:1303-1312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 80]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
50.  Zhou YL, Li YM, He WT. Application of Mesenchymal Stem Cells in the Targeted Gene Therapy for Gastric Cancer. Curr Stem Cell Res Ther. 2016;11:434-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
51.  Lin YH, Chen ZR, Lai CH, Hsieh CH, Feng CL. Active Targeted Nanoparticles for Oral Administration of Gastric Cancer Therapy. Biomacromolecules. 2015;16:3021-3032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
52.  Zhu ZG. [Current status and prospect of perioperative therapy for locally advanced gastric cancer]. Zhonghua Wei Chang Wai Ke Za Zhi. 2021;24:101-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
53.  Lee SY, Oh SC. Changing strategies for target therapy in gastric cancer. World J Gastroenterol. 2016;22:1179-1189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
54.  Costa G, Younes H, Kourie HR, Kattan J. The rapidly evolving landscape of advanced gastric cancer therapy. Future Oncol. 2022;18:1413-1416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
55.  Wu L, Zheng Y, Ruan X, Wu D, Xu P, Liu J, Li X. Long-chain noncoding ribonucleic acids affect the survival and prognosis of patients with esophageal adenocarcinoma through the autophagy pathway: construction of a prognostic model. Anticancer Drugs. 2022;33:e590-e603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
56.  Gerson JN, Skariah S, Denlinger CS, Astsaturov I. Perspectives of HER2-targeting in gastric and esophageal cancer. Expert Opin Investig Drugs. 2017;26:531-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
57.  Majeed W, Iftikhar A, Khaliq T, Aslam B, Muzaffar H, Atta K, Mahmood A, Waris S. Gastric Carcinoma: Recent Trends in Diagnostic Biomarkers and Molecular Targeted Therapies. Asian Pac J Cancer Prev. 2016;17:3053-3060.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Davidson M, Okines AF, Starling N. Current and Future Therapies for Advanced Gastric Cancer. Clin Colorectal Cancer. 2015;14:239-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
59.  Sunkar S, Neeharaika D, Nellore J, Nachiyar VC, Peela S. Small-Molecule-Targeted Therapies for Gastrointestinal Cancer: Successes and Failures. Crit Rev Oncog. 2020;25:311-333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
60.  Joo MK, Park JJ, Chun HJ. Recent updates of precision therapy for gastric cancer: Towards optimal tailored management. World J Gastroenterol. 2016;22:4638-4650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
61.  Durães C, Almeida GM, Seruca R, Oliveira C, Carneiro F. Biomarkers for gastric cancer: prognostic, predictive or targets of therapy? Virchows Arch. 2014;464:367-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 124]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
62.  Asaoka Y, Ikenoue T, Koike K. New targeted therapies for gastric cancer. Expert Opin Investig Drugs. 2011;20:595-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
63.  Chen J, Röcken C, Malfertheiner P, Ebert MP. Recent advances in molecular diagnosis and therapy of gastric cancer. Dig Dis. 2004;22:380-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
64.  Zhang LH, Ji JF. [Prognostic biomarkers and targeted therapies for gastric cancer]. Zhonghua Wei Chang Wai Ke Za Zhi. 2013;16:193-196.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Le DT, Ott PA, Korytowsky B, Le H, Le TK, Zhang Y, Maglinte GA, Abraham P, Patel D, Shangguan T, Chau I. Real-world Treatment Patterns and Clinical Outcomes Across Lines of Therapy in Patients With Advanced/Metastatic Gastric or Gastroesophageal Junction Cancer. Clin Colorectal Cancer. 2020;19:32-38.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
66.  Aguiar PN Jr, Muniz TP, Miranda RR, Tadokoro H, Forones NM, Monteiro ID, Castelo-Branco P, Janjigian YY, De Mello RA. Current advances in targeted therapies for metastatic gastric cancer: improving patient care. Future Oncol. 2016;12:839-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
67.  Wu L, Liu Q, Ruan X, Luan X, Zhong Y, Liu J, Yan J, Li X. Multiple Omics Analysis of the Role of RBM10 Gene Instability in Immune Regulation and Drug Sensitivity in Patients with Lung Adenocarcinoma (LUAD). Biomedicines. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
68.  Yoong J, Michael M, Leong T. Targeted therapies for gastric cancer: current status. Drugs. 2011;71:1367-1384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
69.  Lianos GD, Mangano A, Cho WC, Roukos DH. From standard to new genome-based therapy of gastric cancer. Expert Rev Gastroenterol Hepatol. 2015;9:1023-1026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
70.  Mu YP, Sun WJ, Lu CW, Su XL. MicroRNAs May Serve as Emerging Molecular Biomarkers for Diagnosis and Prognostic Assessment or as Targets for Therapy in Gastric Cancer. Asian Pac J Cancer Prev. 2015;16:4813-4820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
71.  Athauda A, Segelov E, Ali Z, Chau I. Integrative molecular analysis of colorectal cancer and gastric cancer: What have we learnt? Cancer Treat Rev. 2019;73:31-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
72.  Yong J, Li Y, Lin S, Wang Z, Xu Y. Inhibitors Targeting YAP in Gastric Cancer: Current Status and Future Perspectives. Drug Des Devel Ther. 2021;15:2445-2456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
73.  Ouyang J, Long Z, Li G. Circular RNAs in Gastric Cancer: Potential Biomarkers and Therapeutic Targets. Biomed Res Int. 2020;2020:2790679.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
74.  Irmak-Yazicioglu MB. Mechanisms of MicroRNA Deregulation and MicroRNA Targets in Gastric Cancer. Oncol Res Treat. 2016;39:136-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
75.  Shin VY, Chu KM. MiRNA as potential biomarkers and therapeutic targets for gastric cancer. World J Gastroenterol. 2014;20:10432-10439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 223]  [Cited by in F6Publishing: 257]  [Article Influence: 25.7]  [Reference Citation Analysis (1)]
76.  Zhang M, Deng W, Cao X, Shi X, Zhao H, Duan Z, Lv B, Liu B. Concurrent apatinib and local radiation therapy for advanced gastric cancer: A case report and review of the literature. Medicine (Baltimore). 2017;96:e6241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
77.  Iyikesici MS. Survival outcomes of metabolically supported chemotherapy combined with ketogenic diet, hyperthermia, and hyperbaric oxygen therapy in advanced gastric cancer. Niger J Clin Pract. 2020;23:734-740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
78.  Janjigian YY, Shah MA. Molecularly targeted therapies in advanced gastric cancer. Minerva Gastroenterol Dietol. 2011;57:75-88.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Ngeow J, Tan IB, Choo SP. Targeted therapies in the treatment of gastric cancer. Asia Pac J Clin Oncol. 2011;7:224-235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
80.  Lin F, Tan HJ, Guan JS, Lim YP. Divide and conquer: subproteomic approaches toward gastric cancer biomarker and drug target discovery. Expert Rev Proteomics. 2014;11:515-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
81.  Zhang F, Yin J, Zhang C, Han M, Wang X, Fu S, Du J, Zhang H, Li W. Affibody-Conjugated RALA Polymers Delivering Oligomeric 5-Fluorodeoxyuridine for Targeted Therapy of HER2 Overexpressing Gastric Cancer. Macromol Biosci. 2020;20:e2000083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
82.  Kang X, Li W, Liu W, Liang H, Deng J, Wong CC, Zhao S, Kang W, To KF, Chiu PWY, Wang G, Yu J, Ng EKW. LIMK1 promotes peritoneal metastasis of gastric cancer and is a therapeutic target. Oncogene. 2021;40:3422-3433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
83.  De Vita F, Giuliani F, Silvestris N, Rossetti S, Pizzolorusso A, Santabarbara G, Galizia G, Colucci G, Ciardiello F, Orditura M. Current status of targeted therapies in advanced gastric cancer. Expert Opin Ther Targets. 2012;16 Suppl 2:S29-S34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
84.  Tyczyńska M, Kędzierawski P, Karakuła K, Januszewski J, Kozak K, Sitarz M, Forma A. Treatment Strategies of Gastric Cancer-Molecular Targets for Anti-angiogenic Therapy: a State-of-the-art Review. J Gastrointest Cancer. 2021;52:476-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
85.  Ren Z, Sun J, Sun X, Hou H, Li K, Ge Q. Efficacy and safety of different molecular targeted agents based on chemotherapy for gastric cancer patients treatment: a network meta-analysis. Oncotarget. 2017;8:48253-48262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
86.  Kuo HY, Yeh KH. Molecular-targeted therapy for chemotherapy-refractory gastric cancer: a case report and literature review. Anticancer Res. 2014;34:3695-3699.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Ciliberto D, Staropoli N, Caglioti F, Gualtieri S, Fiorillo L, Chiellino S, De Angelis AM, Mendicino F, Botta C, Caraglia M, Tassone P, Tagliaferri P. A systematic review and meta-analysis of randomized trials on the role of targeted therapy in the management of advanced gastric cancer: Evidence does not translate? Cancer Biol Ther. 2015;16:1148-1159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
88.  Zagouri F, Papadimitriou CA, Dimopoulos MA, Pectasides D. Molecularly targeted therapies in unresectable-metastatic gastric cancer: a systematic review. Cancer Treat Rev. 2011;37:599-610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
89.  Weidle UH, Birzele F, Brinkmann U, Auslaender S. Gastric Cancer: Identification of microRNAs Inhibiting Druggable Targets and Mediating Efficacy in Preclinical In Vivo Models. Cancer Genomics Proteomics. 2021;18:497-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
90.  Kim HS, Lee H, Shin SJ, Beom SH, Jung M, Bae S, Lee EY, Park KH, Choi YY, Son T, Kim HI, Cheong JH, Hyung WJ, Park JC, Shin SK, Lee SK, Lee YC, Koom WS, Lim JS, Chung HC, Noh SH, Rha SY, Kim H, Paik S. Complementary utility of targeted next-generation sequencing and immunohistochemistry panels as a screening platform to select targeted therapy for advanced gastric cancer. Oncotarget. 2017;8:38389-38398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
91.  Chandra R, Balachandar N, Wang S, Reznik S, Zeh H, Porembka M. The changing face of gastric cancer: epidemiologic trends and advances in novel therapies. Cancer Gene Ther. 2021;28:390-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
92.  Aprile G, Giampieri R, Bonotto M, Bittoni A, Ongaro E, Cardellino GG, Graziano F, Giuliani F, Fasola G, Cascinu S, Scartozzi M. The challenge of targeted therapies for gastric cancer patients: the beginning of a long journey. Expert Opin Investig Drugs. 2014;23:925-942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
93.  Liu J, Liu YP. [Progress and prospects of international cancer drug clinical research on gastric cancer]. Zhonghua Wei Chang Wai Ke Za Zhi. 2020;23:196-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
94.  Weidle UH, Birzele F, Auslaender S, Brinkmann U. Down-regulated MicroRNAs in Gastric Carcinoma May Be Targets for Therapeutic Intervention and Replacement Therapy. Anticancer Res. 2021;41:4185-4202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
95.  Xie S, Zhang H, Wang X, Ge Q, Hu J. The relative efficacy and safety of targeted agents used in combination with chemotherapy in treating patients with untreated advanced gastric cancer: a network meta-analysis. Oncotarget. 2017;8:26959-26968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
96.  Zou K, Yang S, Zheng L, Yang C, Xiong B. Efficacy and safety of target combined chemotherapy in advanced gastric cancer: a meta-analysis and system review. BMC Cancer. 2016;16:737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
97.  Hescheler DA, Plum PS, Zander T, Quaas A, Korenkov M, Gassa A, Michel M, Bruns CJ, Alakus H. Identification of targeted therapy options for gastric adenocarcinoma by comprehensive analysis of genomic data. Gastric Cancer. 2020;23:627-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
98.  Wagner AD, Moehler M. Development of targeted therapies in advanced gastric cancer: promising exploratory steps in a new era. Curr Opin Oncol. 2009;21:381-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
99.  Koizumi K, Kato S, Sakurai H, Hashimoto I, Yasumoto K, Saiki I. Therapeutics target of CXCR4 and its downstream in peritoneal carcinomatosis of gastric cancer. Front Biosci (Schol Ed). 2012;4:269-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
100.  Carneiro F, Oliveira C, Leite M, Seruca R. Molecular targets and biological modifiers in gastric cancer. Semin Diagn Pathol. 2008;25:274-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
101.  Wu L, Zhong Y, Wu D, Xu P, Ruan X, Yan J, Liu J, Li X. Immunomodulatory Factor TIM3 of Cytolytic Active Genes Affected the Survival and Prognosis of Lung Adenocarcinoma Patients by Multi-Omics Analysis. Biomedicines. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
102.  Cao HY, Yuan AH, Chen W, Shi XS, Miao Y. A DNA aptamer with high affinity and specificity for molecular recognition and targeting therapy of gastric cancer. BMC Cancer. 2014;14:699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
103.  Ohtsu A, Sasako M. Overview of adjuvant therapy for resected gastric cancer: differences in Japan and the United States. Semin Oncol. 2005;32:S101-S104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
104.  Dionisi F, Avery S, Lukens JN, Ding X, Kralik J, Kirk M, Roses RE, Amichetti M, Metz JM, Plastaras JP. Proton therapy in adjuvant treatment of gastric cancer: planning comparison with advanced x-ray therapy and feasibility report. Acta Oncol. 2014;53:1312-1320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
105.  Wang J, Du L, Chen X. Adenosine signaling: Optimal target for gastric cancer immunotherapy. Front Immunol. 2022;13:1027838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
106.  Kim S, Barzi A, Rajdev L. Biomarker-driven targeted therapies for gastric/gastro-esophageal junction malignancies. Semin Oncol. 2018;45:133-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
107.  Wang X, Li G, Zhang Y, Bai S, Xu F, Wei Y, Gong Y. Single-arc volumetric-modulated arc therapy (sVMAT) as adjuvant treatment for gastric cancer: dosimetric comparisons with three-dimensional conformal radiotherapy (3D-CRT) and intensity-modulated radiotherapy (IMRT). Med Dosim. 2013;38:395-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
108.  Curea FG, Hebbar M, Ilie SM, Bacinschi XE, Trifanescu OG, Botnariuc I, Anghel RM. Current Targeted Therapies in HER2-Positive Gastric Adenocarcinoma. Cancer Biother Radiopharm. 2017;32:351-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
109.  Lee JA, Ahn YC, Lim DH, Park HC, Asranbaeva MS. Dosimetric and Clinical Influence of 3D Versus 2D Planning in Postoperative Radiation Therapy for Gastric Cancer. Cancer Res Treat. 2015;47:727-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
110.  Wu L, Li X, Qian X, Wang S, Liu J, Yan J. Lipid Nanoparticle (LNP) Delivery Carrier-Assisted Targeted Controlled Release mRNA Vaccines in Tumor Immunity. Vaccines (Basel). 2024;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
111.  Du S, Fang Z, Ye L, Sun H, Deng G, Wu W, Zeng F. Pretreatment neutrophil-to-lymphocyte ratio predicts the benefit of gastric cancer patients with systemic therapy. Aging (Albany NY). 2021;13:17638-17654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
112.  Shi J, Liu S, Yu Y, He C, Tan L, Shen YM. RGD peptide-decorated micelles assembled from polymer-paclitaxel conjugates towards gastric cancer therapy. Colloids Surf B Biointerfaces. 2019;180:58-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
113.  Arkenau HT, Saggese M, Lemech C. Advanced gastric cancer: is there enough evidence to call second-line therapy standard? World J Gastroenterol. 2012;18:6376-6378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
114.  Hashemi M, Aparviz R, Beickzade M, Paskeh MDA, Kheirabad SK, Koohpar ZK, Moravej A, Dehghani H, Saebfar H, Zandieh MA, Salimimoghadam S, Rashidi M, Taheriazam A, Entezari M, Samarghandian S. Advances in RNAi therapies for gastric cancer: Targeting drug resistance and nanoscale delivery. Biomed Pharmacother. 2023;169:115927.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
115.  Abdel-Rahman O. Targeting vascular endothelial growth factor (VEGF) pathway in gastric cancer: preclinical and clinical aspects. Crit Rev Oncol Hematol. 2015;93:18-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
116.  Padmanabhan N, Ushijima T, Tan P. How to stomach an epigenetic insult: the gastric cancer epigenome. Nat Rev Gastroenterol Hepatol. 2017;14:467-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 104]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
117.  Fährmann M. Targeting protein kinase C (PKC) in physiology and cancer of the gastric cell system. Curr Med Chem. 2008;15:1175-1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
118.  Yoshida K, Yamaguchi K, Okumura N, Osada S, Takahashi T, Tanaka Y, Tanabe K, Suzuki T. The roles of surgical oncologists in the new era: minimally invasive surgery for early gastric cancer and adjuvant surgery for metastatic gastric cancer. Pathobiology. 2011;78:343-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 36]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
119.  Yu L, Zhou GQ, Li DC. MiR-136 triggers apoptosis in human gastric cancer cells by targeting AEG-1 and BCL2. Eur Rev Med Pharmacol Sci. 2018;22:7251-7256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
120.  Khan U, Shah MA. Optimizing Therapies in the Perioperative Management of Gastric Cancer. Curr Treat Options Oncol. 2019;20:57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
121.  Li M, Chang J, Ren H, Song D, Guo J, Peng L, Zhou X, Zhao K, Lu S, Liu Z, Hu P. Downregulation of CCKBR Expression Inhibits the Proliferation of Gastric Cancer Cells, Revealing a Potential Target for Immunotoxin Therapy. Curr Cancer Drug Targets. 2022;22:257-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
122.  Pan SY, Fang JY. [Research progresses in targeted therapies for gastric cancer]. Zhonghua Nei Ke Za Zhi. 2020;59:148-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
123.  Azimi M, Totonchi M, Rahimi M, Firouzi J, Sahranavard P, Emami Razavi A, Memari F, Kamali F, Ebrahimi M. An integrated analysis to predict micro-RNAs targeting both stemness and metastasis in human gastric cancer. J Gastroenterol Hepatol. 2021;36:436-445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
124.  Lei Z, Fan J, Li X, Chen Y, Shi D, Xie H, Liu B. Biomimetic graphene oxide quantum dots nanoparticles targeted photothermal-chemotherapy for gastric cancer. J Drug Target. 2023;31:320-333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
125.  Wu L, Li H, Liu Y, Fan Z, Xu J, Li N, Qian X, Lin Z, Li X, Yan J. Research progress of 3D-bioprinted functional pancreas and in vitro tumor models. IJB. 2024;10:1256.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Briasoulis E, Liakakos T, Dova L, Fatouros M, Tsekeris P, Roukos DH, Kappas AM. Selecting a specific pre- or postoperative adjuvant therapy for individual patients with operable gastric cancer. Expert Rev Anticancer Ther. 2006;6:931-939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 71]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
127.  Abdel-Rahman O. Targeting the hepatocyte growth factor/mesenchymal epithelial transition pathway in gastric cancer: biological rationale and clinical applications. Expert Rev Anticancer Ther. 2015;15:235-245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
128.  Zheng Y, Li Z, Wang Y, Chen W, Lin Y, Guo J, Ye G. CircRNA: A new class of targets for gastric cancer drug resistance therapy. Pathol Oncol Res. 2023;29:1611033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
129.  Kim HM, Jeong I, Kim HJ, Kang SK, Kwon WS, Kim TS, Park KH, Jung M, Soong J, Lin SC, Chung HC, Rha SY. Casein Kinase 2 Inhibitor, CX-4945, as a Potential Targeted Anticancer Agent in Gastric Cancer. Anticancer Res. 2018;38:6171-6180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
130.  Geng H, Qian R, Zhang L, Yang C, Xia X, Wang C, Zhao G, Zhang Z, Zhu C. Clinical outcomes and potential therapies prediction of subgroups based on a ferroptosis-related long non-coding RNA signature for gastric cancer. Aging (Albany NY). 2022;14:6358-6376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
131.  Lee J, Kim ST, Kim K, Lee H, Kozarewa I, Mortimer PGS, Odegaard JI, Harrington EA, Lee J, Lee T, Oh SY, Kang JH, Kim JH, Kim Y, Ji JH, Kim YS, Lee KE, Kim J, Sohn TS, An JY, Choi MG, Lee JH, Bae JM, Kim S, Kim JJ, Min YW, Min BH, Kim NKD, Luke S, Kim YH, Hong JY, Park SH, Park JO, Park YS, Lim HY, Talasaz A, Hollingsworth SJ, Kim KM, Kang WK. Tumor Genomic Profiling Guides Patients with Metastatic Gastric Cancer to Targeted Treatment: The VIKTORY Umbrella Trial. Cancer Discov. 2019;9:1388-1405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 126]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
132.  Ayyappan S, Prabhakar D, Sharma N. Epidermal growth factor receptor (EGFR)-targeted therapies in esophagogastric cancer. Anticancer Res. 2013;33:4139-4155.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Yoshida K, Tsujimoto H, Matsumura K, Kinoshita M, Takahata R, Matsumoto Y, Hiraki S, Ono S, Seki S, Yamamoto J, Hase K. CD47 is an adverse prognostic factor and a therapeutic target in gastric cancer. Cancer Med. 2015;4:1322-1333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 85]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
134.  Feng Y, Sun T, Yu Y, Gao Y, Wang X, Chen Z. MicroRNA-370 inhibits the proliferation, invasion and EMT of gastric cancer cells by directly targeting PAQR4. J Pharmacol Sci. 2018;138:96-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
135.  Xu L, Zhang XY, Zhang SY. [In vitro and in vivo targeting therapy of immunoliposomes against human gastric cancer]. Zhonghua Yi Xue Za Zhi. 1994;74:83-86, 126.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Moehler M, Galle PR, Gockel I, Junginger T, Schmidberger H. The multidisciplinary management of gastrointestinal cancer. Multimodal treatment of gastric cancer. Best Pract Res Clin Gastroenterol. 2007;21:965-981.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
137.  Xia J, Wang H, Li S, Wu Q, Sun L, Huang H, Zeng M. Ion channels or aquaporins as novel molecular targets in gastric cancer. Mol Cancer. 2017;16:54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
138.  Chen Z, Huang W, Tian T, Zang W, Wang J, Liu Z, Li Z, Lai Y, Jiang Z, Gao J, Shen L. Characterization and validation of potential therapeutic targets based on the molecular signature of patient-derived xenografts in gastric cancer. J Hematol Oncol. 2018;11:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
139.  Zhang W. TCGA divides gastric cancer into four molecular subtypes: implications for individualized therapeutics. Chin J Cancer. 2014;33:469-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
140.  Pentheroudakis G, Stoyianni A. Incorporation of targeted agents in the management of patients with advanced gastric cancer. Curr Med Chem. 2011;18:1629-1639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
141.  Zhang SJ, Feng JF, Wang L, Guo W, Du YW, Ming L, Zhao GQ. miR-1303 targets claudin-18 gene to modulate proliferation and invasion of gastric cancer cells. Dig Dis Sci. 2014;59:1754-1763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
142.  Yashiro M, Matsuoka T. Fibroblast growth factor receptor signaling as therapeutic targets in gastric cancer. World J Gastroenterol. 2016;22:2415-2423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 26]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (2)]
143.  Matsukura N, Onda M, Shimada T. [Possibility and future problems of gene therapy for gastric cancer]. Nihon Geka Gakkai Zasshi. 2001;102:778-782.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Lu M, Wu Y, Zhang Y, Yu Y, Wang S, Su X. Immunotherapeutic strategy in the management of gastric cancer: molecular profiles, current practice, and ongoing trials. J Egypt Natl Canc Inst. 2023;35:32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
145.  Kataoka K, Deleersnijder A, Lordick F. Will molecular target agents enable the multidisciplinary treatment in stage IV gastric cancer? Eur J Surg Oncol. 2017;43:1835-1845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
146.  Carneiro P, Figueiredo J, Bordeira-Carriço R, Fernandes MS, Carvalho J, Oliveira C, Seruca R. Therapeutic targets associated to E-cadherin dysfunction in gastric cancer. Expert Opin Ther Targets. 2013;17:1187-1201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
147.  Wen L, Cheng F, Zhou Y, Yin C. MiR-26a enhances the sensitivity of gastric cancer cells to cisplatin by targeting NRAS and E2F2. Saudi J Gastroenterol. 2015;21:313-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 52]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
148.  Wu L, Zheng Y, Liu J, Luo R, Wu D, Xu P, Li X. Comprehensive evaluation of the efficacy and safety of LPV/r drugs in the treatment of SARS and MERS to provide potential treatment options for COVID-19. Aging (Albany NY). 2021;13:10833-10852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
149.  Sun W, Jiang C, Ji Y, Xiao C, Song H. Long Noncoding RNAs: New Regulators of Resistance to Systemic Therapies for Gastric Cancer. Biomed Res Int. 2021;2021:8853269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
150.  Ma YY, Tao HQ. Role of urokinase plasminogen activator receptor in gastric cancer: a potential therapeutic target. Cancer Biother Radiopharm. 2012;27:285-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
151.  Liu Y, Li D, Chen Y, Liu Y, Lin Y, Huang X, Wu T, Wang C, Ding J. Integrated bioinformatics analysis for conducting a prognostic model and identifying immunotherapeutic targets in gastric cancer. BMC Bioinformatics. 2023;24:191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
152.  Zhang Y, Zhu WG, Han JH, Zhang XY, Tao GZ, Li T, Yu CH. [Application of simplified intensity modulated radiation therapy in gastric cancer after operation]. Zhonghua Wei Chang Wai Ke Za Zhi. 2013;16:268-272.  [PubMed]  [DOI]  [Cited in This Article: ]
153.  Li Y, Qin C. MiR-1179 inhibits the proliferation of gastric cancer cells by targeting HMGB1. Hum Cell. 2019;32:352-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
154.  Labots M, Buffart TE, Haan JC, van Grieken NC, Tijssen M, van de Velde CJ, Grabsch HI, Ylstra B, Carvalho B, Fijneman RJ, Verheul HM, Meijer GA. High-level copy number gains of established and potential drug target genes in gastric cancer as a lead for treatment development and selection. Cell Oncol (Dordr). 2014;37:41-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
155.  Pandian J, Ganesan K. Delineation of gastric tumors with activated ERK/MAPK signaling cascades for the development of targeted therapeutics. Exp Cell Res. 2022;410:112956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
156.  Ali SM, Sanford EM, Klempner SJ, Rubinson DA, Wang K, Palma NA, Chmielecki J, Yelensky R, Palmer GA, Morosini D, Lipson D, Catenacci DV, Braiteh F, Erlich R, Stephens PJ, Ross JS, Ou SH, Miller VA. Prospective comprehensive genomic profiling of advanced gastric carcinoma cases reveals frequent clinically relevant genomic alterations and new routes for targeted therapies. Oncologist. 2015;20:499-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
157.  Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, Zhang S, Lee M, Wu J, Lim KH, Lei Z, Goh G, Lim QY, Tan AL, Sin Poh DY, Riahi S, Bell S, Shi MM, Linnartz R, Zhu F, Yeoh KG, Toh HC, Yong WP, Cheong HC, Rha SY, Boussioutas A, Grabsch H, Rozen S, Tan P. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut. 2012;61:673-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 478]  [Cited by in F6Publishing: 499]  [Article Influence: 41.6]  [Reference Citation Analysis (0)]
158.  Alani S, Soyfer V, Strauss N, Schifter D, Corn BW. Limited advantages of intensity-modulated radiotherapy over 3D conformal radiation therapy in the adjuvant management of gastric cancer. Int J Radiat Oncol Biol Phys. 2009;74:562-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
159.  Choi ES, Kim H, Kim HP, Choi Y, Goh SH. CD44v8-10 as a potential theranostic biomarker for targeting disseminated cancer cells in advanced gastric cancer. Sci Rep. 2017;7:4930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
160.  Lee J, Ou SH. Towards the goal of personalized medicine in gastric cancer--time to move beyond HER2 inhibition. Part I: Targeting receptor tyrosine kinase gene amplification. Discov Med. 2013;15:333-341.  [PubMed]  [DOI]  [Cited in This Article: ]
161.  Corso S, Giordano S. How Can Gastric Cancer Molecular Profiling Guide Future Therapies? Trends Mol Med. 2016;22:534-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
162.  DI Cicilia R, Mordenti P, Anselmi E, Paties C, Carella E, Cavanna L. HER2-positive gastric cancer showing marked thickening of the gastric wall on ultrasonographic and computed tomographic scans. a chance phenomenon or a specific behaviour of this cancer type? Report of three cases. Anticancer Res. 2014;34:5083-5086.  [PubMed]  [DOI]  [Cited in This Article: ]
163.  Xu C, Ooi WF, Qamra A, Tan J, Chua BY, Ho SWT, Das K, Adam Isa ZF, Li Z, Yao X, Yan T, Xing M, Huang KK, Lin JS, Nandi T, Tay ST, Lee MH, Tan ALK, Ong X, Ashktorab H, Smoot D, Li S, Ng SC, Teh BT, Tan P. HNF4α pathway mapping identifies wild-type IDH1 as a targetable metabolic node in gastric cancer. Gut. 2020;69:231-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
164.  Choi KM, Cho E, Bang G, Lee SJ, Kim B, Kim JH, Park SG, Han EH, Chung YH, Kim JY, Kim E. Activity-Based Protein Profiling Reveals Potential Dasatinib Targets in Gastric Cancer. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
165.  Liu J, Wang H, Liao X. MicroRNA-223-5p targets long non-coding RNA TP73 antisense RNA1 to promote the invasion of gastric cancer. Hum Cell. 2020;33:676-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
166.  Zou P, Xia Y, Chen W, Chen X, Ying S, Feng Z, Chen T, Ye Q, Wang Z, Qiu C, Yang S, Liang G. EF24 induces ROS-mediated apoptosis via targeting thioredoxin reductase 1 in gastric cancer cells. Oncotarget. 2016;7:18050-18064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
167.  Zhang H, Tian B, Yu H, Yao H, Gao Z. LAPTM4B-35 protein as a potential therapeutic target in gastric cancer. Tumour Biol. 2014;35:12737-12742.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
168.  Zhang X, Zhang J, Ma Y, Pei X, Liu Q, Lu B, Jin L, Wang J, Liu J. A cell-based single-stranded DNA aptamer specifically targets gastric cancer. Int J Biochem Cell Biol. 2014;46:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
169.  Iwagami S, Watanabe M, Baba H. Current status and problems in development of molecular target agents for gastric cancer in Japan. Nihon Shokakibyo Gakkai Zasshi. 2011;108:11-18.  [PubMed]  [DOI]  [Cited in This Article: ]
170.  Zhi Q, Guo X, Guo L, Zhang R, Jiang J, Ji J, Zhang J, Chen X, Cai Q, Li J, Liu B, Zhu Z, Yu Y. Oncogenic miR-544 is an important molecular target in gastric cancer. Anticancer Agents Med Chem. 2013;13:270-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
171.  Gong M, Lu Z, Fang G, Bi J, Xue X. A small interfering RNA targeting osteopontin as gastric cancer therapeutics. Cancer Lett. 2008;272:148-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
172.  Diniz F, Lamas S, Osório H, Aguiar P, Freitas D, Gärtner F, Sarmento B, Reis CA, Gomes J. Nanoparticles targeting Sialyl-Tn for efficient tyrosine kinase inhibitor delivery in gastric cancer. Acta Biomater. 2023;170:142-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
173.  Sohal DP, Sun W. What is the reference cytotoxic regimen for advanced gastric cancer: standard versus optimal therapy? Expert Rev Anticancer Ther. 2011;11:1843-1850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
174.  Fuereder T, Hoeflmayer D, Jaeger-Lansky A, Rasin-Streden D, Strommer S, Fisker N, Hansen BJ, Crevenna R, Wacheck V. Sphingosine kinase 1 is a relevant molecular target in gastric cancer. Anticancer Drugs. 2011;22:245-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
175.  Wang K, Gong Z, Chen Y, Zhang M, Wang S, Yao S, Liu Z, Huang Z, Fei B. KDM4C-mediated senescence defense is a targetable vulnerability in gastric cancer harboring TP53 mutations. Clin Epigenetics. 2023;15:163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
176.  Seo JH, Jeong ES, Choi YK. Therapeutic effects of lentivirus-mediated shRNA targeting of cyclin D1 in human gastric cancer. BMC Cancer. 2014;14:175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
177.  Chang HR, Nam S, Lee J, Kim JH, Jung HR, Park HS, Park S, Ahn YZ, Huh I, Balch C, Ku JL, Powis G, Park T, Jeong JH, Kim YH. Systematic approach identifies RHOA as a potential biomarker therapeutic target for Asian gastric cancer. Oncotarget. 2016;7:81435-81451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
178.  Kim Y, Hosoda Y, Nishino M, Okano M, Kawada J, Yamasaki M, Nagai K, Yasui M, Okuyama M, Tsujinaka T. [Hepatic Resection of Multiple Liver Metastases from Gastric Cancer after Molecular Targeted Chemotherapy(S-1 plus Cisplatin plus Trastuzumab)]. Gan To Kagaku Ryoho. 2015;42:1635-1637.  [PubMed]  [DOI]  [Cited in This Article: ]
179.  Wu L, Chen X, Zeng Q, Lai Z, Fan Z, Ruan X, Li X, Yan J. NR5A2 gene affects the overall survival of LUAD patients by regulating the activity of CSCs through SNP pathway by OCLR algorithm and immune score. Heliyon. 2024;10:e28282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
180.  Inoue M, Matsumoto S, Saito H, Tsujitani S, Ikeguchi M. Intraperitoneal administration of a small interfering RNA targeting nuclear factor-kappa B with paclitaxel successfully prolongs the survival of xenograft model mice with peritoneal metastasis of gastric cancer. Int J Cancer. 2008;123:2696-2701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
181.  Michiura T, Miyazaki S, Fujita S, Hirose H, Miyazaki Y, Sugimoto S, Yamabe K, Nagaoka M. [A case of advanced HER2- positive gastric cancer with a partial response to molecular targeted chemotherapy (S-1+CDDP+Trastuzumab)]. Gan To Kagaku Ryoho. 2014;41:2346-2348.  [PubMed]  [DOI]  [Cited in This Article: ]
182.  Yamamoto K, Nishikawa K, Hirao M, Fujitani K, Tsujinaka T, Maeda S, Uemura M, Miyake M, Hama N, Miyamoto A, Omiya H, Ikeda M, Takami K, Nakamori S, Sekimoto M. [Optimal Target Population for Adjuvant Chemotherapy for Pathological T1 Gastric Cancer]. Gan To Kagaku Ryoho. 2015;42:2084-2087.  [PubMed]  [DOI]  [Cited in This Article: ]
183.  Akamo Y, Mizuno I, Ichino T, Yamamoto T, Yasui T, Itabashi Y, Saito T, Kurahashi S, Tanimoto N, Shinagawa N. [Endoscopic injection of liposomal adriamycin targeting lymph node metastasis of gastric cancer]. Gan To Kagaku Ryoho. 1991;18:1822-1824.  [PubMed]  [DOI]  [Cited in This Article: ]
184.  Akamo Y, Mizuno I, Ichino T, Yamamoto T, Yasui T, Itabashi Y, Saito T, Kurahashi S, Tanimoto N, Shinagawa N. [Efficacy of preoperative adjuvant chemotherapy using adriamycin targeting the regional lymph nodes for gastric cancer--lymph node-targeting delivery of adriamycin in rabbits]. Gan To Kagaku Ryoho. 1992;19:451-455.  [PubMed]  [DOI]  [Cited in This Article: ]
185.  D'Ugo D, Agnes A, Grieco M, Biondi A, Persiani R. Global updates in the treatment of gastric cancer: a systematic review. Part 2: perioperative management, multimodal therapies, new technologies, standardization of the surgical treatment and educational aspects. Updates Surg. 2020;72:355-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
186.  Zhang Y, Wen Y. The Role of LncRNA NEAT1 in Gastric Cancer: Tumor Development and Potential Therapeutic Target. Curr Pharm Des. 2023;29:2213-2222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
187.  Iinuma H, Maruyama K, Okinaga K, Sasaki K, Sekine T, Ishida O, Ogiwara N, Johkura K, Yonemura Y. Intracellular targeting therapy of cisplatin-encapsulated transferrin-polyethylene glycol liposome on peritoneal dissemination of gastric cancer. Int J Cancer. 2002;99:130-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 180]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
188.  He H, Song F, Gao Q, Lu Z, Yuan Y, Li X, Chen L, Jia C, Yang R, Yang J, Duan W, Hou Y. The APEX1/miRNA-27a-5p axis plays key roles in progression, metastasis and targeted chemotherapy of gastric cancer. Int J Pharm. 2021;599:120446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
189.  Yang Y, Ma Y, Xiang X, Xing P, Wu Y, Zhang L, Tian Y. The prognostic value of the lymph node ratio for local advanced gastric cancer patients with intensity-modulated radiation therapy and concurrent chemotherapy after radical gastrectomy in China. Radiat Oncol. 2020;15:237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
190.  Aoyama T, Yoshikawa T, Fujikawa H, Hayashi T, Ogata T, Cho H, Yamada T, Hasegawa S, Tsuchida K, Yukawa N, Oshima T, Rino Y, Masuda M. Exploratory Analysis to Find Unfavorable Subset of Stage II Gastric Cancer for Which Surgery Alone Is the Standard Treatment; Another Target for Adjuvant Chemotherapy. Int Surg. 2014;99:835-841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
191.  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;470:350-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
192.  Liang H, Li Z, Huang Z, Wu C, Qiu Y, Liang Y, Chen X, Li F, Xu Z, Li G, Liu H, Zhao L. Prognostic characteristics and clinical response to immunotherapy targeting programmed cell death 1 for patients with advanced gastric cancer with liver metastases. Front Immunol. 2022;13:1015549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
193.  Li X, Xu J, Xie J, Yang W. Research progress in targeted therapy and immunotherapy for gastric cancer. Chin Med J (Engl). 2022;135:1299-1313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Reference Citation Analysis (0)]
194.  Wieland P, Dobler B, Mai S, Hermann B, Tiefenbacher U, Steil V, Wenz F, Lohr F. IMRT for postoperative treatment of gastric cancer: covering large target volumes in the upper abdomen: a comparison of a step-and-shoot and an arc therapy approach. Int J Radiat Oncol Biol Phys. 2004;59:1236-1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 74]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
195.  Sato T, Saito M, Nakajima S, Saito K, Katagata M, Fukai S, Okayama H, Sakamoto W, Saze Z, Momma T, Mimura K, Kono K. ARID1A deficiency is targetable by AKT inhibitors in HER2-negative gastric cancer. Gastric Cancer. 2023;26:379-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
196.  Yuan DD, Zhu ZX, Zhang X, Liu J. Targeted therapy for gastric cancer: Current status and future directions (Review). Oncol Rep. 2016;35:1245-1254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
197.  Pandey SS, Shaikh FI, Gupta AR, Vaidya RJ. Mannosylated Solid Lipid Nanocarriers of Chrysin to Target Gastric Cancer: Optimization and Cell Line Study. Curr Drug Deliv. 2021;18:1574-1584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
198.  Dong W, Li B, Wang J, Song Y, Zhang Z, Fu C. Exploration of the Radiotherapeutic Clinical Target Volume Delineation for Gastric Cancer from Lymph Node Metastases. Anticancer Res. 2017;37:4629-4634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]