BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 21, 2025; 31(39): 110986
Published online Oct 21, 2025. doi: 10.3748/wjg.v31.i39.110986
Targetable pathways for drug repurposing in gastric cancer
Alba Valero-Almingol, Sheyla Montori, Nayra Felípez, Gastroenterology Research Unit, Instituto de Investigación Sanitaria de Navarra, Navarrabiomed, Universidad Pública de Navarra, Hospital Universitario de Navarra, Pamplona 31008, Navarre, Spain
Enrique Santamaría, Joaquín Fernandez-Irigoyen, Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra, Universidad Pública de Navarra, Hospital Universitario de Navarra, Pamplona 31008, Navarre, Spain
Irina Luzko, Anabella A Cuestas, Carlos Pardo, Ricard Prat, Leticia Moreira, Department of Gastroenterology, Hospital Clínic Instituto de Investigaciones Biomédicas August Pi i Sunyer, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona 08036, Catalonia, Spain
Eduardo Albéniz, Department of Gastroenterology, Hospital Universitario de Navarra, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra, Universidad Pública de Navarra, Pamplona 31008, Navarre, Spain
ORCID number: Alba Valero-Almingol (0009-0007-6535-8301); Sheyla Montori (0000-0003-1836-1966); Nayra Felípez (0009-0001-8561-4794); Enrique Santamaría (0000-0001-8046-8102); Joaquín Fernandez-Irigoyen (0000-0001-5072-4099); Irina Luzko (0000-0002-8292-1596); Anabella A Cuestas (0009-0008-2632-5731); Ricard Prat (0000-0003-2852-2591); Leticia Moreira (0000-0002-4518-8591); Eduardo Albéniz (0000-0002-9311-3746).
Co-corresponding authors: Leticia Moreira and Eduardo Albéniz.
Author contributions: Valero-Almingol A, Montori S, Moreira L and Albéniz E conceptualized the study, reviewed and edited the manuscript; Valero-Almingol A performed the literature review and drafted the manuscript; Felípez N, Santamaría E, Fernández-Irigoyen J, Luzko I, Cuestas AA, Pardo C and Prat R made critical revisions; All the authors read and approved the final manuscript; Moreira L and Albéniz E contributed equally to the manuscript as co-corresponding authors by supervising and reviewing the work.
Supported by the Instituto de Salud Carlos III, No. PI24/00784, No. PI24/00737, No. PI21/01181, No. PI21/00333 and No. INT22/00112; and Departamento de Universidad, Innovación y Transformación Digital Gobierno de Navarra, No. 0011-1408-2024-000011 and No. 0011-1408-2022-000010.
Conflict-of-interest statement: Albéniz E reports a consultant agreement with Creo Medical and AGS Medtech and a grant from ISCIII- INT 22/00112 during this study. The other authors declare that they have no conflicts 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: Eduardo Albéniz, MD, Senior Researcher, Department of Gastroenterology, Hospital Universitario de Navarra, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra, Universidad Pública de Navarra, Calle Irunlarrea, 3, Pamplona 31008, Navarre, Spain. eduardo.albeniz.arbizu@navarra.es
Received: June 20, 2025
Revised: July 31, 2025
Accepted: September 19, 2025
Published online: October 21, 2025
Processing time: 123 Days and 19.5 Hours

Abstract

Gastric cancer (GC) is both the fifth most common cancer worldwide and the fifth in mortality. Owing to a lack of symptoms in the early stages and unspecific clinical presentation in the later stages, GC is usually diagnosed at advanced stages. This means that only approximately 60% of patients are eligible for curative treatment, and overall, GC patients have a 5-year survival rate of only 28.3%, underscoring the importance of developing new treatment strategies. Drug repurposing involves identifying new therapeutic uses for approved drugs and is a promising strategy for cancer treatment because of its lower cost and faster development time. A variety of targetable pathways are involved in GC progression, including the mitogen-activated protein kinase, phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin, p53, Janus kinase 2/signal transducer and activator of transcription 3, hypoxia-inducible factor-1α, wingless-type mouse mammary tumor virus integration site family/beta-catenin (Wnt/β-catenin), nuclear factor kappa B, and Hippo pathways. Therefore, the repurposing of drugs targeting these pathways represents an interesting option in the search for new treatments for GC. In this review, we explore some relevant pathways involved in the development of GC and the possibilities of repurposing drugs that target them.

Key Words: Gastric cancer; Drug repurposing; Signaling pathways; Correa’s cascade; Cancer therapy

Core Tip: Gastric cancer patients have a dismal prognosis when it is detected in advanced stages, so the search for new treatments is imperative to improve survival rates. In this review, we explored the potential of repurposed drugs for targeting pathways related to the development and progression of gastric cancer, as their lower costs and faster development make them promising candidates in the search for new therapies to treat this disease.



INTRODUCTION

Gastric cancer (GC) is both the fifth most common cancer worldwide and the fifth in mortality, representing almost 1000000 new cases and 650000 deaths in 2022[1]. The incidence of GC differs by region. The highest incidence rates are in East Asia, Central and Eastern Europe, and South America, and a lower incidence occurs in Western Europe and North America[2,3].

The histological classification of GC distinguishes between diffuse- and intestinal-type GC, with the latter type being more common and usually associated with Helicobacter pylori (H. pylori) infection[4]. Intestinal-type GC accounts for approximately 54% of GC cases, diffuse-type GC represents approximately 32%, and approximately 15% of cases present an indeterminate histology. Diffuse-type GC is more common in women and younger individuals; meanwhile, intestinal-type GC is more typically found in men and older individuals[5].

The risk factors for GC include genetic predisposition, age, sex, diet, obesity, smoking status, alcohol intake, Epstein-Barr virus infection, and H. pylori infection. Among these, H. pylori is the main risk factor and is responsible for most GC cases worldwide[4,6]. This bacterium causes chronic inflammation of the stomach that leads to GC through a serial step-by-step progression known as Correa’s cascade, which involves a transition from normal stomach tissue to atrophic gastritis, intestinal metaplasia, dysplasia, and finally invasive adenocarcinoma[7].

Owing to the absence of early-stage symptoms and the nonspecific nature of advanced-stage symptoms, GC is usually diagnosed in advanced stages, meaning that approximately 60% of patients are not eligible for curative treatment[3]. Although early-stage patients are eligible for surgery, which is sometimes combined with chemotherapy before or after surgery, for unresectable advanced-stage patients, chemotherapy is the main treatment used to prevent disease progression for as long as possible[8]. Overall, patients diagnosed with GC have a 28.3% 5-year survival rate[7], which underscores the importance of developing new treatment strategies for this disease.

Drug repurposing has emerged as a promising strategy for GC treatment. Drug repurposing consists of the use of already clinically approved drugs for new applications[9]. This can be accomplished through computational and experimental methods, although the most typical approach involves complementary use of these strategies[10,11].

Moreover, drug repurposing offers several advantages over traditional drug discovery methods, including reduced time and cost. De novo drug discovery is a highly expensive and risky process; it may take 10-15 years and cost more than 1 billion United States dollars, but only approximately 1% of compounds reach clinical trials. Drug repurposing significantly reduces these numbers, as the drugs have already undergone preclinical and clinical trials, and information about the drug compounds is already available[10,12]. In this review, we explore key molecular pathways involved in GC pathogenesis and explore potential drug repurposing opportunities targeting them.

PRENEOPLASTIC GASTRIC LESIONS

Correa’s cascade is a model of gastric carcinogenesis which proposes that intestinal-type GC arises as a result of a gradual change in the gastric mucosa. Normal gastric tissue transitions to non-atrophic gastritis, atrophic gastritis, intestinal metaplasia, low-grade dysplasia, high-grade dysplasia, and finally adenocarcinoma[7]. Each stage presents distinct molecular alterations that can lead to progression to the next stage[13].

Gastritis

The starting point of Correa’s cascade is chronic inflammation of the stomach, known as gastritis, which is often related to H. pylori infection[14,15] and can evolve into atrophic gastritis after the loss of gastric glands. The bacterium liberates toxins such as CagA that produce inflammation and dysregulate the molecular landscape of the stomach[13].

Several pathways are involved in the development of chronic atrophic gastritis. The nuclear factor kappa B (NF-κB) signaling pathway is closely related to inflammation and malignancies; thus, this pathway could represent a target for reversing damage to the gastric mucosa. Hedgehog signaling is also relevant, as its loss indicates the progression of chronic atrophic gastritis to subsequent stages[16].

The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK), and Wnt/β-catenin signaling pathways are also activated by H. pylori infection and are associated with disease progression[16]. In fact, a recent study showed that H. pylori activates the Wnt/β-catenin pathway in a mouse model and that the use of MoLuoDan, a traditional Chinese medicine, may inhibit this pathway and reverse some molecular damage present in gastritis[17].

Another relevant pathway is associated with the tumor suppressor p53, which is degraded during H. pylori infection via AKT1. The gene encoding this protein, TP53, has been found to be mutated as early as the gastritis phase[18].

Finally, the effects of the Hippo pathway on the development of gastritis are contradictory, with studies indicating both induction and protection from metaplasia due to its activation[16,19].

Intestinal metaplasia

Intestinal metaplasia is the next step in the GC cascade and is characterized by loss of the gastric epithelium and its transformation into an intestinal epithelium[14]. Intestinal metaplasia is already considered a precancerous lesion, greatly increasing the risk of GC, and it seems to be an irreversible point in the precancerous cascade[20,21].

Bile acids are important factors in the development of intestinal metaplasia, even in the absence of H. pylori[15,22], and one mechanism of action is the activation of the NF-κB signaling pathway[22]. Other pathways involved include Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3)[21], MAPK, and other molecular pathways involved in intestinal development such as the Wnt and Hedgehog pathways[20].

Several compounds have demonstrated efficacy in treating intestinal metaplasia. Mitogen-activated protein (MEK) inhibitors have been shown to reduce growth and signs of dysplasia in mouse organoid models[23]. Meanwhile, resveratrol, a natural compound that is present in the traditional Chinese medicine Polygonum cuspidatum, has been shown to inhibit the expression of metaplasia markers via regulation of the PI3K-AKT signaling pathway[24].

Dysplasia

In the dysplastic stage, the epithelium shows typical characteristics of neoplasia but no evidence of lamina propria invasion. Dysplasia is classified as either low-grade dysplasia, in which the architecture and features of the cell are more preserved, or high-grade dysplasia, in which a more atypical structure is observed. In high-grade dysplasia, complex glandular architecture and cytologic atypia with marked nuclei and large nucleoli are present along with a loss of cell polarity and high mitosis rates[14].

With respect to dysregulated pathways, evidence has shown that yes-associated protein (YAP), one of the main proteins in the Hippo signaling pathway, is overexpressed in high-grade dysplasia, and its crosstalk with the Wnt pathway has been connected to chemoresistance in GC[19]. The p53 pathway is also relevant at this stage, as approximately 88% of dysplasia samples have mutations in TP53[13], and inhibition of the NF-κB signaling pathway seems to promote dysplasia development[16]. Other important pathways include the MAPK and STAT3 pathways. These pathways have been shown to be inhibited by pyrvinim, which is effective against dysplastic cells[25].

CURRENT TREATMENT FOR GC

GC is defined by the penetration of neoplastic cells into the surrounding stroma through degradation of the stromal matrix. Depending on the depth of invasion of the gastric wall, GC can be classified as early, when it is limited to the mucosa or superficial submucosa, or advanced[7,14], when the neoplastic cells have invaded the deep submucosal layer or beyond, with subsequent distant dissemination potential.

Current European clinical guidelines consider three types of treatment according to the stage at diagnosis. Early-stage GC is treated with endoscopic resection, whereas for advanced stages, a combination of surgery and perioperative chemotherapy is the standard. Finally, in the case of advanced metastatic cancer, different lines of chemotherapy are applied with the goal of prolonging survival and improving the quality of life of patients[3,5,26].

Early GC treatment

According to European guidelines, early-stage IA cancers can be treated with either endoscopic or surgical resection, depending on whether deep submucosal infiltration exists. Survival rates at this stage are relatively high, and chemotherapy is not a standard treatment, as no benefit has been demonstrated[5].

Advanced GC treatment

From stages IB to III, surgical resection is the typical treatment for GC, but neoadjuvant (before surgery) and adjuvant (after surgery) chemotherapy may be added[26,27]. Although some Asian clinical guidelines do not have a strong recommendation for neoadjuvant chemotherapy, in Europe, the application of both (called perioperative chemotherapy) is standard[3,28]. European guidelines recommend four cycles of preoperative and postoperative 5-fluorouracil-leucovorin-oxaliplatin-docetaxel for patients who are able to tolerate that treatment, which includes a fluoropyrimidine, a platinum compound, and docetaxel. For unfit patients, a modified regimen may be administered that includes a reduced dose or a platinum and fluoropyrimidine doublet[3,26].

In stage IV patients, median survival is low, but chemotherapy has been shown to prolong survival and quality of life[5]. First-line chemotherapy commonly includes a combination of a platinum drug and a fluoropyrimidine. Complementary therapy is used for human epidermal growth factor receptor 2-positive (HER2 +) and programmed cell death ligand 1-positive patients, with the addition of trastuzumab and nivolumab/pembrolizumab, respectively, being beneficial[3,5].

For second-line therapy, paclitaxel, docetaxel, and irinotecan are used, sometimes with the addition of ramucirumab or immunotherapy. Finally, oral treatment with trifluridine and tipiracil is a typical third-line treatment, although taxane and irinotecan can also be considered[26]. Gastrectomy is generally not recommended for advanced metastatic stages[3].

POTENTIAL TARGETS FOR DRUG REPURPOSING IN GC

Drug repurposing is becoming an important part of drug development because of its lower costs and reduced risks, and several pathways may be targeted for the treatment of GC[8,29]. These pathways include the MAPK, PI3K/AKT/mTOR, p53, JAK2/STAT3, hypoxia-inducible factor-1α (HIF-1α), Wnt/β-catenin, NF-κB, and Hippo signaling pathways. A summary of the drugs and their target pathways is presented in Table 1.

Table 1 Candidates for drug repurposing in gastric cancer and target pathways[29-41,43-67,70-80,83,84,86-98,101-107,110-113].
Drug
Type
Approved use
Target pathway(s)/molecule(s)
Effects
Ref.
DoxyciclineAntibioticBacterial infectionMAPK, WntInhibit proliferation, colony formation, and spheroid growthMagnelli et al[30]; Pandian et al[31]
LovastatinStatinLower cholesterolMAPKInhibit growth, increase sensitivity of anti-HER2 therapy, radioprotective effects in healthy tissuesGao et al[32]; Zhang et al[33]; Du et al[34]; Rao et al[35]
SilymarinNatural productEpilepsy and hepato-protectionMAPKInhibit growth and proliferation in vivo and in vitroKoltai and Fliegel[36]; Kim et al[37]
Tegaserod5-HTR4 receptor agonistIrritable bowel syndromeMAPKInhibit growth in vivo and in vitroChen et al[38]; Wang et al[39]
TrametinibMEK inhibitorMelanomaMAPKSuppress tumor development and liver metastasis in mouse organoidsYamasaki et al[40]; Wang et al[41]
ATRALigand for retinoic acid and retinoic X receptorPromyelocytic leukemiaPI3K, WntSuppress proliferation in vivo and in vitroBouriez et al[43]; Zhang et al[44]; Jin et al[45]
Danusertib/alisertibAurora kinase inhibitorsNot approved (phase II for prostate cancer and phase III for lymphoma, respectively)MAPKInhibit proliferation, induce cell cycle arrest, and promote apoptosis and autophagyYuan et al[46]; Yuan et al[47]; Novais et al[48]
DeferasiroxIron chelatorIron overload diseasemTORInduce apoptosisChoi et al[49]
DronedaroneAntiarrhythmicAuricular fibrillationPI3KSuppress cell proliferation and colony formationLu et al[50]; CIMA[113]
MetforminBiguanideType 2 diabetesPI3KInduce apoptosis in vitroLu et al[51]; Lan et al[52]; Hu et al[53]
Mycophenolic acidIMPDH inhibitorOrgan transplant, lupus eritrematosusPI3KInhibit proliferationHsieh et al[54]; Dun et al[55]
NaftopidilΑ-1-adrenoceptor blockerBenign prostate hyperplasiaPI3KInduce apoptosisNakamura et al[56]; Kaku et al[57]; Florent et al[58]
RapamycinmTOR inhibitorOrgan transplantmTOR, HIF-1αReduce cell proliferation and invasivenessChen et al[59]; Morgos et al[60]; Liu et al[61]; CIMA[113]
SertralineAntidepressant (SSRI)Depression in cancer patientsPI3K, HIF-1αResensitize drug resistant GCMu et al[62]; Sánchez-Castillo et al[63]; Sun et al[64]; Wang et al[65]
VortioxetineAntidepressantDepressionPI3K, JAK2/STAT3Inhibit cell proliferation, migration, and invasion and induce apoptosis and autophagyChen et al[38]; Lv et al[66]; Li et al[67]
ThioridazineAntipsychoticSchizophreniaPI3K, WntReduce cell viability, induce apoptosis, and enhance anti-HER2 therapyYang et al[70]
TigecyclineAntibioticComplicated infectionsPI3KInduce autophagy and inhibit cell proliferationDong et al[71]; Tang et al[72]
Valproic acidHDAC1/2 inhibitorBipolar disorder, epilepsyPI3KInduce apoptosisZhang et al[33]; Fushida et al[73]; Sun et al[74]
EconazoleAntifungalSkin infectionsp53, PI3KCaspase-dependent apoptosis (p53 activation); inhibit metastasis (MMP2/9)Choi et al[75]
BazedoxifeneEstrogen receptor modulatorPostmenopausal osteoporosisJAK2/STAT3Reduce tumor burdenThilakasiri et al[76]; Wu et al[77]
FedratinibJAK2 inhibitorMyelofibrosisJAK2/STAT3Inhibit cell proliferationWang et al[41]; Passamonti et al[78]; Miyazaki et al[79]; Mao et al[80]
DisulfiramAcetaldehyde dehydrogenase inhibitorAlcohol abuseWnt, NF-κB, MAPKInhibit proliferation and induce apoptosisZhang et al[86]; Liu et al[87]
PropofolSedative-hypnoticAnesthesiaWnt, JAK2/STAT3Inhibit cell growth, invasion, and migrationZhan et al[88]; Chen et al[89]
TipifarnibFarnesyltransferase inhibitorNot approved (phase II for HNSCC)mTOR, HIF-1αInhibit proliferation and migrationEgawa et al[83]; Zhou et al[84]
SimvastatinStatinLower cholesterolWnt, HippoInhibit proliferation and metastasis and reduce radiotoxicity of healthy tissuesRao et al[35]; Liu et al[90]
BortezomibProteasome inhibitorMultiple myelomaNF-κBInhibit growth and induce apoptosisGuo et al[91]; Nakata et al[92]; Ocean et al[93]; Bui et al[94]
Propranololβ-AR receptor blockerCardiovascular diseases, postmenopausal osteoporosisNF-κBIncrease sensitivity to chemotherapy, cell cycle arrest, apoptosis, and inhibit tumor growth in xenograph modelsLiao et al[95]; Koh et al[96]
ResveratrolNatural productMetabolic and cardiovascular diseasesNF-κBDecrease cell viabilityTrautmann et al[97]; Rojo et al[98]
VerteporfinBenzoporphyrinPhotodinamic therapyHippoInhibit cell proliferationGiraud et al[101]
AbacavirNucleoside reverse transcriptase inhibitorHIV infectionTERTInsufficiently suppress cell proliferation and activate tumorigenic pathwaysPanneerpandian et al[102]
CandesartanAngiotensin II type I receptor blockerHypertension, heart failure, diabetic neuropathyTGF-β1Suppress tumor cell proliferation and stromal fibrosis in xenograph modelsAraújo et al[29]; Okazaki et al[103]
TelmisartanAngiotensin II type I receptor blockerHypertension, heart failure, diabetic neuropathyEGFRInhibit proliferationAraújo et al[29]; Fujita et al[104]
FluoxetineAntidepressantDepression in cancer patientsDeath receptor pathwayInduce apoptosisAraújo et al[29]; Po et al[105]
ItraconazoleAntifungalFungal infectionsHedgehogInhibit proliferation and increase apoptosisHu et al[106]
LevobupivacaineLocal anestheticAnesthesiaFerroptosisInhibit growth in vitro and in vivo and induce ferroptosisMao et al[107]
ParoxetineAntidepressant (SSRI)DepressionDNA repair proteinsInduce apoptosis and inhibit cell viabilityLiu et al[110]; Kowalska et al[111]
RisperidoneAntipsychoticSchizophreniaUnknown (proposed: Histone deacethylation, dopamine receptor and cholesterol homeostasis)Induce apoptosis and inhibit growthChen et al[112]
MAPK signaling pathway

MAPKs are a family of serine/threonine kinases that regulate different functions in GC, such as proliferation, migration, invasion, and metastasis. The signaling pathway is composed of five cascades: The Jun amino-terminal kinases cascade, the p38/MAPK cascade, and three extracellular signal-regulated kinase (ERK) cascades. One of the most relevant cascades is the ERK/MAPK cascade. ERK/MAPK controls matrix metalloproteinases (MMPs), which are proteins that degrade the extracellular matrix and play a role in cell migration and invasion. The signaling cascade starts with RAS, which interacts with RAF. This activates MEK1/2 kinases, which in turn activate ERK[8,30]. MAPK/ERK signaling is deregulated in approximately one-third of human cancers, and inhibitors of this pathway have been shown to be effective against GC[30,31] (Figure 1).

Figure 1
Figure 1 Mechanisms of action of repurposed drugs targeting the mitogen-activated protein kinase signaling pathway. Purple form indicated main pathway proteins; light blue form indicated drugs; dark blue form indicated proteins involved in the mechanism of action of the drug; “?” symbol indicated target is not clear. KLF: Krupple-like factor; DDIT4: DNA damage inducible transcript 4; MEK: Mitogen-activated protein; ERK: Extracellular signal-regulated kinase; MAPK: Mitogen-activated protein kinase.

Twenty-six antimicrobial agents were screened in a GC cell line to study their role as ERK/MAPK inhibitors, and among them, doxycycline had strong inhibitory effects. This drug is an antibiotic whose mechanism of action consists of binding with the 30S ribosomal subunit and interfering with translation. Doxycycline has already been shown to have anticancer effects on breast and prostate cancers. In GC, doxycycline inhibits proliferation, colony formation, and spheroid growth[31]. Moreover, the compound also inhibits several pathways expressed in a subset of GCs, such as the Wnt signaling pathway[30].

Lovastatin is a statin typically used to lower cholesterol levels that has shown anticancer effects in breast cancer, ovarian cancer, and multiple myeloma[32]. Lovastatin has been identified as a promising drug for treating GC because of its ability to inhibit the growth of GC cells[33]. In leukemia, lovastatin inhibited leukemogenesis through the induction of Krupple-like factor 5 and subsequent downregulation of DNA damage inducible transcript 4 (DDIT4), which inhibited the MAPK pathway[32]. Given that DDIT4 has been shown to promote GC tumorigenesis through the MAPK and p53 pathways[34], this pathway could also be a potential target of lovastatin in GC. In addition, a recent study demonstrated that lovastatin increased the efficacy of anti-HER2 therapy in HER2 + GC patients when it was combined with trastuzumab. Lovastatin acts by increasing the availability of membrane-bound HER2 while simultaneously having radioprotective effects in normal tissues[35].

Silymarin, a product obtained from the milk thistle plant (Silybum marianum), has been used for the treatment of epilepsy and for hepatoprotection in patients with cirrhosis or alcoholic hepatitis, among other conditions[36,37]. In both in vitro and in vivo studies, silymarin inhibited growth and proliferation in both GC cells and tumor xenograft mouse models by downregulating the MAPK pathway[37].

Tegaserod is a 5-hydroxytryptamine receptor 4 (5-HTR4) agonist that is typically employed to treat irritable bowel syndrome[38]. In GC, the drug acts by inhibiting MEK1/2 kinases, whose high levels are correlated with poor patient prognosis. Their inhibition decreases ERK1/2 signaling and therefore inhibits GC growth in vitro and in vivo[39].

The MEK inhibitor trametinib is already used in patients with melanoma and has been demonstrated to be effective in GC mouse organoid models with KRAS mutations[40,41]. KRAS mutations are present in many human cancers and result in lower overall survival in patients with GC. They activate signaling pathways such as the PI3K and MAPK pathways. In a recent study, the use of trametinib in mouse organoid models reduced the number of phosphorylated ERK proteins and suppressed tumor development as well as liver metastasis[40]. However, in another study, trametinib alone was not enough to suppress the proliferation of GC cells, but combination therapy with the JAK2/STAT3 inhibitor fedratinib reduced cell survival and induced apoptosis[41].

PI3K/AKT/mTOR signaling pathway

The PI3K/AKT pathway is relevant in cancer and regulates the inhibition of apoptosis, the induction of drug resistance, metastasis, and angiogenesis[8,42]. Downstream signaling of PI3K results in the recruitment of the serine-threonine kinase AKT, which has mTOR as a downstream effector. Effective inhibitors could lead to better outcomes for cancer patients, and some have already been approved for clinical use[42] (Figure 2).

Figure 2
Figure 2 Mechanisms of action of repurposed drugs targeting the phosphoinositide 3-kinase, p53, and hypoxia-inducible factor-1α signaling pathways. Purple form indicated main pathway proteins; light blue form indicated drugs; dark blue form indicated proteins involved in the mechanism of action of the drug; “?” symbol indicated target is not clear. ATRA: All-trans retinoic acid; Pin1: Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1; AMPK: Adenosine 5’-monophosphate-activated protein kinase; SRC: Proto-oncogene tyrosine-protein kinase Src; DRD2: Dopamine receptor D2; HDAC1/2: Histone deacetylase 1/2; PI3K: Phosphoinositide 3-kinase; AKT: Protein kinase B; mTOR: Mammalian target of rapamycin; SMHT2: Serine hydroxymethyl transferase 2.

Retinoic acids are compounds derived from vitamin A and are ligands for retinoic acid receptors and retinoic X receptors. Both receptors form a heterodimer and act as transcription factors for proteins that play a role in cell differentiation[43]. All-trans retinoic acid (ATRA) is used to treat promyelocytic leukemia and has been previously reported to exert antitumor effects in other cancers, such as breast and liver cancer, through the inhibition of peptidyl-prolyl cis-trans isomerase NIMA-interacting 1. This protein is also confirmed to be a target in GC cells, suppressing proliferation in vivo and in vitro. The protein is involved in the PI3K and Wnt signaling pathways and in crosstalk between these pathways, both of which are highly relevant in GC[44]. In addition, ATRA affects gastric dysplasia. The addition of omeprazole and sucralfate to conventional treatment for gastritis has been shown to improve the prognosis of patients with gastric dysplasia by increasing retinoblastoma levels and reducing HER2 levels[45]. ATRA also has two isomers, 13-cis-retinoic acid and 9-cis-retinoic acid, which have anticancer effects on GC. However, these isomers are too toxic for use in GC patients[43].

Aurora kinases are important regulators of mitosis, and their overexpression promotes the development of GC. As such, they have been proposed as promising targets[46]. Danusertib and alisertib are aurora kinase inhibitors; danusertib is a pan-inhibitor and alisertib specifically inhibits aurora kinase A. Studies in GC cell lines have shown that both compounds inhibit cell proliferation, induce cell cycle arrest, and promote apoptosis and autophagy. These compounds exert their effects through adenosine 5’-monophosphate-activated protein kinase (AMPK) and p38-MAPK, which are upstream regulators of AKT/mTOR[46,47]. Although not approved, danusertib and alisertib have passed phase II clinical trials, and alisertib has a single completed phase III trial in peripheral T-cell lymphoma. A trial of danusertib as a single agent in hormone-refractory prostate cancer patients showed no objective response, but alisertib has shown some efficacy in combination therapy for solid and hematological tumors[48].

Deferasirox is an iron chelator used for the treatment of iron overload disease. Iron chelators are being explored as anticancer agents because iron metabolism is implicated in tumor initiation and growth. Despite adverse events such as gastrointestinal disturbance or renal toxicity, deferasirox is generally well tolerated and has been shown to have antineoplastic effects on leukemias through downregulation of the NF-κB and mTOR pathways. In GC, deferasirox induces apoptosis and exerts its antitumor effects through several pathways, which include the inhibition of cell cycle progression and effects on the mTOR and metastasis pathways[49].

Dronedarone hydrochloride is an antiarrhythmic drug that targets the proto-oncogene tyrosine-protein kinase Src (SRC). This kinase regulates processes such as proliferation, migration, and invasion and plays a role in tumorigenesis by phosphorylating AKT1. In GC, cell proliferation and colony formation were suppressed in cells treated with dronedarone. The mechanism of action consists of the drug binding to and inhibiting SRC activity, thereby downregulating the AKT signaling pathway[50].

Metformin is used for the treatment of type 2 diabetes and is associated with a decreased risk of cancer. Metformin acts by suppressing respiratory complex I, which results in the activation of AMPK signaling, reducing blood glucose levels and enhancing insulin sensitivity. Metformin has been shown to induce the apoptosis of GC cells in vitro through this mechanism, as treatment with metformin resulted in the upregulation of AMPK. In turn, this protein modulates the PI3K/AKT pathway[51]. Interestingly, metformin is not only a promising treatment for GC but also a promising preventive agent. Metformin was shown to reduce GC risk[52] and had therapeutic effects in an intestinal metaplasia mouse model[53].

Mycophenolic acid is an inhibitor of inosine monophosphate dehydrogenase, which reduces de novo purine synthesis, inhibiting T and B lymphocytes. Therefore, mycophenolic acid results in immunosuppression and, as such, is used for applications such as organ transplantation and lupus erythematosus[54,55]. In GC, mycophenolic acid has been shown to decrease AKT activity by reducing its phosphorylation, which reduced the capacity for proliferation of GC cells.

Naftopidil is a drug used for the treatment of benign prostate hyperplasia that acts by blocking the α-1 adrenoceptor. Naftopidil has been shown to reduce prostate cancer risk and induce death in these cancer cells. In GC cells, naftopidil induces apoptosis through the inhibition of the PI3K/AKT signaling pathway by reducing the phosphorylation levels of AKT, and treatment with naftopidil plus an autophagy inhibitor enhances its apoptotic effects[56]. In a previous study, a synthetic analogous compound of naftopidil was shown to have an even stronger effect for suppressing cell proliferation in GC cells[57]. However, the application of naftopidil is approved only in Japan, and clinical trials have been performed only in Asian populations, which present some metabolic differences compared with non-Asian populations. In addition, naftopidil has not been tested in women, so further information about its safety is needed[58].

Rapamycin is an mTOR inhibitor and can reduce the proliferation and invasion of GC cells. As HIF-1α is downstream of mTOR, rapamycin can also inhibit HIF-1α- and HIF-1-dependent transcription induced by hypoxia[59,60]. An analogous compound, 42-(2-tetrazolyl) rapamycin, was shown to inhibit Forkhead box protein P4, downregulating the Hippo pathway. This suppressed tumor growth and increased the efficacy of the chemotherapeutic drug 5-fluorouracil that is used in standard treatment[61].

Sertraline is an antidepressant of the serotonin reuptake inhibitor (SSRI) type, and the use of SSRIs has been shown to prolong survival and quality of life in cancer patients with depression. Sertraline has been shown to resensitize drug-resistant GC through the induction of apoptosis via the PI3K pathway. The effects of normal sertraline are limited; thus, modified sertraline derivatives have been used with better results[62]. Sertraline has also been shown to have an effect on non-small cell lung cancer, in which the target is serine hydroxymethyl transferase 2[63]. Given that this target has been shown to upregulate the PI3K signaling pathway[64] and is upregulated in GC and other cancers (in which it regulates other pathways, such as the HIF-1α pathway[65]), this is a plausible mechanism of action in GC.

Vortioxetine is another antidepressant that affects the serotonin pathway by inhibiting 5-HTR3A[38]. A recent study revealed that vortioxetine could inhibit cell proliferation, migration, and invasion in a GC cell line. Vortioxetine induced apoptosis and autophagy in cells via the PI3K pathway by decreasing the phosphorylation levels of AKT and mTOR[66]. Another study revealed that vortioxetine exerts anticancer activity by directly binding to JAK2 and SRC, which inhibits them and thus inhibits the JAK2/STAT3 signaling pathway[67].

Thioridazine is an antipsychotic drug with reported effects on different cancers, including leukemia and breast cancer. In GC cells, thioridazine has been shown to reduce cell viability and induce cell apoptosis via the inhibition of dopamine receptor D2 (DRD2), a receptor that controls AKT and Wnt. Therefore, the observed effects could be related to these pathways[68]. A posterior analysis of 84 GC samples revealed the relevance of DRD2 in GC; higher levels of this receptor were found in GC and patients with higher levels of DRD2 had a worse prognosis[69]. Another interesting point in favor of repurposing thioridazine against GC was demonstrated in a recent study in which the therapeutic effect of anti-HER2 chemotherapy was enhanced when this drug was combined with thioridazine. This was mediated through the inhibition of S-phase kinase associated protein 2 (Skp2), a component of the Skp2-stem cell factor complex. This complex plays a role in ubiquitination, cell cycle progression, proliferation, and apoptosis[70].

Tigecycline is a glycilcycline-type antibiotic that is used to treat complicated infections and has shown important anticancer effects on solid and liquid tumors[71]. An in vitro study revealed that tigecycline induced autophagy and inhibited cell proliferation in GC cells through the upregulation of AMPK, which led to the inhibition of the PI3K signaling pathway[72].

Valproic acid is a histone deacetylase inhibitor that is used to treat bipolar disorder and epilepsy[73]. High expression of the drug’s target, histone deacetylase 1/2, has been linked to poor survival in patients with GC. Through this target, valproic acid downregulates the activity of the PI3K signaling pathway by inhibiting AKT, which results in apoptosis in GC cells. The main disadvantage is that the therapeutic concentrations reached in in vitro experiments are not achievable in the clinic; thus, other strategies such as a combination of valproic acid with chemotherapy should be used[74]. Another study reported similar results in different GC cell lines[33]. However, the drug has already been tested in a phase II clinical trial to determine whether it offered survival advantages in advanced GC when combined with paclitaxel, compared with paclitaxel alone, and negative results were obtained[73].

p53 signaling pathway

The p53 signaling pathway is involved in the regulation of DNA repair and control of the cell cycle, apoptosis, and differentiation. This pathway can block the cell cycle in vertebrates by controlling checkpoints in the G1/S and G2/M phases; thus, its function is related to cyclins and cyclin-dependent kinases. More than 75% of GC patients have elevated levels of p53. H. pylori infection can induce mutations in the TP53 gene, so inhibition of this pathway may be a therapeutic strategy for this stage[8] (Figure 2).

Econazole is an antifungal drug and a calcium ion channel agonist that is used for the treatment of skin infections. According to a recent study, this drug activates p53 and induces caspase-dependent apoptosis in GC cells. Moreover, econazole affects different signaling pathways, producing decreased levels of phosphorylated PI3K, AKT, and ERK. Downregulation of MMP2 and MMP9 has also been shown, which inhibits the metastatic capacity of the cells[75].

JAK2/STAT3

STAT3 is an oncogene that is overexpressed in many cancers, including GC. The JAK2/STAT3 signaling pathway is triggered by the binding of an interleukin (IL) or an elongation factor G family member to transmembrane cytokine receptors. After that, the JAK2 receptor is dimerized and transphosphorylated, which activates STAT3. STAT3 is subsequently translocated to the nucleus, where it acts as a transcription factor that regulates (among other genes) genes related to cancer cell growth, tumor invasion, and chemoresistance. In GC, STAT3 signaling has been linked to tumor progression and metastasis[8] (Figure 3).

Figure 3
Figure 3 Mechanisms of action of repurposed drugs targeting the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway. Purple form indicated main pathway proteins; light blue form indicated drugs; dark blue form indicated proteins involved in the mechanism of action of the drug; IL11: Interleukin 11; JAK2: Janus kinase 2; STAT3: Signal transducer and activator of transcription 3.

Bazedoxifene is a selective estrogen receptor modulator used for the treatment and prevention of postmenopausal osteoporosis. This drug has demonstrated antitumor effects in pancreatic cancer and other gastrointestinal cancers[76,77]. In mouse models of gastrointestinal cancers, bazedoxifene reduced the tumor burden and interfered with the binding of IL-11 to the receptor β subunit gp130, which inhibited JAK2/STAT3. In addition, as the mechanism of action is independent of the modulation of the estrogen receptor, bazedoxifene has been shown to have the same tumor burden-reducing effect in males and females[76].

Fedratinib is a JAK2-selective inhibitor that is approved for the treatment of myelofibrosis[78]. When applied in combination with other drugs, it has anticancer effects on pancreatic cancer and colorectal cancer[79,80]. In fact, high doses of the drug have been reported to have serious adverse effects; thus, its combination with other drugs could enable the use of a lower dose[79]. In a recent study, fedratinib was tested alone and in combination with the MEK inhibitor trametinib. Although fedratinib had a stronger inhibitory effect on GC than trametinib when both were applied alone, their combination had an even stronger effect, and the combination had similar effects on pancreatic cancer[41].

HIF-1α

The HIF family genes include HIF-1, HIF-2, and HIF-3. HIF-1 is composed of two subunits, HIF-1α and HIF-1β. Under normoxic conditions, HIF-1α is degraded, but under hypoxic conditions, HIF-1α enters the nucleus and combines with HIF-1β to promote gene transcription. These genes play important roles in cancer, especially HIF-1α, which inhibits apoptosis and promotes processes such as proliferation, metastasis, drug resistance, and angiogenesis in GC[81]. In fact, HIF-1α not only contributes to tumor progression but also to the carcinogenetic process, as HIF-1α expression increases with progression through Correa’s cascade. The frequency of HIF-1α positivity is greater in diffuse-type GC than in intestinal-type GC; thus, HIF-1α likely plays an even more important role in this type of cancer. This pathway also interacts with others, and the upregulation of MAPK and PI3K potentiate HIF-1α activity, which indicates that the inhibition of these pathways could also inhibit HIF-1α[82] (Figure 2).

A drug that targets this pathway, tipifarnib, is a farnesyltransferase inhibitor. This drug was granted fast track designation by the United States Food and Drug Administration for the treatment of head and neck squamous cell carcinoma in 2019 after positive results were obtained in a phase II clinical trial, and tipifarnib has already demonstrated its efficacy in triple-negative breast cancer[83,84]. Low doses have been tested in GC cell lines, and tipifarnib inhibited the proliferation and migration of HIF-1α-positive cells but had no effect on a HIF-1α-negative cell line. The mechanism of action involves the inhibition of Rheb farnesylation, which suppresses the mTOR/HIF-1α signaling pathway[83].

Wnt/β-catenin signaling pathway

The Wnt signaling pathways include the classical Wnt/β-catenin pathway, which is involved in cell proliferation, and the nonclassical Wnt/planar cell polarity and Wnt/calcium ion pathways, which are involved in cell polarity and migration. β-catenin is a transcriptional coactivator involved in proliferation, apoptosis, and infiltration in tumor cells. Dysregulation of the Wnt/β-catenin signaling pathway is present in more than half of GC patients and is related to treatment resistance and epithelial–mesenchymal transition[8,85] (Figure 4).

Figure 4
Figure 4 Mechanisms of action of repurposed drugs targeting the Wnt, nuclear factor kappa B, and Hippo signaling pathways. Purple form indicated main pathway proteins; light blue form indicated drugs; dark blue form indicated proteins involved in the mechanism of action of the drug; “?” symbol indicated target is not clear. β-AR: Beta-adrenergic receptor; NF-κB: Nuclear factor kappa B; DKK1: Dickkopf-related protein 1; YAP: Yes-associated protein; TAZ: Transcriptional coactivator with PDZ-binding motif; HMGCR: Hydroxymethyl glutaryl-CoA.

Disulfiram is a drug that is used to treat alcohol abuse through the inhibition of acetaldehyde dehydrogenase. Disulfiram has been shown to have anticancer effects and to reverse drug resistance, inhibit DNA methylation, and induce apoptosis. In a recent study, disulfiram inhibited proliferation in two GC cell lines and downregulated the Wnt and NF-κB signaling pathways[86]. Another possible mechanism of action depends on the presence of cupric ions. Disulfiram is catalyzed into diethyldithiocarbamate, which chelates into complexes that contain cupric ion. These factors enhance the antitumor properties of these compounds by increasing the level of cellular reactive oxygen species, which leads to apoptosis through the MAPK signaling pathway[86,87].

Propofol is a sedative-hypnotic drug that is used for anesthesia, and its use in the surgical resection of tumors has been shown to reduce the likelihood of recurrence. Propofol has been reported to inhibit GC cell growth, invasion, and migration through increased expression of miR-493-3p. This microRNA (miRNA) targets Dickkopf-related protein 1 (DKK1), an activator of the Wnt signaling pathway, whose high expression has been linked to poor survival in GC patients. Increased expression of this miRNA decreases the expression of DKK1 and inhibits the Wnt signaling pathway[88]. Propofol has also been shown to upregulate the expression of other miRNAs, such as miR-125b-5p, which inhibits the JAK2/STAT3 signaling pathway[89].

Like other statins, simvastatin is used to decrease blood cholesterol levels by inhibiting hydroxymethylglutaryl-CoA reductase. This target protein is upregulated in GC and is related to cancer cell growth and migration. In one study, the mechanism of action was proposed to involve targeting hydroxymethylglutaryl-CoA, which downregulates geranylgeranyl pyrophosphate. This is necessary for the posttranslational modification of RhoA, so this process reduces active RhoA levels and decreases the levels of β–catenin and YAP. In addition, a positive feedback loop was found to exist between YAP and Wnt, despite previous findings that Hippo inhibits Wnt. Therefore, simvastatin was found to inhibit proliferation and metastasis via this mechanism[90]. Another point in favor of repurposing this drug for GC treatment is its ability to reduce radiotoxicity in normal tissues (similar to lovastatin and other statins)[35].

NF-κB

NF-κB is a family of transcription factors comprising RelA, V-Rel reticuloendotheliosis viral oncogene homolog B, c-Rel, NF-κB1, and NF-κB2. NF-κB can be activated by tumor necrosis factor receptors, toll-like receptors, and the IL-1 receptor. Under normal conditions, NF-κB remains in the cytoplasm in an inactive state and is bound to a family of proteins called inhibitor of NF-κB (IκB). However, when the IκB kinase complex is stimulated, the transcription factor is liberated and translocated to the nucleus where it promotes the transcription of different target genes, and thus the progression of GC[8]. In cancer, NF-κB is aberrantly activated, which leads to tumor proliferation, the inhibition of apoptosis, and the facilitation of metastasis[91] (Figure 4).

Bortezomib is a selective and reversible proteasome inhibitor that inhibits the activity of the 26S proteasome, which functions in protein degradation[91]. This drug is approved for treatment of multiple myeloma, and a previous study showed that bortezomib inhibited growth and induced apoptosis in seven GC cell lines and in vivo xenograft models. Its effect occurred through the inhibition of NF-κB, and it had a greater effect on cell lines with lower levels of NF-κB[92]. In contrast, in a clinical trial of bortezomib alone and in combination with irinotecan in advanced gastroesophageal junction and gastric adenocarcinoma, bortezomib alone had limited effects and did not enhance the effectiveness of irinotecan[93]. However, a recent study revealed that it could overcome tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resistance. TRAIL is a compound that causes apoptosis in many cancer cell types, including GC, but resistance to treatment with TRAIL has been observed. The application of bortezomib caused cell cycle arrest and apoptosis, and these effects were thought to be mediated by the upregulation of dopamine receptor 5[94].

Propranolol is a nonselective β-adrenergic receptor (β-AR) blocker that is used to treat cardiovascular diseases and other conditions such as post-menopausal osteoporosis. In addition, propranolol has been shown to have anticancer effects for neuroblastoma and GC. β-AR inhibition has been shown to increase the sensitivity of cancer cells to radiotherapy through NF-κB inhibition. Moreover, propranolol was shown to reduce the side effects of radiation on surrounding tissues[95]. Another study revealed that propranolol caused cell cycle arrest and apoptosis in GC cell lines and also affected metastasis by suppressing the expression of MMP2, MMP9, and vascular endothelial growth factor A. These effects were consistent with those in xenograft mouse models, in which tumor growth was inhibited[96].

Resveratrol is a natural polyphenol that is present in foods such as nuts, apples, and black olives. This polyphenol can interact with reactive oxygen species and cytoplasmic and nuclear proteins in cells, thus conferring its antioxidant, anti-inflammatory, and antimicrobial properties. These characteristics have made resveratrol an alternative and complementary therapy for metabolic and cardiovascular diseases, and it has also been shown to be useful for treating GC[97]. The antitumor effects of resveratrol are exerted partly through inhibition of the NF-κB signaling pathway. In a recent study, resveratrol was tested in two GC cell lines and was shown to decrease cell viability in a dose-dependent manner; cytotoxic effects occurred at higher concentrations and inhibition of the invasion potential of cells occurred at noncytotoxic concentrations. The proposed mechanism of action was the inhibition of heparanase as a result of the downregulation of NF-κB signaling. Heparanase plays an important role in the metastatic process of GC[98]. In addition, resveratrol has synergistic effects with some currently used chemotherapeutic agents such as 5-fluorouracil[97].

Hippo pathway

The Hippo signaling cascade starts with mammalian sterile20-like kinases 1/2 phosphorylating large tumor suppressor 1/2 kinases. These phosphorylate the transcription coactivators YAP/transcriptional coactivator with PDZ-binding motif (TAZ), which are taken to the cytoplasm and degraded. However, when no Hippo signaling occurs, these proteins translocate to the nucleus and form a complex with the TEAD family. This promotes the expression of downstream target genes and, as a result, increases proliferation and decreases apoptosis. Current therapies focus on either suppressing YAP nuclear translocation by enhancing the Hippo pathway or disrupting the nuclear YAP/TAZ-TEAD complex[99] (Figure 4).

Therefore, the Hippo signaling pathway is considered a tumor-suppressor pathway that inhibits YAP/TAZ[19]. This pathway appears to be downregulated in GC, which indicates that YAP/TAZ and TEAD are upregulated; high YAP1 expression is associated with poor outcomes, promoting growth and metastasis[100]. In addition, YAP1 is overactivated in dysplasia and is involved in the processes stimulated by H. pylori that lead to carcinogenesis[19].

Verteporfin is a benzoporphyrin that is used in ophthalmology for photodynamic therapy, and its anticancer effects have been tested in breast, pancreatic, and GCs. In a recent study, YAP1 and TAZ1 were shown to be overexpressed in chemoresistant stem cells, and the effects of verteporfin were tested in GC cell lines. Verteporfin targets YAP1/TAZ and decreases the transcriptional activity of YAP/TAZ-TEAD and cell proliferation[101].

Others

Other important pathways and proteins can be targeted for GC treatment, such as reverse transcriptase, transforming growth factor-beta (TGF-β), epidermal growth factor receptor (EGFR), autophagocytosis, Hedgehog, ferroptosis, and even DNA repair protein expression (Figure 5).

Figure 5
Figure 5 Mechanisms of action of repurposed drugs with other target proteins/pathways. Purple form indicated main pathway proteins; light blue form indicated drugs; dark blue form indicated proteins involved in the mechanism of action of the drug; “?” symbol indicated target is not clear. ERK: Extracellular signal-regulated kinase; HIF-1α: Hypoxia-inducible factor-1α; NF-κB: Nuclear factor kappa B; TERT: Telomerase reverse transcriptase; TGF-β1: Transforming growth factor-β1; EGFR: Epidermal growth factor receptor; DR: Dopamine receptor.

Abacavir is a nucleoside reverse transcriptase inhibitor that is used to treat human immunodeficiency virus infection, and as such, it causes telomere shortening, chromosomal instability, and cell death. Telomerase reverse transcriptase is overexpressed in tumor tissues and thus represents an interesting target for anticancer therapies. However, a recent study revealed that abacavir did not have enough potential to suppress GC cells, even at high concentrations, and it activated several tumorigenic pathways, including the Wnt, ERK, HIF-1α, and NF-κB pathways[102].

Candesartan is an angiotensin II type I receptor blocker that is used to treat hypertension, heart failure, and diabetic nephropathy[29,103]. Angiotensin II has been linked to cancer progression, and the mechanism of action of its inhibitors consists of antagonizing the angiotensin II type 1 receptor[104]. Candesartan was tested in a GC cell line and in mouse xenograft models and was found to inhibit TGF-β1. In addition, candesartan suppressed tumor cell proliferation and stromal fibrosis in xenograft models[103]. The effects of TGF-β1 can differ depending on the cell type and physiological environment. In GC, TGF-β1 has been shown to inhibit proliferation in some cell lines, but other studies have shown that it can promote tumor growth. In addition, high expression of TGF-β1 in patients with GC has been linked to poor 5-year survival[8].

Another angiotensin receptor blocker is telmisartan, which is used for the same purposes as candesartan[29]. Telmisartan has been shown to be effective against a variety of cancers, including T-cell leukemia, esophageal adenocarcinoma, and hepatocellular carcinoma. This drug was tested in GC cell lines, and it inhibited proliferation through cell cycle arrest in the G0/G1 phase. The proposed mechanism of action is the inactivation of EGFR, which is observed at 24 hours after treatment but not at 48 hours, and the changes in the levels of some miRNAs[104]. EGFR, which is also known as HER1 and is overexpressed in GC, is part of the EGFR family, whose members participate in the regulation of tumor cell growth, proliferation, and migration[8].

The antidepressant fluoxetine is typically used to treat cancer-related depression. Fluoxetine has also been shown to have anticancer activity[105] and may be a promising candidate for drug repurposing[29]. According to a recent study, fluoxetine induces apoptosis through the death receptor pathway. However, fluoxetine also increases autophagocytosis, which can reduce apoptosis. The combination of fluoxetine and an autophagy inhibitor, 3-methyladenine, increased the apoptotic effect of fluoxetine. Considering that the mechanism of action of 3-methyladenine involves a reduction in AKT levels[105], the combination of fluoxetine with a PI3K inhibitor may be beneficial in GC treatment.

Itraconazole is an antifungal drug that inhibits endothelial cell proliferation and angiogenesis. Hedgehog signaling has been revealed to be overactivated in GC tissues, as shown by overexpression of the marker zinc finger protein Gli1 in GC tissues compared with adjacent normal tissues. Itraconazole inhibited proliferation and increased apoptosis in GC cells, and when it was used in combination with the chemotherapeutic agent 5-fluorouracil, the antiproliferative effects increased. The mechanism of action involves the inhibition of Gli1 and, as a result, the Hedgehog signaling pathway[106]. The Hedgehog pathway plays important roles in embryonic tissue development and in maintaining tissue homeostasis. Aberrant activation of this pathway is associated with carcinogenesis in medulloblastoma, non-small cell lung cancer, breast cancer, and GC[8,106].

Levobupivacaine is a local anesthetic that has been demonstrated to be effective against several cancers including GC and breast cancer. In one study, levobupivacaine inhibited GC cell growth in vivo and in vitro through the induction of ferroptosis. Levobupivacaine upregulated the expression of miR-489-3p, which targets the cystine/glutamate transporter gene SLC7A11 and subsequently induces ferroptosis[107]. Ferroptosis is a type of cell death induced by iron-dependent lipid peroxidation that has been linked to the development of GC, possibly through changes in iron metabolism related to H. pylori infection[108]. However, the induction of ferroptosis is also a feasible strategy for treating different types of cancers. Several compounds are being tested[109].

Paroxetine is another SSRI-type antidepressant that has been shown to have anticancer effects through various processes including the induction of cell death or the arrest of cell proliferation. The mechanism of action in GC involves inhibition of the expression of DNA repair proteins, which leads to DNA damage and the activation of apoptosis. Paroxetine was also shown to inhibit cell viability in AGS and MKN-45 cell lines but had a much stronger effect on AGS[110]. Moreover, in some types of cancers, paroxetine can also act as a JAK inhibitor[111].

Risperidone is an antipsychotic drug, and its effects on cancer are contradictory. Studies have reported that risperidone increases the risk of some cancers, and other studies have indicated a negative correlation with other types of cancer. Recently, risperidone was found to have anticancer effects on the GC cell line KATO-III, and it produced apoptosis and inhibited growth in xenograft tumor mouse models. In addition, a population-based study revealed a correlation between risperidone use and decreased GC risk. However, the mechanism of action of risperidone remains poorly understood; inhibition of histone deacetylation, modulation of the dopamine receptor pathway, and disruption of cholesterol homeostasis have been proposed as mechanisms[112].

CONCLUSION

GC is a worldwide challenge, and the discovery of new treatments may lead to more favorable outcomes for patients. In this context, drug repurposing has emerged as a promising strategy because of its lower costs and fewer challenges for drug implementation. In most cases, the development of GC starts with H. pylori-induced gastritis and then progresses through a series of steps leading to the development of gastric adenocarcinoma. The progression through these steps is controlled by several pathways, some of which are also involved in GC. Therefore, drugs targeting these pathways could be effective against GC and also against its precursor lesions. This could lead to new treatment approaches that target GC development from an early stage, reducing its global burden. However, the scarcity of research on premalignant lesions makes it difficult to pinpoint the exact stage at which a compound could be effective. Moreover, a lack of studies on most drugs in preneoplastic lesion models, such as patient-derived organoids or primary cell culture, means that there is still a long way before repurposed drugs can be implemented in the clinic for the prevention of GC development. Patient-derived organoid models could be especially advantageous for research on these types of lesions, as they allow the different stages of precancerous lesions and GC to be recreated. Among the candidate compounds for drug repurposing in GC, several target molecular pathways involved in processes such as cell proliferation, invasion, or migration, and have the potential to eliminate tumor cells or reduce their growth. Some relevant pathways in GC include the MAPK, PI3K/AKT/mTOR, p53, JAK2/STAT3, HIF-1α, Wnt/β-catenin, NF-κB, and Hippo pathways. Although the PI3K/AKT/mTOR pathway is the target of most repurposed drugs presented in this review, some drugs act on more than one pathway. A better understanding of drug mechanisms of action and their target pathways could enable the development of new synergistic combinations. This would enhance the efficacy of chemotherapy and potentially reduce the required dose for individual drugs, thereby minimizing side effects. Another area that could be potentially beneficial to explore in the future includes repurposed immunotherapeutic drugs. Research on this topic is lacking for GC, but it could represent a promising approach for future studies.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Spain

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade B, Grade B

Novelty: Grade B, Grade B, Grade B, Grade C

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

Scientific Significance: Grade B, Grade B, Grade B, Grade C

P-Reviewer: Xie Y, PhD, China; Zao XB, MD, Professor, China S-Editor: Fan M L-Editor: A P-Editor: Lei YY

References
1.  Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5690]  [Cited by in RCA: 9160]  [Article Influence: 9160.0]  [Reference Citation Analysis (3)]
2.  Lin JL, Lin JX, Lin GT, Huang CM, Zheng CH, Xie JW, Wang JB, Lu J, Chen QY, Li P. Global incidence and mortality trends of gastric cancer and predicted mortality of gastric cancer by 2035. BMC Public Health. 2024;24:1763.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 37]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
3.  Lordick F, Carneiro F, Cascinu S, Fleitas T, Haustermans K, Piessen G, Vogel A, Smyth EC; ESMO Guidelines Committee. Gastric cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33:1005-1020.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 196]  [Cited by in RCA: 727]  [Article Influence: 242.3]  [Reference Citation Analysis (0)]
4.  Marqués-Lespier JM, González-Pons M, Cruz-Correa M. Current Perspectives on Gastric Cancer. Gastroenterol Clin North Am. 2016;45:413-428.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 88]  [Cited by in RCA: 90]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
5.  Lordick F, Al-Batran SE, Arnold D, Borner M, Bruns CJ, Eisterer W, Faber G, Gockel I, Köberle D, Lorenzen S, Möhler M, Pritzkuleit R, Stahl M, Thuss-Patience P, Wöll E, Zander T, Maschmeyer G; Germany Society of Hematology and Medical Oncology (DGHO, Austrian Society of Hematology and Oncology (OeGHO), Swiss Society of Hematology (SSH), Swiss Society of Medical Oncology (SGMO);  in collaboration with the Arbeitsgemeinschaft Internistische Onkologie (AIO) of the German Cancer Society. German, Austrian, and Swiss guidelines for systemic treatment of gastric cancer. Gastric Cancer. 2024;27:6-18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 10]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
6.  Alagesan P, Goodwin JC, Garman KS, Epplein M. Cancer Progress and Priorities: Gastric Cancer. Cancer Epidemiol Biomarkers Prev. 2023;32:473-486.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
7.  Correa P, Piazuelo MB. The gastric precancerous cascade. J Dig Dis. 2012;13:2-9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 355]  [Cited by in RCA: 539]  [Article Influence: 41.5]  [Reference Citation Analysis (0)]
8.  Lei ZN, Teng QX, Tian Q, Chen W, Xie Y, Wu K, Zeng Q, Zeng L, Pan Y, Chen ZS, He Y. Signaling pathways and therapeutic interventions in gastric cancer. Signal Transduct Target Ther. 2022;7:358.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 179]  [Article Influence: 59.7]  [Reference Citation Analysis (1)]
9.  Al Khzem AH, Gomaa MS, Alturki MS, Tawfeeq N, Sarafroz M, Alonaizi SM, Al Faran A, Alrumaihi LA, Alansari FA, Alghamdi AA. Drug Repurposing for Cancer Treatment: A Comprehensive Review. Int J Mol Sci. 2024;25:12441.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
10.  Xia Y, Sun M, Huang H, Jin WL. Drug repurposing for cancer therapy. Signal Transduct Target Ther. 2024;9:92.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 153]  [Cited by in RCA: 134]  [Article Influence: 134.0]  [Reference Citation Analysis (0)]
11.  Tanoli Z, Fernández-Torras A, Özcan UO, Kushnir A, Nader KM, Gadiya Y, Fiorenza L, Ianevski A, Vähä-Koskela M, Miihkinen M, Seemab U, Leinonen H, Seashore-Ludlow B, Tampere M, Kalman A, Ballante F, Benfenati E, Saunders G, Potdar S, Gómez García I, García-Serna R, Talarico C, Beccari AR, Schaal W, Polo A, Costantini S, Cabri E, Jacobs M, Saarela J, Budillon A, Spjuth O, Östling P, Xhaard H, Quintana J, Mestres J, Gribbon P, Ussi AE, Lo DC, de Kort M, Wennerberg K, Fratelli M, Carreras-Puigvert J, Aittokallio T. Computational drug repurposing: approaches, evaluation of in silico resources and case studies. Nat Rev Drug Discov. 2025;24:521-542.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
12.  Hua Y, Dai X, Xu Y, Xing G, Liu H, Lu T, Chen Y, Zhang Y. Drug repositioning: Progress and challenges in drug discovery for various diseases. Eur J Med Chem. 2022;234:114239.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 29]  [Cited by in RCA: 119]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
13.  Businello G, Angerilli V, Parente P, Realdon S, Savarino E, Farinati F, Grillo F, Vanoli A, Galuppini F, Paccagnella S, Pennelli G, Mastracci L, Saragoni L, Fassan M. Molecular Landscapes of Gastric Pre-Neoplastic and Pre-Invasive Lesions. Int J Mol Sci. 2021;22:9950.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
14.  Gullo I, Grillo F, Mastracci L, Vanoli A, Carneiro F, Saragoni L, Limarzi F, Ferro J, Parente P, Fassan M. Precancerous lesions of the stomach, gastric cancer and hereditary gastric cancer syndromes. Pathologica. 2020;112:166-185.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 93]  [Cited by in RCA: 98]  [Article Influence: 19.6]  [Reference Citation Analysis (1)]
15.  Zhang S, Shen Y, Liu H, Zhu D, Fang J, Pan H, Liu W. Inflammatory microenvironment in gastric premalignant lesions: implication and application. Front Immunol. 2023;14:1297101.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
16.  Kuang W, Xu J, Xu F, Huang W, Majid M, Shi H, Yuan X, Ruan Y, Hu X. Current study of pathogenetic mechanisms and therapeutics of chronic atrophic gastritis: a comprehensive review. Front Cell Dev Biol. 2024;12:1513426.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
17.  Wang YM, Luo ZW, Shu YL, Zhou X, Wang LQ, Liang CH, Wu CQ, Li CP. Effects of Helicobacter pylori and Moluodan on the Wnt/β-catenin signaling pathway in mice with precancerous gastric cancer lesions. World J Gastrointest Oncol. 2024;16:979-990.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
18.  Sethi NS, Kikuchi O, Duronio GN, Stachler MD, McFarland JM, Ferrer-Luna R, Zhang Y, Bao C, Bronson R, Patil D, Sanchez-Vega F, Liu JB, Sicinska E, Lazaro JB, Ligon KL, Beroukhim R, Bass AJ. Early TP53 alterations engage environmental exposures to promote gastric premalignancy in an integrative mouse model. Nat Genet. 2020;52:219-230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 43]  [Cited by in RCA: 50]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
19.  Messina B, Lo Sardo F, Scalera S, Memeo L, Colarossi C, Mare M, Blandino G, Ciliberto G, Maugeri-Saccà M, Bon G. Hippo pathway dysregulation in gastric cancer: from Helicobacter pylori infection to tumor promotion and progression. Cell Death Dis. 2023;14:21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 47]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
20.  Ni Z, Min Y, Han C, Yuan T, Lu W, Ashktorab H, Smoot DT, Wu Q, Wu J, Zeng W, Shi Y. TGR5-HNF4α axis contributes to bile acid-induced gastric intestinal metaplasia markers expression. Cell Death Discov. 2020;6:56.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 26]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
21.  Jin D, Huang K, Xu M, Hua H, Ye F, Yan J, Zhang G, Wang Y. Deoxycholic acid induces gastric intestinal metaplasia by activating STAT3 signaling and disturbing gastric bile acids metabolism and microbiota. Gut Microbes. 2022;14:2120744.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 49]  [Cited by in RCA: 54]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
22.  Yu JH, Zheng JB, Qi J, Yang K, Wu YH, Wang K, Wang CB, Sun XJ. Bile acids promote gastric intestinal metaplasia by upregulating CDX2 and MUC2 expression via the FXR/NF-κB signalling pathway. Int J Oncol. 2019;54:879-892.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
23.  Riera KM, Jang B, Min J, Roland JT, Yang Q, Fesmire WT, Camilleri-Broet S, Ferri L, Kim WH, Choi E, Goldenring JR. Trop2 is upregulated in the transition to dysplasia in the metaplastic gastric mucosa. J Pathol. 2020;251:336-347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 31]  [Cited by in RCA: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
24.  Han L, Ma Y, Wu W, Ni Q, Xie J, Huang Y, Jin Q, Wu L, Zhang Y. Research progress on the therapeutic effects of effective components of traditional Chinese medicine in the treatment of gastric cancer precursors through modulation of multiple signaling pathways. Front Oncol. 2025;15:1555274.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
25.  Kim H, Jang B, Zhang C, Caldwell B, Park DJ, Kong SH, Lee HJ, Yang HK, Goldenring JR, Choi E. Targeting Stem Cells and Dysplastic Features With Dual MEK/ERK and STAT3 Suppression in Gastric Carcinogenesis. Gastroenterology. 2024;166:117-131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 23]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
26.  Rivera F, Longo F, Martín Richard M, Richart P, Alsina M, Carmona A, Custodio AB, Fernández Montes A, Gallego J, Fleitas Kanonnikoff T. SEOM-GEMCAD-TTD clinical guideline for the diagnosis and treatment of gastric cancer (2023). Clin Transl Oncol. 2024;26:2826-2840.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
27.  Sexton RE, Al Hallak MN, Diab M, Azmi AS. Gastric cancer: a comprehensive review of current and future treatment strategies. Cancer Metastasis Rev. 2020;39:1179-1203.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 459]  [Cited by in RCA: 480]  [Article Influence: 96.0]  [Reference Citation Analysis (0)]
28.  Eom SS, Ryu KW, Han HS, Kong SH. A Comprehensive and Comparative Review of Global Gastric Cancer Treatment Guidelines: 2024 Update. J Gastric Cancer. 2025;25:153-176.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 13]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
29.  Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. Molecules. 2022;28:319.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
30.  Magnelli L, Schiavone N, Staderini F, Biagioni A, Papucci L. MAP Kinases Pathways in Gastric Cancer. Int J Mol Sci. 2020;21:2893.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 57]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
31.  Pandian J, Panneerpandian P, Devanandan HJ, Sekar BT, Balakrishnan K, Selvarasu K, Muthupandi K, Ganesan K. Identification of the targeted therapeutic potential of doxycycline for a subset of gastric cancer patients. Ann N Y Acad Sci. 2020;1467:94-111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
32.  Gao J, Hu J, Yu F, Wang C, Sheng D, Liu W, Hu A, Yu K, Xiao X, Kuang Y, Zacksenhaus E, Gajendran B, Ben-David Y. Lovastatin inhibits erythroleukemia progression through KLF2-mediated suppression of MAPK/ERK signaling. BMC Cancer. 2023;23:306.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
33.  Zhang L, Kang W, Lu X, Ma S, Dong L, Zou B. Weighted gene co-expression network analysis and connectivity map identifies lovastatin as a treatment option of gastric cancer by inhibiting HDAC2. Gene. 2019;681:15-25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 59]  [Cited by in RCA: 55]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
34.  Du F, Sun L, Chu Y, Li T, Lei C, Wang X, Jiang M, Min Y, Lu Y, Zhao X, Nie Y, Fan D. DDIT4 promotes gastric cancer proliferation and tumorigenesis through the p53 and MAPK pathways. Cancer Commun (Lond). 2018;38:45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 40]  [Cited by in RCA: 74]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
35.  Rao Y, Samuels Z, Carter LM, Monette S, Panikar SS, Pereira PMR, Lewis JS. Statins enhance the efficacy of HER2-targeting radioligand therapy in drug-resistant gastric cancers. Proc Natl Acad Sci U S A. 2023;120:e2220413120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 20]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
36.  Koltai T, Fliegel L. Role of Silymarin in Cancer Treatment: Facts, Hypotheses, and Questions. J Evid Based Integr Med. 2022;27:2515690X211068826.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 51]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
37.  Kim SH, Choo GS, Yoo ES, Woo JS, Han SH, Lee JH, Jung JY. Silymarin induces inhibition of growth and apoptosis through modulation of the MAPK signaling pathway in AGS human gastric cancer cells. Oncol Rep. 2019;42:1904-1914.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
38.  Chen L, Huang S, Wu X, He W, Song M. Serotonin signalling in cancer: Emerging mechanisms and therapeutic opportunities. Clin Transl Med. 2024;14:e1750.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 20]  [Reference Citation Analysis (0)]
39.  Wang Z, Chen Y, Li X, Zhang Y, Zhao X, Zhou H, Lu X, Zhao L, Yuan Q, Shi Y, Zhao J, Dong Z, Jiang Y, Liu K. Tegaserod Maleate Suppresses the Growth of Gastric Cancer In Vivo and In Vitro by Targeting MEK1/2. Cancers (Basel). 2022;14:3592.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
40.  Yamasaki J, Hirata Y, Otsuki Y, Suina K, Saito Y, Masuda K, Okazaki S, Ishimoto T, Saya H, Nagano O. MEK inhibition suppresses metastatic progression of KRAS-mutated gastric cancer. Cancer Sci. 2022;113:916-925.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
41.  Wang X, Dai C, Yin Y, Wu L, Jin W, Fu Y, Chen Z, Hao K, Lu B. Blocking the JAK2/STAT3 and ERK pathways suppresses the proliferation of gastrointestinal cancers by inducing apoptosis. J Zhejiang Univ Sci B. 2021;22:492-503.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
42.  He Y, Sun MM, Zhang GG, Yang J, Chen KS, Xu WW, Li B. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther. 2021;6:425.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 161]  [Cited by in RCA: 930]  [Article Influence: 232.5]  [Reference Citation Analysis (0)]
43.  Bouriez D, Giraud J, Gronnier C, Varon C. Efficiency of All-Trans Retinoic Acid on Gastric Cancer: A Narrative Literature Review. Int J Mol Sci. 2018;19:3388.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 34]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
44.  Zhang Z, Yu W, Zheng M, Liao X, Wang J, Yang D, Lu W, Wang L, Zhang S, Liu H, Zhou XZ, Lu KP. Pin1 inhibition potently suppresses gastric cancer growth and blocks PI3K/AKT and Wnt/β-catenin oncogenic pathways. Mol Carcinog. 2019;58:1450-1464.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
45.  Jin J, Li X, Xing L, Chang Y, Wu L, Jin Z, Su X, Bai Y, Zheng Y, Jiang Y, Zhao X, Lu L, Gao Q. Addition of all-trans-retinoic acid to omeprazole and sucralfate therapy improves the prognosis of gastric dysplasia. J Int Med Res. 2015;43:204-216.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
46.  Yuan CX, Zhou ZW, Yang YX, He ZX, Zhang X, Wang D, Yang T, Pan SY, Chen XW, Zhou SF. Danusertib, a potent pan-Aurora kinase and ABL kinase inhibitor, induces cell cycle arrest and programmed cell death and inhibits epithelial to mesenchymal transition involving the PI3K/Akt/mTOR-mediated signaling pathway in human gastric cancer AGS and NCI-N78 cells. Drug Des Devel Ther. 2015;9:1293-1318.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
47.  Yuan CX, Zhou ZW, Yang YX, He ZX, Zhang X, Wang D, Yang T, Wang NJ, Zhao RJ, Zhou SF. Inhibition of mitotic Aurora kinase A by alisertib induces apoptosis and autophagy of human gastric cancer AGS and NCI-N78 cells. Drug Des Devel Ther. 2015;9:487-508.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
48.  Novais P, Silva PMA, Amorim I, Bousbaa H. Second-Generation Antimitotics in Cancer Clinical Trials. Pharmaceutics. 2021;13:1011.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 37]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
49.  Choi JH, Kim JS, Won YW, Uhm J, Park BB, Lee YY. The potential of deferasirox as a novel therapeutic modality in gastric cancer. World J Surg Oncol. 2016;14:77.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
50.  Lu X, Zhang W, Yang X, Yan X, Hussain Z, Wu Q, Zhao J, Yuan B, Yao K, Dong Z, Liu K, Jiang Y. Dronedarone hydrochloride inhibits gastric cancer proliferation in vitro and in vivo by targeting SRC. Transl Oncol. 2024;50:102136.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
51.  Lu CC, Chiang JH, Tsai FJ, Hsu YM, Juan YN, Yang JS, Chiu HY. Metformin triggers the intrinsic apoptotic response in human AGS gastric adenocarcinoma cells by activating AMPK and suppressing mTOR/AKT signaling. Int J Oncol. 2019;54:1271-1281.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
52.  Lan WH, Lin TY, Yeh JA, Feng CL, Hsu JT, Lin HJ, Kuo CJ, Lai CH. Mechanism Underlying Metformin Action and Its Potential to Reduce Gastric Cancer Risk. Int J Mol Sci. 2022;23:14163.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
53.  Hu R, Xue X, Sun X, Mi Y, Wen H, Xi H, Li F, Zheng P, Liu S. Revealing the role of metformin in gastric intestinal metaplasia treatment. Front Pharmacol. 2024;15:1340309.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
54.  Hsieh CH, Huang YW, Tsai TF. Oral Conventional Synthetic Disease-Modifying Antirheumatic Drugs with Antineoplastic Potential: a Review. Dermatol Ther (Heidelb). 2022;12:835-860.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
55.  Dun B, Sharma A, Teng Y, Liu H, Purohit S, Xu H, Zeng L, She JX. Mycophenolic acid inhibits migration and invasion of gastric cancer cells via multiple molecular pathways. PLoS One. 2013;8:e81702.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
56.  Nakamura A, Matsunaga W, Gotoh A. Autophagy Induced by Naftopidil Inhibits Apoptosis of Human Gastric Cancer Cells. Anticancer Res. 2018;38:803-809.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
57.  Kaku Y, Tsuchiya A, Kanno T, Nakao S, Shimizu T, Tanaka A, Nishizaki T. The newly synthesized anticancer drug HUHS1015 is useful for treatment of human gastric cancer. Cancer Chemother Pharmacol. 2015;75:527-535.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
58.  Florent R, Poulain L, N'Diaye M. Drug Repositioning of the α(1)-Adrenergic Receptor Antagonist Naftopidil: A Potential New Anti-Cancer Drug? Int J Mol Sci. 2020;21:5339.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
59.  Chen M, Lu J, Wei W, Lv Y, Zhang X, Yao Y, Wang L, Ling T, Zou X. Effects of proton pump inhibitors on reversing multidrug resistance via downregulating V-ATPases/PI3K/Akt/mTOR/HIF-1α signaling pathway through TSC1/2 complex and Rheb in human gastric adenocarcinoma cells in vitro and in vivo. Onco Targets Ther. 2018;11:6705-6722.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 26]  [Cited by in RCA: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
60.  Morgos DT, Stefani C, Miricescu D, Greabu M, Stanciu S, Nica S, Stanescu-Spinu II, Balan DG, Balcangiu-Stroescu AE, Coculescu EC, Georgescu DE, Nica RI. Targeting PI3K/AKT/mTOR and MAPK Signaling Pathways in Gastric Cancer. Int J Mol Sci. 2024;25:1848.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 48]  [Cited by in RCA: 80]  [Article Influence: 80.0]  [Reference Citation Analysis (0)]
61.  Liu X, Chen B, Xie F, Wong KY, Cheung AHK, Zhang J, Wu Q, Fang C, Hu J, Wang S, Xu D, Chen J, Wang Y, Wong CC, Chen H, Wu WKK, Yu J, Chan MWY, Tsang CM, Lo KW, Tse GMK, To KF, Kang W. FOXP4 Is a Direct YAP1 Target That Promotes Gastric Cancer Stemness and Drives Metastasis. Cancer Res. 2024;84:3574-3588.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
62.  Mu C, Peng RK, Guo CL, Li A, Yang XM, Zeng R, Li YL, Gu J, Ouyang Q. Discovery of sertraline and its derivatives able to combat drug-resistant gastric cancer cell via inducing apoptosis. Bioorg Med Chem Lett. 2021;41:127997.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
63.  Sánchez-Castillo A, Heylen E, Hounjet J, Savelkouls KG, Lieuwes NG, Biemans R, Dubois LJ, Reynders K, Rouschop KM, Vaes RDW, De Keersmaecker K, Lambrecht M, Hendriks LEL, De Ruysscher DKM, Vooijs M, Kampen KR. Targeting serine/glycine metabolism improves radiotherapy response in non-small cell lung cancer. Br J Cancer. 2024;130:568-584.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
64.  Sun M, Zhao M, Li R, Zhang Y, Shi X, Ding C, Ma C, Lu J, Yue X. SHMT2 promotes papillary thyroid cancer metastasis through epigenetic activation of AKT signaling. Cell Death Dis. 2024;15:87.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 12]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
65.  Wang W, Wang M, Du T, Hou Z, You S, Zhang S, Ji M, Xue N, Chen X. SHMT2 Promotes Gastric Cancer Development through Regulation of HIF1α/VEGF/STAT3 Signaling. Int J Mol Sci. 2023;24:7150.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
66.  Lv GB, Wang TT, Zhu HL, Wang HK, Sun W, Zhao LF. Vortioxetine induces apoptosis and autophagy of gastric cancer AGS cells via the PI3K/AKT pathway. FEBS Open Bio. 2020;10:2157-2165.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 18]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
67.  Li M, Duan L, Wu W, Li W, Zhao L, Li A, Lu X, He X, Dong Z, Liu K, Jiang Y. Vortioxetine hydrobromide inhibits the growth of gastric cancer cells in vivo and in vitro by targeting JAK2 and SRC. Oncogenesis. 2023;12:24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
68.  Mu J, Xu H, Yang Y, Huang W, Xiao J, Li M, Tan Z, Ding Q, Zhang L, Lu J, Wu X, Liu Y. Thioridazine, an antipsychotic drug, elicits potent antitumor effects in gastric cancer. Oncol Rep. 2014;31:2107-2114.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36]  [Cited by in RCA: 57]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
69.  Mu J, Huang W, Tan Z, Li M, Zhang L, Ding Q, Wu X, Lu J, Liu Y, Dong Q, Xu H. Dopamine receptor D2 is correlated with gastric cancer prognosis. Oncol Lett. 2017;13:1223-1227.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
70.  Yang ZY, Zhao YW, Xue JR, Guo R, Zhao Z, Liu HD, Ren ZG, Shi M. Thioridazine reverses trastuzumab resistance in gastric cancer by inhibiting S-phase kinase associated protein 2-mediated aerobic glycolysis. World J Gastroenterol. 2023;29:5974-5987.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
71.  Dong Z, Abbas MN, Kausar S, Yang J, Li L, Tan L, Cui H. Biological Functions and Molecular Mechanisms of Antibiotic Tigecycline in the Treatment of Cancers. Int J Mol Sci. 2019;20:3577.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 35]  [Cited by in RCA: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
72.  Tang C, Yang L, Jiang X, Xu C, Wang M, Wang Q, Zhou Z, Xiang Z, Cui H. Antibiotic drug tigecycline inhibited cell proliferation and induced autophagy in gastric cancer cells. Biochem Biophys Res Commun. 2014;446:105-112.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 53]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
73.  Fushida S, Kinoshita J, Kaji M, Oyama K, Hirono Y, Tsukada T, Fujimura T, Ohta T. Paclitaxel plus valproic acid versus paclitaxel alone as second- or third-line therapy for advanced gastric cancer: a randomized Phase II trial. Drug Des Devel Ther. 2016;10:2353-2358.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
74.  Sun J, Piao J, Li N, Yang Y, Kim KY, Lin Z. Valproic acid targets HDAC1/2 and HDAC1/PTEN/Akt signalling to inhibit cell proliferation via the induction of autophagy in gastric cancer. FEBS J. 2020;287:2118-2133.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 65]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
75.  Choi EK, Park EJ, Phan TT, Kim HD, Hoe KL, Kim DU. Econazole Induces p53-Dependent Apoptosis and Decreases Metastasis Ability in Gastric Cancer Cells. Biomol Ther (Seoul). 2020;28:370-379.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
76.  Thilakasiri P, Huynh J, Poh AR, Tan CW, Nero TL, Tran K, Parslow AC, Afshar-Sterle S, Baloyan D, Hannan NJ, Buchert M, Scott AM, Griffin MD, Hollande F, Parker MW, Putoczki TL, Ernst M, Chand AL. Repurposing the selective estrogen receptor modulator bazedoxifene to suppress gastrointestinal cancer growth. EMBO Mol Med. 2019;11:e9539.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 20]  [Cited by in RCA: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
77.  Wu X, Cao Y, Xiao H, Li C, Lin J. Bazedoxifene as a Novel GP130 Inhibitor for Pancreatic Cancer Therapy. Mol Cancer Ther. 2016;15:2609-2619.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 72]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
78.  Passamonti F, Lou Y, Chevli M, Abraham P. Real-world outcomes with fedratinib therapy in patients who discontinued ruxolitinib for primary myelofibrosis. Future Oncol. 2024;20:1165-1174.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
79.  Miyazaki S, Kitazawa M, Nakamura S, Koyama M, Yamamoto Y, Hondo N, Kataoka M, Tanaka H, Takeoka M, Komatsu D, Soejima Y. Targeting KRAS-mutant pancreatic cancer through simultaneous inhibition of KRAS, MEK, and JAK2. Mol Oncol. 2025;19:377-390.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
80.  Mao Y, Wang W, Yang J, Zhou X, Lu Y, Gao J, Wang X, Wen L, Fu W, Tang F. Drug repurposing screening and mechanism analysis based on human colorectal cancer organoids. Protein Cell. 2024;15:285-304.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 61]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
81.  Li M, Li G, Yang X, Yin W, Lv G, Wang S. HIF in Gastric Cancer: Regulation and Therapeutic Target. Molecules. 2022;27:4893.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
82.  Ucaryilmaz Metin C, Ozcan G. The HIF-1α as a Potent Inducer of the Hallmarks in Gastric Cancer. Cancers (Basel). 2022;14:2711.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 16]  [Cited by in RCA: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
83.  Egawa N, Tanaka T, Matsufuji S, Yamada K, Ito K, Kitagawa H, Okuyama K, Kitajima Y, Noshiro H. Antitumor effects of low-dose tipifarnib on the mTOR signaling pathway and reactive oxygen species production in HIF-1α-expressing gastric cancer cells. FEBS Open Bio. 2021;11:1465-1475.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
84.  Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther. 2024;9:132.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 227]  [Cited by in RCA: 219]  [Article Influence: 219.0]  [Reference Citation Analysis (0)]
85.  Han R, Yang J, Zhu Y, Gan R. Wnt signaling in gastric cancer: current progress and future prospects. Front Oncol. 2024;14:1410513.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
86.  Zhang J, Pu K, Bai S, Peng Y, Li F, Ji R, Guo Q, Sun W, Wang Y. The anti-alcohol dependency drug disulfiram inhibits the viability and progression of gastric cancer cells by regulating the Wnt and NF-κB pathways. J Int Med Res. 2020;48:300060520925996.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
87.  Liu Y, Guan X, Wang M, Wang N, Chen Y, Li B, Xu Z, Fu F, Zheng Z, Du C. Disulfiram/Copper induces antitumor activity against gastric cancer via the ROS/MAPK and NPL4 pathways. Bioengineered. 2022;13:6579-6589.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
88.  Zhan K, Song X, Zhang Q, Yang J, Lu S. Propofol-Induced miR-493-3p Inhibits Growth and Invasion of Gastric Cancer through Suppression of DKK1-Mediated Wnt/β-Catenin Signaling Activation. Dis Markers. 2023;2023:7698706.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
89.  Chen M, Wang T, Tian D, Hai C, Qiu Z. Induction, growth, drug resistance, and metastasis: A comprehensive summary of the relationship between STAT3 and gastric cancer. Heliyon. 2024;10:e37263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
90.  Liu Q, Xia H, Zhou S, Tang Q, Zhou J, Ren M, Bi F. Simvastatin Inhibits the Malignant Behaviors of Gastric Cancer Cells by Simultaneously Suppressing YAP and β-Catenin Signaling. Onco Targets Ther. 2020;13:2057-2066.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
91.  Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther. 2024;9:53.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 655]  [Cited by in RCA: 649]  [Article Influence: 649.0]  [Reference Citation Analysis (0)]
92.  Nakata W, Hayakawa Y, Nakagawa H, Sakamoto K, Kinoshita H, Takahashi R, Hirata Y, Maeda S, Koike K. Anti-tumor activity of the proteasome inhibitor bortezomib in gastric cancer. Int J Oncol. 2011;39:1529-1536.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
93.  Ocean AJ, Christos P, Sparano JA, Shah MA, Yantiss RK, Cheng J, Lin J, Papetti M, Matulich D, Schnoll-Sussman F, Besanceney-Webler C, Xiang J, Ward M, Dilts KT, Keresztes R, Holloway S, Chen EX, Wright JJ, Lane ME. Phase II trial of bortezomib alone or in combination with irinotecan in patients with adenocarcinoma of the gastroesophageal junction or stomach. Invest New Drugs. 2014;32:542-548.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
94.  Bui HTT, Le NH, Le QA, Kim SE, Lee S, Kang D. Synergistic apoptosis of human gastric cancer cells by bortezomib and TRAIL. Int J Med Sci. 2019;16:1412-1423.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 18]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
95.  Liao X, Chaudhary P, Qiu G, Che X, Fan L. The role of propranolol as a radiosensitizer in gastric cancer treatment. Drug Des Devel Ther. 2018;12:639-645.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
96.  Koh M, Takahashi T, Kurokawa Y, Kobayashi T, Saito T, Ishida T, Serada S, Fujimoto M, Naka T, Wada N, Yamashita K, Tanaka K, Miyazaki Y, Makino T, Nakajima K, Yamasaki M, Eguchi H, Doki Y. Propranolol suppresses gastric cancer cell growth by regulating proliferation and apoptosis. Gastric Cancer. 2021;24:1037-1049.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
97.  Trautmann D, Suazo F, Torres K, Simón L. Antitumor Effects of Resveratrol Opposing Mechanisms of Helicobacter pylori in Gastric Cancer. Nutrients. 2024;16:2141.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
98.  Rojo D, Madrid A, Martín SS, Párraga M, Silva Pinhal MA, Villena J, Valenzuela-Valderrama M. Resveratrol Decreases the Invasion Potential of Gastric Cancer Cells. Molecules. 2022;27:3047.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 20]  [Reference Citation Analysis (0)]
99.  Cao Z, Hou Y, Zhao Z, Zhang H, Tian L, Zhang Y, Dong C, Guo F, Tan L, Han Y, Wang W, Jiao S, Tang Y, An L, Zhou Z. Reactivating Hippo by drug compounds to suppress gastric cancer and enhance chemotherapy sensitivity. J Biol Chem. 2024;300:107311.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
100.  Mohajan S, Jaiswal PK, Vatanmakarian M, Yousefi H, Sankaralingam S, Alahari SK, Koul S, Koul HK. Hippo pathway: Regulation, deregulation and potential therapeutic targets in cancer. Cancer Lett. 2021;507:112-123.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 77]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
101.  Giraud J, Molina-Castro S, Seeneevassen L, Sifré E, Izotte J, Tiffon C, Staedel C, Boeuf H, Fernandez S, Barthelemy P, Megraud F, Lehours P, Dubus P, Varon C. Verteporfin targeting YAP1/TAZ-TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Int J Cancer. 2020;146:2255-2267.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 102]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
102.  Panneerpandian P, Devanandan HJ, Marimuthu A, Karthikeyan C, Ganesan K. Abacavir induces the transcriptional activity of YY1 and other oncogenic transcription factors in gastric cancer cells. Antiviral Res. 2020;174:104695.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
103.  Okazaki M, Fushida S, Harada S, Tsukada T, Kinoshita J, Oyama K, Tajima H, Ninomiya I, Fujimura T, Ohta T. The angiotensin II type 1 receptor blocker candesartan suppresses proliferation and fibrosis in gastric cancer. Cancer Lett. 2014;355:46-53.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
104.  Fujita N, Fujita K, Iwama H, Kobara H, Fujihara S, Chiyo T, Namima D, Yamana H, Kono T, Takuma K, Hirata M, Kobayashi K, Kato K, Kamada H, Morishita A, Tsutsui K, Himoto T, Okano K, Suzuki Y, Masaki T. Antihypertensive drug telmisartan suppresses the proliferation of gastric cancer cells in vitro and in vivo. Oncol Rep. 2020;44:339-348.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
105.  Po WW, Thein W, Khin PP, Khing TM, Han KWW, Park CH, Sohn UD. Fluoxetine Simultaneously Induces Both Apoptosis and Autophagy in Human Gastric Adenocarcinoma Cells. Biomol Ther (Seoul). 2020;28:202-210.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
106.  Hu Q, Hou YC, Huang J, Fang JY, Xiong H. Itraconazole induces apoptosis and cell cycle arrest via inhibiting Hedgehog signaling in gastric cancer cells. J Exp Clin Cancer Res. 2017;36:50.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 33]  [Cited by in RCA: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
107.  Mao SH, Zhu CH, Nie Y, Yu J, Wang L. Levobupivacaine Induces Ferroptosis by miR-489-3p/SLC7A11 Signaling in Gastric Cancer. Front Pharmacol. 2021;12:681338.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 50]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
108.  Wang L, Wang H. The putative role of ferroptosis in gastric cancer: a review. Eur J Cancer Prev. 2023;32:575-583.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
109.  Luo Y, Bai XY, Zhang L, Hu QQ, Zhang N, Cheng JZ, Hou MZ, Liu XL. Ferroptosis in Cancer Therapy: Mechanisms, Small Molecule Inducers, and Novel Approaches. Drug Des Devel Ther. 2024;18:2485-2529.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
110.  Liu BH, Yuan TM, Huang CJ, Hsu DT, Chen SW, Hsiao NW, Lin SC, Wu SW, Lin YJ, Chuang SM. DNA repair proteins as the targets for paroxetine to induce cytotoxicity in gastric cancer cell AGS. Am J Cancer Res. 2022;12:1465-1483.  [PubMed]  [DOI]
111.  Kowalska M, Nowaczyk J, Fijałkowski Ł, Nowaczyk A. Paroxetine-Overview of the Molecular Mechanisms of Action. Int J Mol Sci. 2021;22:1662.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
112.  Chen VC, Hsu TC, Lin CF, Huang JY, Chen YL, Tzang BS, McIntyre RS. Association of Risperidone With Gastric Cancer: Triangulation Method From Cell Study, Animal Study, and Cohort Study. Front Pharmacol. 2022;13:846455.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
113.  CIMA  Medicine Online Information Center of AEMPS - CIMA. [cited September 16, 2025]. Available from: https://cima.aemps.es/cima/publico/home.html#.  [PubMed]  [DOI]