Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Jun 24, 2025; 16(6): 104314
Published online Jun 24, 2025. doi: 10.5306/wjco.v16.i6.104314
Immunotherapy in gastrointestinal stromal tumors: Current landscape and future horizons
Stavros P Papadakos, Alexandra Argyrou, First Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens 11527, Greece
Stavros P Papadakos, Stamatios Theocharis, First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
Ioannis Karniadakis, Upper Gastrointestinal Surgery, Department of General Surgery, St. George's Hospital, St. George's University Hospitals NHS Foundation Trust, London 84790, United Kingdom
Stamatina Vogli, Department of Gastroenterology, Metaxa Oncologic Hospital of Piraeus, Athens, Greece, Athens 18537, Greece
ORCID number: Stavros P Papadakos (0000-0003-1583-1125); Alexandra Argyrou (0000-0002-1569-5592); Ioannis Karniadakis (0000-0001-9924-0044); Stamatina Vogli (0000-0003-4944-2611); Stamatios Theocharis (0000-0002-0930-1657).
Co-first authors: Stavros P Papadakos and Alexandra Argyrou.
Author contributions: Papadakos SP and Argyrou A contributed equally to the work and should be considered co-first authors; Papadakos SP and Theocharis S contributed to the conceptualization and study design; Papadakos SP wrote the first draft of the manuscript; Argyrou A, Karniadakis I, and Vogli S contributed to the literature review and manuscript drafting; Theocharis S and Papadakos SP critically reviewed and edited the manuscript for important intellectual content. All authors have read and approved the final manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
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: Stamatios Theocharis, MD, Senior Researcher, First Department of Pathology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma Street, Goudi, Athens 11527, Greece. stamtheo@med.uoa.gr
Received: December 20, 2024
Revised: February 10, 2025
Accepted: March 5, 2025
Published online: June 24, 2025
Processing time: 185 Days and 11.3 Hours

Abstract

Gastrointestinal stromal tumors (GISTs) feature a unique tumor microenvironment (TME) with abundant immune infiltrates, including CD8+ T cells and tertiary lymphoid structures, alongside significant immune escape mechanisms such as indoleamine 2,3-dioxygenase (IDO) overexpression, MHC I loss, and regulatory T-cell activity. These factors contribute to an immunosuppressive TME, limiting the effectiveness of immune responses. Recent proteomic and immune profiling has identified distinct immune clusters, ranging from highly infiltrated "hot" tumors to immune-desert "cold" tumors, offering new insights into immune heterogeneity and prognostic stratification. While tyrosine kinase inhibitors (TKIs) like imatinib have shown immunomodulatory effects, clinical trials with immune checkpoint inhibitors (ICIs) alone or in combination have yielded modest outcomes. This editorial examines the immunologic landscape of GIST, explores the interplay between ICIs and TKIs, and highlights emerging therapeutic strategies such as IDO inhibition, bispecific antibodies, and patient selection based on TME characteristics. These insights pave the way for more effective immunotherapy approaches in GIST.

Key Words: Gastrointestinal stromal tumors; Tumor microenvironment; Immune checkpoint inhibitors; Biomarker-driven therapy; Immunotherapy strategies

Core Tip: Immunotherapy for gastrointestinal stromal tumors (GISTs) remains an emerging field with promising but early-stage developments. This editorial summarizes the current landscape of immunotherapy in GISTs, focusing on the immune microenvironment, advances in biomarker discovery, and innovative strategies such as immune checkpoint inhibitors, cytokine-based therapies, and adoptive cell therapies. Future directions emphasize the integration of immune profiling and personalized combination therapies to optimize treatment outcomes. Understanding these dynamics will be essential for overcoming resistance and improving precision oncology in GIST management.



INTRODUCTION

According to the latest guidelines, tyrosine kinase inhibitors (TKIs) are the cornerstone of therapeutic management for gastrointestinal stromal tumors (GISTs)[1,2]. Their use is tailored to the tumor's mutational profile, with imatinib serving as the first-line therapy for GISTs harboring KIT or PDGFRA mutations sensitive to this agent. For cases with imatinib-resistant mutations, such as PDGFRA exon 18 D842V, avapritinib is preferred[3]. Subsequent lines of therapy include sunitinib, regorafenib, and ripretinib for patients whose disease progresses on prior TKIs. Dose adjustments and alternative options are employed to manage resistance, highlighting the pivotal role of TKIs across all stages of GIST treatment[3,4]. At present, immunotherapy for GISTs is still in the clinical trial phase and has not yet been adopted into clinical practice. However, there is a growing interest in this approach, as emerging data highlight the potential of targeting the immune microenvironment in GISTs. Table 1 summarizes the results from clinical trials evaluating immunotherapeutic approaches in GIST[5-10].

Table 1 summarizes the results of clinical trials evaluating various immunotherapeutic approaches in gastrointestinal stromal tumors.
Ref.
Treatment
Phase
Number of patients
ORR (%)
Median PFS
Median OS
Discussion
Chen et al[5], 2012Peg-IFNa2b + ImatinibII8100%NR (> 3 years)NRNew PR achieved after reintroduction of Peg-IFNa2b in a patient who progressed on imatinib maintenance therapy
D’Angelo et al[6], 2017Dasatinib + IpilimumabIb2053.8% (by Choi criteria)2.8 months13.5 months7/13 evaluable GISTs had PR by Choi criteria
Toulmonde et al[7], 2018Pembrolizumab + CyclophosphamideII90%6 months PFS: 11%-63% of GISTs showed high IDO expression
Singh et al[8], 2019Nivolumab +/- IpilimumabIIN: 15, N + I: 12N: 0%, N + I: 8.3%N: 8.57 weeks, N + I: 9.1 weeksN:9.1 months, N + I: 12.1 monthsResponses and disease control observed, drugs well tolerated
Chen et al[9], 2020Nivolumab +/- IpilimumabIIN: 9, N + I: 9N: 0%, N + I: 0%N: 1.5 months, N + I: 2.9 months-No confirmed responses in GIST
NCT05152472[10]Imatinib + Atezolizumab vs Imatinib aloneII110 (planned)OngoingOngoingOngoingTrial ongoing to assess PFS improvement with combination therapy
NCT03609424[10]PDR001 + ImatinibI/II39Not reportedNot reportedNot reportedNot reported
ADVANCING GIST IMMUNOTHERAPY THROUGH COMPREHENSIVE IMMUNE PROFILING

Cancer immunotherapy has revolutionized oncology by utilizing the immune system to target and eliminate malignancies, including GISTs[11]. Techniques such as adoptive cell transfer (ACT) and immune checkpoint inhibitors (ICIs) have achieved substantial success, delivering durable responses in a subset of patients. However, the efficacy of these therapies is closely intertwined with the complex and dynamic nature of the tumor microenvironment (TME), particularly the roles and characteristics of tumor-infiltrating immune cells[12,13]. Advances in single-cell technologies, such as single-cell RNA sequencing (scRNA-seq) and mass cytometry, have enabled detailed profiling of the GIST TME, providing insights into the heterogeneity and functional diversity of immune cells within tumors[14,15]. Key findings from recent research underscore the significant roles of cytotoxic T-cells[16,17], regulatory T-cells[18], tumor-associated macrophages (TAMs)[19,20], and other immune populations in shaping the response to immunotherapy in GISTs[18]. For instance, increased presence of cytotoxic CD8+ T-cells often correlates with better responses to ICIs, whereas an abundance of immunosuppressive TAMs and regulatory T-cells may predict resistance to such therapies[15].

The emerging understanding of these immune dynamics informs strategies for enhancing immunotherapy in GISTs. For example, modulating TAM polarization[21], reactivating exhausted T-cells[22], or targeting immune checkpoint proteins such as PD-1/PD-L1 and CTLA-4 have become central to therapeutic innovation[16,23]. Furthermore, the identification of new biomarkers through single-cell analysis can guide patient stratification and improve the precision of immunotherapeutic interventions[7,24]. Imatinib, widely recognized for its role in targeting c-kit and PDGFRa, also exhibits notable immunomodulatory effects in the management of GISTs[14]. Beyond its oncogenic inhibition, imatinib promotes immune activation by reversing M2 macrophage polarization[25], impairing dendritic cell differentiation[26], and enhancing NK cell activity through c-kit inhibition[27]. This leads to increased Th1 responses, CD8+ T-cell infiltration, and a reduction in regulatory T cells and indoleamine 2,3-dioxygenase (IDO) expression, thereby mitigating immune escape and supporting tumor suppression[14]. Overall, the integration of advanced single-cell profiling technologies with clinical research is accelerating progress in GIST immunotherapy.

EMERGING IMMUNOTHERAPY STRATEGIES FOR GIST: TARGETING THE TME

Immunotherapy is a promising strategy in the management of GISTs. The immune microenvironment plays a critical role in GIST progression, and targeting it has been proposed to enhance the effectiveness of therapies like imatinib. Immunotherapeutic strategies for GISTs can be classified into several key categories: Cytokine-based therapies, ICIs, antibody-based treatments, antibody-drug conjugates (ADCs), vaccine-based immunotherapies, and ACTs[14,15].

Cytokine-based therapies, such as pegylated interferon-α-2b (PegIFNα2b), have shown promise in modulating immune responses. In patients with advanced stage III/IV GISTs, the combination of PegIFNα2b and imatinib showed notable immunological and clinical benefits. The therapy enhanced the infiltration of immune cells producing IFN-γ, such as CD4+ and CD8+ T cells and natural killer (NK) cells, into the TME. These immune responses correlated with a 100% overall response rate, highlighting the synergistic potential of using PegIFNα2b alongside imatinib[5]. The combination of Peg-IFNα-2b and imatinib synergistically inhibits proliferation and promotes apoptosis in imatinib-resistant GIST cells by downregulating p-mTOR and Bcl-2 expression, targeting the PI3K/Akt/mTOR pathway. This suggests a potential strategy for overcoming imatinib resistance[28]. ICIs, including anti-PD-1[7-9,29-31]. Anti-PD-L1, and anti-CTLA-4[6,32,33] antibodies, have shown limited success in advanced GIST cases, partly due to the immunosuppressive TME characterized by low PD-L1 expression, high levels of M2 macrophages, and regulatory T cells[14]. However, patients with specific features, such as PDGFRA D842V mutations[34], high PD-L1 expression, or TLS presence, may derive more benefit from these therapies[8]. Early intervention with ICIs or their use in combination with imatinib may further improve their effectiveness.

Antibody-based therapies and ADCs are being developed to target key molecules in GISTs, such as KIT, PDGFRA, and SSTR2. For instance, anti-KIT monoclonal antibodies like SR1 have demonstrated potential in reducing KIT expression and enhancing macrophage-mediated tumor cell death, independent of imatinib sensitivity[35]. Edris et al[36] demonstrated that the anti-KIT monoclonal antibody SR1 effectively inhibited the growth of both imatinib-sensitive and imatinib-resistant GISTs. In vitro, SR1 reduced cell proliferation equally across several GIST cell lines by downregulating cell-surface KIT expression. Additionally, SR1 enhanced macrophage-mediated phagocytosis of GIST cells, indicating an immune-mediated mechanism of tumor clearance[36]. In vivo, SR1 significantly suppressed tumor growth in xenograft models, reducing imatinib-resistant GIST430 tumor size by 10-fold and imatinib-sensitive GIST882 tumors by fivefold. These findings suggest SR1’s dual action—direct KIT inhibition and stimulation of innate immune responses—offers a promising approach for overcoming imatinib resistance in GIST therapy[36]. ADCs, including anti-KIT conjugates like LOP628-DM1 and anti-GPR20 ADCs, have shown efficacy in preclinical models[37], though clinical application remains limited by side effects such as hypersensitivity reactions[38].

Vaccine-based therapies and ACTs are also emerging as innovative approaches. Ilixadencel, an allogeneic dendritic cell vaccine, has shown tumor responses in some GIST patients[39]. Ilixadencel is an allogeneic, cell-based immune primer administered directly into tumors to stimulate an anti-cancer immune response[40]. It releases pro-inflammatory cytokines and chemokines that recruit and activate NK cells and dendritic cells at the tumor site. Activated NK cells kill tumor cells, releasing tumor-associated antigens, while the cytokines from both ilixadencel and NK cells promote the maturation and cross-presentation ability of recruited dendritic cells. These dendritic cells then present the antigens to cytotoxic CD8+ T cells, triggering a systemic immune response against the tumor[40]. Additionally, ilixadencel helps reduce local immunosuppression by inhibiting suppressive cells like regulatory T cells and myeloid-derived suppressor cells[40]. CAR-T cell therapies targeting KIT and other tumor antigens have demonstrated effectiveness in preclinical studies. Adoptive transfer of cytokine-induced killer (CIK) cells has shown promise in overcoming resistance to standard therapies[41]. The therapy functions by utilizing CIK lymphocytes and interferon-alpha (IFN-α) to target KIT/PDGFRA wild-type GIST (wtGIST)[41]. CIK cells eliminate tumor cells through the release of granzyme B and secrete high levels of IFN-α and IFN-γ, which stimulate immune responses. Additionally, CIK cells produce factors that increase the expression of PD-L1/2 and HLA-I on tumor cells, improving their visibility to the immune system[41]. IFN-α independently exerts antitumor effects in certain wtGIST cells, particularly those with elevated IFN receptor expression. Tumor cells that persist after IFN-α exposure remain vulnerable to CIK cell-mediated killing, indicating a synergistic effect between the two treatments[41]. Additionally, novel targets such as M2 macrophages, Treg cells, LAG3, Tim-3, WT1, and CSPG4 are being explored to address immune escape mechanisms in GISTs[14].

ADVANCING IMMUNOTHERAPY IN GIST: INSIGHTS INTO BIOMARKER-DRIVEN STRATEGIES

While these approaches hold significant promise, further refinement of immunotherapy strategies is required. This includes identifying reliable biomarkers for patient selection, and developing personalized combination therapies to optimize immune responses and overcome treatment resistance. Two recent studies provide valuable insights in this direction[22,42]. Petitprez et al[22] explored the significance of B cells and tertiary lymphoid structures (TLSs) in the TME of soft tissue sarcomas, including GISTs, and their impact on survival and immunotherapy outcomes. By analyzing gene expression profiles from 608 samples, five sarcoma immune classes (SICs) were identified, with SIC E standing out for its immune-rich characteristics, including a high density of B cells within TLSs. Notably, tumors with abundant B cell infiltration, as seen in SIC E, were associated with improved overall survival, even when CD8+ T cell levels were low, highlighting the independent prognostic value of B cells. TLSs in SIC E tumors were particularly rich in B cells, T cells, and dendritic cells, and are believed to serve as hubs for generating antitumor immune responses. Additionally, SIC E tumors exhibited a higher response rate to PD1 blockade therapies like pembrolizumab compared to other immune classes, with a 50% objective response rate. This underscores the potential of B cell-rich TMEs to enhance the effectiveness of immunotherapy. Biomarkers such as the presence of TLSs, increased CXCL13 expression, and B cell abundance emerged as critical predictors of response to ICI[22]. Sun et al[42] investigated the proteomic and phosphoproteomic characteristics of GISTs in 193 patients. They identified four distinct immune clusters, each with varying levels of immune and stromal cell infiltration. These clusters demonstrated significant differences in progression-free survival. The first two clusters (Im-I and Im-II) were characterized by higher levels of T-cell infiltration, including CD4+ and CD8+ T cells, and upregulation of antigen-presentation pathways. These clusters showed improved survival outcomes, highlighting the potential protective role of T-cell activity. However, cluster Im-I exhibited elevated levels of CD276, a molecule associated with immune evasion[43], which could suppress CD8+ T-cell activity and suggest potential benefits from therapies targeting immune checkpoint pathways. Cluster Im-III, predominantly associated with high-risk tumors, was enriched in pathways related to ERK1 and ERK2 cascades, indicating a more aggressive tumor biology. In contrast, cluster Im-IV exhibited the lowest immune scores and minimal immune cell infiltration, suggesting a "cold" TME with poor immune engagement. By integrating immune and proteomic clustering, they further refined the classification of stomach GISTs, revealing combinations of immune and molecular profiles that influence prognosis. This analysis underscores the heterogeneity of immune responses in GISTs and highlights the potential for immune-based therapeutic strategies to improve outcomes in specific patient subgroups.

Building on these insights into the TME and immune profiling of GISTs, it is crucial to consider how underlying genetic mutations further shape the immune landscape and influence therapeutic outcomes. KIT-mutant GISTs typically show lower levels of immune cell infiltration, including CD8+ T lymphocytes and NK cells, and exhibit reduced expression of MHC-I molecules, contributing to immune evasion and potentially limiting the efficacy of immunotherapies[27,44]. Conversely, PDGFRA-mutant GISTs, particularly those with the D842V mutation, demonstrate a more immunogenic profile with higher infiltration of CD8+ T cells, NK cells, and dendritic cells, along with increased expression of immune checkpoint molecules such as PD-L1 and IDO[34,44]. This enhanced immune surveillance correlates with better clinical outcomes and suggests a potential for improved responsiveness to ICIs[34,44,45]. In contrast, KIT/PDGFRA-wild-type GISTs, which include succinate dehydrogenase (SDH)-deficient and SDH-competent subtypes, present a more complex immune landscape. SDH-deficient GISTs often exhibit a pronounced immunosuppressive microenvironment characterized by low levels of MHC-I expression and minimal infiltration of cytotoxic immune cells, which may contribute to their resistance to both TKIs and immunotherapy[44]. Some studies suggest that KIT/PDGFRA-WT GISTs may have varying degrees of immune cell infiltration, with conflicting reports on the abundance of CD3+, CD4+, and CD8+ T cells. While Sun et al[46] reported increased CD8+ T cell infiltration in WT GISTs compared to KIT- and PDGFRA-mutant tumors, Gasparotto et al[44] found reduced T cell infiltration and lower expression of MHC-I and immune checkpoint molecules in WT GISTs, highlighting the heterogeneity within this subgroup. These differences in immune cell infiltration and checkpoint molecule expression have important prognostic implications and may influence the effectiveness of emerging immunotherapeutic strategies.

Beyond genetic drivers, systemic inflammatory biomarkers like the neutrophil-to-lymphocyte ratio[47], platelet-to-lymphocyte ratio, and systemic immune-inflammation index have emerged as prognostic indicators, correlating with tumor progression, recurrence risk, and treatment response[48,49]. The TME also plays a pivotal role, with high infiltration of CD8+ T lymphocytes and NK cells associated with favorable outcomes, while increased M2 macrophage and Treg infiltration contribute to immunosuppression and poorer prognosis[27,50,51]. In addition to traditional biomarkers, the expression of immune checkpoint molecules such as PD-L1, IDO, and Tim-3 has gained attention for their prognostic and therapeutic implications in GIST management. Although PD-L1 expression shows variability, some studies suggest that higher levels correlate with improved recurrence-free survival (RFS), while others associate it with resistance to therapy, underscoring the complexity of its role[7,46,52]. Similarly, elevated IDO expression driven by KIT signaling contributes to an immunosuppressive microenvironment, and its inhibition has shown promise in enhancing the efficacy of imatinib and PD-1 blockade in preclinical models[52,53]. Furthermore, the presence of cancer-testis antigens, such as NY-ESO-1 and MAGE-A3, in 26.7%-40% of GISTs has been linked to poor responses to TKIs and shorter RFS, suggesting their potential as biomarkers for aggressive disease and targets for immunotherapy[54,55]. Finally, GISTs generally exhibit a low tumor mutation burden (TMB), though more aggressive forms show increased genomic alterations like copy number changes and structural variants[56]. Despite the low TMB, certain mutations can lead to the formation of neoantigens. For instance, the KIT A502_Y503 duplication mutation, frequently observed in Chinese GIST patients, results in neoantigen peptides that strongly bind to MHC class I molecules. This mutation, present in around 16% of cases, is often linked to reduced sensitivity to imatinib, but it opens the door for neoantigen-targeted immunotherapies as a potential treatment strategy[57].

These insights highlight the growing importance of integrating molecular, immunological, and systemic biomarkers into personalized GIST management strategies. The above is briefly summarized in Table 2[58,59].

Table 2 summarizes the basic categories of potential gastrointestinal stromal tumors biomarkers.
Type
Biomarker
Clinical relevance
Ref.
GeneticKIT and PDGFRA mutationsGuide TKI therapy; KIT exon 11 mutations respond well, PDGFRA-D842V mutations show resistance[58]
GeneticSDH-deficient/competent GISTShow resistance to TKIs; alternative treatments required[58]
Inflammatory biomarkerNeutrophil-to-lymphocyte ratioHigh ratio indicates poor prognosis due to increased neutrophil-driven inflammation and reduced lymphocyte-mediated immunity[53]
Inflammatory biomarkerSystemic immune-inflammation indexElevated index correlates with tumor progression by promoting pro-tumor inflammation[48,49]
Immune cell markerCD8+ T lymphocyte infiltrationIncreased CD8+ T cells enhance cytotoxic responses against tumor cells, improving survival[27,50,52]
Immune cell markerM2 macrophage infiltrationHigh M2 macrophages suppress immune responses and promote tumor growth and angiogenesis[53]
Immune cell markerB Cell and TLS presenceDense B cells and TLS formation boost antitumor immunity and predict better immunotherapy response[59]
Immune checkpointPD-L1 expressionModulates T cell activity; high levels can inhibit immune responses but may predict immunotherapy success[7,52]
Immune checkpointIndoleamine 2,3-dioxygenaseDepletes tryptophan, suppressing T cell function and fostering immune tolerance in the tumor microenvironment[52-54]]
Immune checkpointTim-3/Gal-9 axisTim-3 promotes T cell exhaustion; Gal-9 association linked to reduced CD8+ T cell infiltration[51]
Tumor antigensCancer-testis antigens (NY-ESO-1, MAGE-A3)Trigger immune responses but are linked to poor TKI response and aggressive tumor behavior[54,55]
Chemokine biomarkerCXCL13 expressionAttracts B cells to tumor sites, facilitating TLS formation and enhancing immune response[59]
Immune profilingImmune clusters (Im-I to Im-IV)High T cell presence in clusters improves survival; Im-I cluster shows immune evasion via CD276 expression[47]
Molecular pathwayERK1/ERK2 pathway activationDrives tumor proliferation and is linked to immune-poor, aggressive GIST subtypes[42]
Immune evasion markerCD276 (B7-H3) expressionInhibits CD8+ T cell activity, contributing to immune evasion and tumor progression[43]

The above emphasize the potential of immune profiling and biomarker discovery to enhance immunotherapy outcomes, offering a path toward more precise and effective treatments for GISTs.

CONCLUSION

While early findings from small, retrospective studies are encouraging, validation through large-scale, prospective research is essential to confirm these results. Future investigations should focus on longitudinal studies to evaluate the long-term efficacy and safety of PD-1/PD-L1 inhibitors and IDO-targeted therapies, particularly in combination with established TKIs[60]. IDO1 inhibitors were explored as a strategy to overcome resistance to immune checkpoint inhibitors by targeting tryptophan metabolism and reducing immunosuppression in the tumor microenvironment[61]. While early trials showed promise[62-64], phase 3 studies[65], including the KEYNOTE-252/ECHO-301 trial, failed to demonstrate clinical benefit[60,66]. The heterogeneity of IDO1 expression and the activation of compensatory pathways, such as AhR and TDO2, may have contributed to these failures[60]. Moreover, exploring the role of M2 TAM polarization and its influence on treatment outcomes remains a critical area for further research. Biomarker-driven approaches, including comprehensive immune profiling and proteomics, offer great potential for identifying novel biomarkers such as B cells and TLSs within the GIST immune microenvironment. These biomarkers could guide more personalized treatment strategies by taking into account the tumor’s immune characteristics and levels of immune cell infiltration. For example, bispecific antibodies (bsAbs) represent a promising class of immunotherapies designed to engage multiple targets simultaneously, enhancing precision and efficacy in cancer treatment[67]. While their success has been particularly evident in hematologic malignancies[68], their application in GISTs, remains an area of active investigation. GISTs are primarily driven by KIT and PDGFRA mutations, with targeted TKI like imatinib being the standard treatment[58]. However, resistance to TKIs often emerges, necessitating alternative therapeutic strategies. Bispecific antibodies could potentially offer a novel approach by leveraging immune activation against GIST cells[67]. For instance, a bsAb targeting KIT-expressing tumor cells while engaging T cells via CD3 could promote direct immune-mediated cytotoxicity[69]. Alternatively, bispecific antibodies that combine checkpoint inhibition with tumor antigen targeting might enhance the immune response within the typically immunosuppressive GIST microenvironment[70]. However, their integration into clinical practice will require robust validation through large, multi-center clinical trials to refine patient selection and optimize immunotherapy outcomes. Ultimately, the successful application of these biomarkers has the potential to significantly improve the precision and effectiveness of GIST treatments, leading to more tailored therapeutic interventions and better outcomes for patients.

Footnotes

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

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: Greece

Peer-review report’s classification

Scientific Quality: Grade B, Grade C, Grade C, Grade D

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

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

Scientific Significance: Grade A, Grade B, Grade C, Grade D

P-Reviewer: Li PM; Wang ZS; Liao HM S-Editor: Liu H L-Editor: A P-Editor: Zhao YQ

References
1.  von Mehren M, Kane JM, Riedel RF, Sicklick JK, Pollack SM, Agulnik M, Bui MM, Carr-Ascher J, Choy E, Connelly M, Dry S, Ganjoo KN, Gonzalez RJ, Holder A, Homsi J, Keedy V, Kelly CM, Kim E, Liebner D, McCarter M, McGarry SV, Mesko NW, Meyer C, Pappo AS, Parkes AM, Petersen IA, Poppe M, Schuetze S, Shabason J, Spraker MB, Zimel M, Bergman MA, Sundar H, Hang LE. NCCN Guidelines® Insights: Gastrointestinal Stromal Tumors, Version 2.2022. J Natl Compr Canc Netw. 2022;20:1204-1214.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 57]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
2.  Akahoshi K, Oya M, Koga T, Shiratsuchi Y. Current clinical management of gastrointestinal stromal tumor. World J Gastroenterol. 2018;24:2806-2817.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 180]  [Cited by in RCA: 232]  [Article Influence: 33.1]  [Reference Citation Analysis (9)]
3.  Serrano C, Martín-Broto J, Asencio-Pascual JM, López-Guerrero JA, Rubió-Casadevall J, Bagué S, García-Del-Muro X, Fernández-Hernández JÁ, Herrero L, López-Pousa A, Poveda A, Martínez-Marín V. 2023 GEIS Guidelines for gastrointestinal stromal tumors. Ther Adv Med Oncol. 2023;15:17588359231192388.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 23]  [Cited by in RCA: 49]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
4.  Papadakos SP, Tsagkaris C, Papadakis M, Papazoglou AS, Moysidis DV, Zografos CG, Theocharis S. Angiogenesis in gastrointestinal stromal tumors: From bench to bedside. World J Gastrointest Oncol. 2022;14:1469-1477.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 8]  [Cited by in RCA: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
5.  Chen LL, Chen X, Choi H, Sang H, Chen LC, Zhang H, Gouw L, Andtbacka RH, Chan BK, Rodesch CK, Jimenez A, Cano P, Jones KA, Oyedeji CO, Martins T, Hill HR, Schumacher J, Willmore C, Scaife CL, Ward JH, Morton K, Randall RL, Lazar AJ, Patel S, Trent JC, Frazier ML, Lin P, Jensen P, Benjamin RS. Exploiting antitumor immunity to overcome relapse and improve remission duration. Cancer Immunol Immunother. 2012;61:1113-1124.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 36]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
6.  D'Angelo SP, Shoushtari AN, Keohan ML, Dickson MA, Gounder MM, Chi P, Loo JK, Gaffney L, Schneider L, Patel Z, Erinjeri JP, Bluth MJ, Sjoberg A, Streicher H, Takebe N, Qin LX, Antonescu C, DeMatteo RP, Carvajal RD, Tap WD. Combined KIT and CTLA-4 Blockade in Patients with Refractory GIST and Other Advanced Sarcomas: A Phase Ib Study of Dasatinib plus Ipilimumab. Clin Cancer Res. 2017;23:2972-2980.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 96]  [Cited by in RCA: 99]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
7.  Toulmonde M, Penel N, Adam J, Chevreau C, Blay JY, Le Cesne A, Bompas E, Piperno-Neumann S, Cousin S, Grellety T, Ryckewaert T, Bessede A, Ghiringhelli F, Pulido M, Italiano A. Use of PD-1 Targeting, Macrophage Infiltration, and IDO Pathway Activation in Sarcomas: A Phase 2 Clinical Trial. JAMA Oncol. 2018;4:93-97.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 266]  [Cited by in RCA: 318]  [Article Influence: 45.4]  [Reference Citation Analysis (0)]
8.  Singh AS, Hecht JR, Rosen L, Wainberg ZA, Wang X, Douek M, Hagopian A, Andes R, Sauer L, Brackert SR, Chow W, DeMatteo R, Eilber FC, Glaspy JA, Chmielowski B. A Randomized Phase II Study of Nivolumab Monotherapy or Nivolumab Combined with Ipilimumab in Patients with Advanced Gastrointestinal Stromal Tumors. Clin Cancer Res. 2022;28:84-94.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
9.  Chen JL, Mahoney MR, George S, Antonescu CR, Liebner DA, Van Tine BA, Milhem MM, Tap WD, Streicher H, Schwartz GK, D'angelo SP. A multicenter phase II study of nivolumab +/- ipilimumab for patients with metastatic sarcoma (Alliance A091401): Results of expansion cohorts. JCO. 2020;38:11511-11511.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 20]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
10.  Mechahougui H, Michael M, Friedlaender A. Precision Oncology in Gastrointestinal Stromal Tumors. Curr Oncol. 2023;30:4648-4662.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
11.  Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 2020;17:807-821.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1361]  [Cited by in RCA: 1701]  [Article Influence: 340.2]  [Reference Citation Analysis (0)]
12.  Papadakos SP, Arvanitakis K, Stergiou IE, Koutsompina ML, Germanidis G, Theocharis S. γδ T Cells: A Game Changer in the Future of Hepatocellular Carcinoma Immunotherapy. Int J Mol Sci. 2024;25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
13.  Papadakos SP, Machairas N, Stergiou IE, Arvanitakis K, Germanidis G, Frampton AE, Theocharis S. Unveiling the Yin-Yang Balance of M1 and M2 Macrophages in Hepatocellular Carcinoma: Role of Exosomes in Tumor Microenvironment and Immune Modulation. Cells. 2023;12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
14.  Li B, Chen H, Yang S, Chen F, Xu L, Li Y, Li M, Zhu C, Shao F, Zhang X, Deng C, Zeng L, He Y, Zhang C. Advances in immunology and immunotherapy for mesenchymal gastrointestinal cancers. Mol Cancer. 2023;22:71.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 26]  [Reference Citation Analysis (0)]
15.  Roulleaux Dugage M, Jones RL, Trent J, Champiat S, Dumont S. Beyond the Driver Mutation: Immunotherapies in Gastrointestinal Stromal Tumors. Front Immunol. 2021;12:715727.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
16.  Seifert AM, Zeng S, Zhang JQ, Kim TS, Cohen NA, Beckman MJ, Medina BD, Maltbaek JH, Loo JK, Crawley MH, Rossi F, Besmer P, Antonescu CR, DeMatteo RP. PD-1/PD-L1 Blockade Enhances T-cell Activity and Antitumor Efficacy of Imatinib in Gastrointestinal Stromal Tumors. Clin Cancer Res. 2017;23:454-465.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 94]  [Cited by in RCA: 114]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
17.  Zhao R, Song Y, Wang Y, Huang Y, Li Z, Cui Y, Yi M, Xia L, Zhuang W, Wu X, Zhou Y. PD-1/PD-L1 blockade rescue exhausted CD8+ T cells in gastrointestinal stromal tumours via the PI3K/Akt/mTOR signalling pathway. Cell Prolif. 2019;52:e12571.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 65]  [Cited by in RCA: 105]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
18.  Dufresne A, Lesluyes T, Ménétrier-Caux C, Brahmi M, Darbo E, Toulmonde M, Italiano A, Mir O, Le Cesne A, Le Guellec S, Valentin T, Chevreau C, Bonvalot S, Robin YM, Coindre JM, Caux C, Blay JY, Chibon F. Specific immune landscapes and immune checkpoint expressions in histotypes and molecular subtypes of sarcoma. Oncoimmunology. 2020;9:1792036.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
19.  Pantaleo MA, Tarantino G, Agostinelli C, Urbini M, Nannini M, Saponara M, Castelli C, Stacchiotti S, Fumagalli E, Gatto L, Santini D, De Leo A, Marafioti T, Akarca A, Sabattini E, Pession A, Ardizzoni A, Indio V, Astolfi A. Immune microenvironment profiling of gastrointestinal stromal tumors (GIST) shows gene expression patterns associated to immune checkpoint inhibitors response. Oncoimmunology. 2019;8:e1617588.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 40]  [Cited by in RCA: 40]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
20.  Cavnar MJ, Zeng S, Kim TS, Sorenson EC, Ocuin LM, Balachandran VP, Seifert AM, Greer JB, Popow R, Crawley MH, Cohen NA, Green BL, Rossi F, Besmer P, Antonescu CR, DeMatteo RP. KIT oncogene inhibition drives intratumoral macrophage M2 polarization. J Exp Med. 2013;210:2873-2886.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 101]  [Cited by in RCA: 111]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
21.  Tamura R, Tanaka T, Yamamoto Y, Akasaki Y, Sasaki H. Dual role of macrophage in tumor immunity. Immunotherapy. 2018;10:899-909.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 57]  [Cited by in RCA: 100]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
22.  Petitprez F, de Reyniès A, Keung EZ, Chen TW, Sun CM, Calderaro J, Jeng YM, Hsiao LP, Lacroix L, Bougoüin A, Moreira M, Lacroix G, Natario I, Adam J, Lucchesi C, Laizet YH, Toulmonde M, Burgess MA, Bolejack V, Reinke D, Wani KM, Wang WL, Lazar AJ, Roland CL, Wargo JA, Italiano A, Sautès-Fridman C, Tawbi HA, Fridman WH. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577:556-560.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1306]  [Cited by in RCA: 1273]  [Article Influence: 254.6]  [Reference Citation Analysis (0)]
23.  Danaher P, Warren S, Lu R, Samayoa J, Sullivan A, Pekker I, Wallden B, Marincola FM, Cesano A. Pan-cancer adaptive immune resistance as defined by the Tumor Inflammation Signature (TIS): results from The Cancer Genome Atlas (TCGA). J Immunother Cancer. 2018;6:63.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 292]  [Cited by in RCA: 316]  [Article Influence: 45.1]  [Reference Citation Analysis (0)]
24.  Ravishankar B, Liu H, Shinde R, Chandler P, Baban B, Tanaka M, Munn DH, Mellor AL, Karlsson MC, McGaha TL. Tolerance to apoptotic cells is regulated by indoleamine 2,3-dioxygenase. Proc Natl Acad Sci U S A. 2012;109:3909-3914.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 137]  [Cited by in RCA: 163]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
25.  van Dongen M, Savage ND, Jordanova ES, Briaire-de Bruijn IH, Walburg KV, Ottenhoff TH, Hogendoorn PC, van der Burg SH, Gelderblom H, van Hall T. Anti-inflammatory M2 type macrophages characterize metastasized and tyrosine kinase inhibitor-treated gastrointestinal stromal tumors. Int J Cancer. 2010;127:899-909.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 61]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
26.  Medina BD, Liu M, Vitiello GA, Seifert AM, Zeng S, Bowler T, Zhang JQ, Cavnar MJ, Loo JK, Param NJ, Maltbaek JH, Rossi F, Balachandran V, DeMatteo RP. Oncogenic kinase inhibition limits Batf3-dependent dendritic cell development and antitumor immunity. J Exp Med. 2019;216:1359-1376.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
27.  Rusakiewicz S, Semeraro M, Sarabi M, Desbois M, Locher C, Mendez R, Vimond N, Concha A, Garrido F, Isambert N, Chaigneau L, Le Brun-Ly V, Dubreuil P, Cremer I, Caignard A, Poirier-Colame V, Chaba K, Flament C, Halama N, Jäger D, Eggermont A, Bonvalot S, Commo F, Terrier P, Opolon P, Emile JF, Coindre JM, Kroemer G, Chaput N, Le Cesne A, Blay JY, Zitvogel L. Immune infiltrates are prognostic factors in localized gastrointestinal stromal tumors. Cancer Res. 2013;73:3499-3510.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 224]  [Cited by in RCA: 251]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
28.  Zhang L. Inhibitory effects of Peg-IFNα 2b and imatinib, alone or in combination, on imatinib-resistant gastrointestinal stromal tumor cell line PC9/GR. WCJD. 2015;23:85.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
29.  Toulmonde M, Penel N, Adam J, Chevreau C, Blay J, Le Cesne A, Bompas E, Piperno-neumann S, Cousin S, Ryckewaert T, Bessede A, Ghiringhelli F, Grellety T, Pulido M, Italiano A. Combination of pembrolizumab and metronomic cyclophosphamide in patients with advanced sarcomas and GIST: A French Sarcoma Group phase II trial. J Clin Oncol. 2017;35:11053.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 8]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
30.  Reddy R, Velagapudi RM, Chitikela SD, Barwad A, Shrivastava S, Dhamija E, Shamim SA, Tripathy S, Pandey R, Rastogi S. Indian experience with immunotherapy in sarcoma and gastrointestinal stromal tumors: a retrospective study. Future Sci OA. 2022;8:FSO795.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
31.  Schroeder BA, Kohli K, O'Malley RB, Kim TS, Jones RL, Pierce RH, Pollack SM. Durable tumor regression in highly refractory metastatic KIT/PDGFRA wild-type GIST following treatment with nivolumab. Oncoimmunology. 2020;9:1710064.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
32.  Shoushtari AN, D'angelo SP, Keohan ML, Dickson MA, Gounder MM, Abdullah AK, Erinjeri JP, Bluth MJ, Ustoyev Y, Condy MM, Streicher H, Takebe N, Dematteo RP, Schwartz GK, Tap WD, Carvajal RD. Combined KIT and CTLA-4 blockade in patients with refractory GIST and other advanced sarcomas. J Clin Oncol. 2014;32:10521-10521.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
33.  Reilley MJ, Bailey A, Subbiah V, Janku F, Naing A, Falchook G, Karp D, Piha-Paul S, Tsimberidou A, Fu S, Lim J, Bean S, Bass A, Montez S, Vence L, Sharma P, Allison J, Meric-Bernstam F, Hong DS. Phase I clinical trial of combination imatinib and ipilimumab in patients with advanced malignancies. J Immunother Cancer. 2017;5:35.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 46]  [Cited by in RCA: 52]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
34.  Sun X, Sun J, Yuan W, Gao X, Fu M, Xue A, Li H, Shu P, Fang Y, Hou Y, Shen K, Sun Y, Qin J, Qin X. Immune Cell Infiltration and the Expression of PD-1 and PD-L1 in Primary PDGFRA-Mutant Gastrointestinal Stromal Tumors. J Gastrointest Surg. 2021;25:2091-2100.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
35.  Edris B, Willingham S, Weiskopf K, Volkmer AK, Volkmer JP, Mühlenberg T, Weissman IL, van de Rijn M. Use of a KIT-specific monoclonal antibody to bypass imatinib resistance in gastrointestinal stromal tumors. Oncoimmunology. 2013;2:e24452.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
36.  Edris B, Willingham SB, Weiskopf K, Volkmer AK, Volkmer JP, Mühlenberg T, Montgomery KD, Contreras-Trujillo H, Czechowicz A, Fletcher JA, West RB, Weissman IL, van de Rijn M. Anti-KIT monoclonal antibody inhibits imatinib-resistant gastrointestinal stromal tumor growth. Proc Natl Acad Sci U S A. 2013;110:3501-3506.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36]  [Cited by in RCA: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
37.  Abrams T, Connor A, Fanton C, Cohen SB, Huber T, Miller K, Hong EE, Niu X, Kline J, Ison-Dugenny M, Harris S, Walker D, Krauser K, Galimi F, Wang Z, Ghoddusi M, Mansfield K, Lee-Hoeflich ST, Holash J, Pryer N, Kluwe W, Ettenberg SA, Sellers WR, Lees E, Kwon P, Abraham JA, Schleyer SC. Preclinical Antitumor Activity of a Novel Anti-c-KIT Antibody-Drug Conjugate against Mutant and Wild-type c-KIT-Positive Solid Tumors. Clin Cancer Res. 2018;24:4297-4308.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 31]  [Cited by in RCA: 43]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
38.  L'Italien L, Orozco O, Abrams T, Cantagallo L, Connor A, Desai J, Ebersbach H, Gelderblom H, Hoffmaster K, Lees E, Maacke H, Schleyer S, Skegro D, Lee-Hoeflich ST. Mechanistic Insights of an Immunological Adverse Event Induced by an Anti-KIT Antibody Drug Conjugate and Mitigation Strategies. Clin Cancer Res. 2018;24:3465-3474.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
39.  Fröbom R, Berglund E, Berglund D, Nilsson IL, Åhlén J, von Sivers K, Linder-Stragliotto C, Suenaert P, Karlsson-Parra A, Bränström R. Phase I trial evaluating safety and efficacy of intratumorally administered inflammatory allogeneic dendritic cells (ilixadencel) in advanced gastrointestinal stromal tumors. Cancer Immunol Immunother. 2020;69:2393-2401.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 20]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
40.  Karlsson-Parra A, Kovacka J, Heimann E, Jorvid M, Zeilemaker S, Longhurst S, Suenaert P. Ilixadencel - an Allogeneic Cell-Based Anticancer Immune Primer for Intratumoral Administration. Pharm Res. 2018;35:156.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
41.  Fiorino E, Merlini A, D'Ambrosio L, Cerviere I, Berrino E, Marchiò C, Giraudo L, Basiricò M, Massa A, Donini C, Leuci V, Rotolo R, Galvagno F, Vitali L, Proment A, Ferrone S, Pisacane A, Pignochino Y, Aglietta M, Grignani G, Mesiano G, Sangiolo D. Integrated Antitumor Activities of Cellular Immunotherapy with CIK Lymphocytes and Interferons against KIT/PDGFRA Wild Type GIST. Int J Mol Sci. 2022;23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
42.  Sun M, Tong Y, Yuan W, Wang Y, Pu Y, Huang W, Lv B, Xu C, Jiang W, Luo R, Fang R, Tang S, Ren L, Wang J, Feng J, Sun C, Shen K, He F, Hou Y, Ding C. Proteomic Characterization Identifies Clinically Relevant Subgroups of Gastrointestinal Stromal Tumors. Gastroenterology. 2024;166:450-465.e33.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
43.  Mochizuki K, Oishi N, Tahara I, Inoue T, Kondo T. Expression of B7-H3 and B7-H4 in Gastric Gastrointestinal Stromal Tumors. Appl Immunohistochem Mol Morphol. 2024;32:484-487.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
44.  Gasparotto D, Sbaraglia M, Rossi S, Baldazzi D, Brenca M, Mondello A, Nardi F, Racanelli D, Cacciatore M, Dei Tos AP, Maestro R. Tumor genotype, location, and malignant potential shape the immunogenicity of primary untreated gastrointestinal stromal tumors. JCI Insight. 2020;5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
45.  Vitiello GA, Bowler TG, Liu M, Medina BD, Zhang JQ, Param NJ, Loo JK, Goldfeder RL, Chibon F, Rossi F, Zeng S, DeMatteo RP. Differential immune profiles distinguish the mutational subtypes of gastrointestinal stromal tumor. J Clin Invest. 2019;129:1863-1877.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 45]  [Cited by in RCA: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
46.  Sun X, Shu P, Fang Y, Yuan W, Zhang Q, Sun J, Fu M, Xue A, Gao X, Shen K, Hou Y, Sun Y, Qin J, Qin X. Clinical and Prognostic Significance of Tumor-Infiltrating CD8+ T Cells and PD-L1 Expression in Primary Gastrointestinal Stromal Tumors. Front Oncol. 2021;11:789915.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
47.  Perez DR, Baser RE, Cavnar MJ, Balachandran VP, Antonescu CR, Tap WD, Strong VE, Brennan MF, Coit DG, Singer S, Dematteo RP. Blood neutrophil-to-lymphocyte ratio is prognostic in gastrointestinal stromal tumor. Ann Surg Oncol. 2013;20:593-599.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 45]  [Cited by in RCA: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
48.  Supsamutchai C, Wilasrusmee C, Hiranyatheb P, Jirasiritham J, Rakchob T, Choikrua P. A cohort study of prognostic factors associated with recurrence or metastasis of gastrointestinal stromal tumor (GIST) of stomach. Ann Med Surg (Lond). 2018;35:1-5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
49.  Mo DC, Liu X, Lv YJ, Qin YQ, Xie GS. Is monocyte-to-lymphocyte ratio the best systematic inflammatory biomarker for the prognosis of gastrointestinal stromal tumors? J Surg Oncol. 2019;119:1024-1025.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
50.  Wei ZW, Wu J, Huang WB, Li J, Lu XF, Yuan YJ, Xiong WJ, Zhang XH, Wang W, He YL, Zhang CH. Immune-infiltration based signature as a novel prognostic biomarker in gastrointestinal stromal tumour. EBioMedicine. 2020;57:102850.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 23]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
51.  Zhuang C, Ni B, Zhang ZZ, Zhao WY, Tu L, Ma XL, Yang LX, Cao H, Wang M. Low Distribution of TIM-3(+) Cytotoxic Tumor-Infiltrating Lymphocytes Predicts Poor Outcomes in Gastrointestinal Stromal Tumors. J Immunol Res. 2021;2021:6647292.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
52.  Blakely AM, Matoso A, Patil PA, Taliano R, Machan JT, Miner TJ, Lombardo KA, Resnick MB, Wang LJ. Role of immune microenvironment in gastrointestinal stromal tumours. Histopathology. 2018;72:405-413.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
53.  Balachandran VP, Cavnar MJ, Zeng S, Bamboat ZM, Ocuin LM, Obaid H, Sorenson EC, Popow R, Ariyan C, Rossi F, Besmer P, Guo T, Antonescu CR, Taguchi T, Yuan J, Wolchok JD, Allison JP, DeMatteo RP. Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat Med. 2011;17:1094-1100.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 396]  [Cited by in RCA: 430]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
54.  Perez D, Hauswirth F, Jäger D, Metzger U, Samartzis EP, Went P, Jungbluth A. Protein expression of cancer testis antigens predicts tumor recurrence and treatment response to imatinib in gastrointestinal stromal tumors. Int J Cancer. 2011;128:2947-2952.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 26]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
55.  Ghadban T, Perez DR, Vashist YK, Bockhorn M, Koenig AM, El Gammal AT, Izbicki JR, Metzger U, Hauswirth F, Frosina D, Jungbluth AA. Expression of cancer testis antigens CT10 (MAGE-C2) and GAGE in gastrointestinal stromal tumors. Eur J Surg Oncol. 2014;40:1307-1312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
56.  Xie F, Luo S, Liu D, Lu X, Wang M, Liu X, Jia F, Pang Y, Shen Y, Zeng C, Ma X, Tang D, Tu L, Yang L, Cheng Y, Luo Y, Xie F, Hou H, Huang T, Ni B, Zhuang C, Zhao W, Li K, Zheng X, Bi W, Jia X, He Y, Wang S, Cao H, Wu K, Wang Y. Genomic and transcriptomic landscape of human gastrointestinal stromal tumors. Nat Commun. 2024;15:9495.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
57.  Liu F, Wang Q, Li Y, Li L, Shi J, Fan X, Pan Q, Li R. KIT A502_Y503 duplication mutation serves as a potential and universal target for neoantigen peptide in Chinese GIST patients. J Gastroenterol Hepatol. 2023;38:1123-1130.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
58.  Blay JY, Kang YK, Nishida T, von Mehren M. Gastrointestinal stromal tumours. Nat Rev Dis Primers. 2021;7:22.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 72]  [Cited by in RCA: 238]  [Article Influence: 59.5]  [Reference Citation Analysis (0)]
59.  Dematteo RP, Ballman KV, Antonescu CR, Maki RG, Pisters PW, Demetri GD, Blackstein ME, Blanke CD, von Mehren M, Brennan MF, Patel S, McCarter MD, Polikoff JA, Tan BR, Owzar K; American College of Surgeons Oncology Group (ACOSOG) Intergroup Adjuvant GIST Study Team. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;373:1097-1104.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1075]  [Cited by in RCA: 964]  [Article Influence: 60.3]  [Reference Citation Analysis (1)]
60.  Fujiwara Y, Kato S, Nesline MK, Conroy JM, DePietro P, Pabla S, Kurzrock R. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treat Rev. 2022;110:102461.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 109]  [Cited by in RCA: 155]  [Article Influence: 51.7]  [Reference Citation Analysis (0)]
61.  Opitz CA, Somarribas Patterson LF, Mohapatra SR, Dewi DL, Sadik A, Platten M, Trump S. The therapeutic potential of targeting tryptophan catabolism in cancer. Br J Cancer. 2020;122:30-44.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 139]  [Cited by in RCA: 196]  [Article Influence: 39.2]  [Reference Citation Analysis (0)]
62.  Soliman HH, Jackson E, Neuger T, Dees EC, Harvey RD, Han H, Ismail-Khan R, Minton S, Vahanian NN, Link C, Sullivan DM, Antonia S. A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors. Oncotarget. 2014;5:8136-8146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 132]  [Cited by in RCA: 138]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
63.  Spranger S, Koblish HK, Horton B, Scherle PA, Newton R, Gajewski TF. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment. J Immunother Cancer. 2014;2:3.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 426]  [Cited by in RCA: 457]  [Article Influence: 41.5]  [Reference Citation Analysis (0)]
64.  Beatty GL, O'Dwyer PJ, Clark J, Shi JG, Bowman KJ, Scherle PA, Newton RC, Schaub R, Maleski J, Leopold L, Gajewski TF. First-in-Human Phase I Study of the Oral Inhibitor of Indoleamine 2,3-Dioxygenase-1 Epacadostat (INCB024360) in Patients with Advanced Solid Malignancies. Clin Cancer Res. 2017;23:3269-3276.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 193]  [Cited by in RCA: 241]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
65.  Mariotti V, Han H, Ismail-Khan R, Tang SC, Dillon P, Montero AJ, Poklepovic A, Melin S, Ibrahim NK, Kennedy E, Vahanian N, Link C, Tennant L, Schuster S, Smith C, Danciu O, Gilman P, Soliman H. Effect of Taxane Chemotherapy With or Without Indoximod in Metastatic Breast Cancer: A Randomized Clinical Trial. JAMA Oncol. 2021;7:61-69.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
66.  Mitchell TC, Hamid O, Smith DC, Bauer TM, Wasser JS, Olszanski AJ, Luke JJ, Balmanoukian AS, Schmidt EV, Zhao Y, Gong X, Maleski J, Leopold L, Gajewski TF. Epacadostat Plus Pembrolizumab in Patients With Advanced Solid Tumors: Phase I Results From a Multicenter, Open-Label Phase I/II Trial (ECHO-202/KEYNOTE-037). J Clin Oncol. 2018;36:3223-3230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 262]  [Cited by in RCA: 286]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]
67.  Labrijn AF, Janmaat ML, Reichert JM, Parren PWHI. Bispecific antibodies: a mechanistic review of the pipeline. Nat Rev Drug Discov. 2019;18:585-608.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 471]  [Cited by in RCA: 843]  [Article Influence: 140.5]  [Reference Citation Analysis (0)]
68.  Klinger M, Brandl C, Zugmaier G, Hijazi Y, Bargou RC, Topp MS, Gökbuget N, Neumann S, Goebeler M, Viardot A, Stelljes M, Brüggemann M, Hoelzer D, Degenhard E, Nagorsen D, Baeuerle PA, Wolf A, Kufer P. Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood. 2012;119:6226-6233.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 322]  [Cited by in RCA: 363]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
69.  Amann M, Brischwein K, Lutterbuese P, Parr L, Petersen L, Lorenczewski G, Krinner E, Bruckmeier S, Lippold S, Kischel R, Lutterbuese R, Kufer P, Baeuerle PA, Schlereth B. Therapeutic window of MuS110, a single-chain antibody construct bispecific for murine EpCAM and murine CD3. Cancer Res. 2008;68:143-151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 61]  [Cited by in RCA: 66]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
70.  Osada T, Patel SP, Hammond SA, Osada K, Morse MA, Lyerly HK. CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1. Cancer Immunol Immunother. 2015;64:677-688.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 70]  [Cited by in RCA: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]