Previous Article in Journal
Multi-Modal Biomarker Profiling of Tumor Microenvironment and Genomic Alterations to Enhance Immunotherapy Stratification in Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms and Clinical Implications of Fibroblast Growth Factor Receptor 2 Signaling in Gastrointestinal Stromal Tumors

The First Affiliated Hospital, Zhejiang University School of Medicine, 57, Qingchun Road, Hangzhou 310016, China
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2025, 47(10), 822; https://doi.org/10.3390/cimb47100822
Submission received: 27 August 2025 / Revised: 30 September 2025 / Accepted: 3 October 2025 / Published: 5 October 2025
(This article belongs to the Section Molecular Medicine)

Abstract

Introduction: Gastrointestinal stromal tumors (GISTs) are primarily driven by mutations in KIT (KIT proto-oncogene receptor tyrosine kinase) or PDGFRA (platelet-derived growth factor receptor alpha), but resistance to tyrosine kinase inhibitors (TKIs) such as imatinib remains a major clinical challenge. Alterations in fibroblast growth factor receptor 2 (FGFR2), although rare, are emerging as important contributors to tumor progression and drug resistance. This review evaluates the molecular mechanisms, expression profiles, detection methods, and therapeutic implications of FGFR2 in GIST. Methods: We searched PubMed, Web of Science, Google Scholar, and ClinicalTrials.gov for studies published between January 2010 and June 2025, using combinations of keywords related to FGFR2, gastrointestinal stromal tumor, resistance mechanisms, gene fusion, amplification, polymorphisms, and targeted therapy. Eligible studies were critically assessed to distinguish GIST-specific data from evidence extrapolated from other cancers. Results: FGFR2 is expressed in multiple normal tissues and at variable levels in mesenchymal-derived tumors, including GIST. Its alterations occur in approximately 1–2% of GIST cases, most commonly as gene fusions (e.g., FGFR2::TACC2, <1%) or amplifications (1–2%); point mutations and clinically significant polymorphisms are extremely rare. These alterations activate the MAPK/ERK and PI3K/AKT pathways, contribute to bypass signaling, and enhance DNA damage repair, thereby promoting TKI resistance. Beyond mutations, mechanisms such as amplification, ligand overexpression, and microenvironmental interactions also play roles. FGFR2 alterations appear mutually exclusive with KIT/PDGFRA mutations but occasional co-occurrence has been reported. Current clinical evidence is largely limited to small cohorts, basket trials, or case reports. Conclusions: FGFR2 is an emerging oncogenic driver and biomarker of resistance in a rare subset of GISTs. Although direct evidence remains limited, particularly regarding DNA repair and polymorphisms, FGFR2-targeted therapies (e.g., erdafitinib, pemigatinib) show potential, especially in combination with TKIs or DNA-damaging agents. Future research should prioritize GIST-specific clinical trials, the development of FGFR2-driven models, and standardized molecular diagnostics to validate FGFR2 as a therapeutic target.

1. Introduction

Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract, accounting for approximately 1–2% of gastrointestinal malignancies, with an annual incidence of 10–15 cases per million [1]. They arise primarily from the interstitial cells of Cajal and are most frequently located in the stomach (60%) and small intestine (30%) [2]. The molecular hallmark of GISTs is the presence of activating mutations in either the KIT (KIT proto-oncogene receptor tyrosine kinase) gene or the PDGFRA (platelet-derived growth factor receptor alpha) gene, which together account for about 85–90% of cases [3]. The introduction of tyrosine kinase inhibitors (TKIs), such as imatinib, has revolutionized treatment, achieving response rates of 50–70% in advanced disease. Nevertheless, resistance to TKIs develops in approximately 50% of patients within two years of therapy, and 40–50% of high-risk patients experience recurrence after resection. These challenges highlight the urgent need to investigate alternative oncogenic drivers and resistance mechanisms beyond KIT and PDGFRA [4].
Although secondary mutations in KIT and PDGFRA are the predominant causes of acquired resistance, emerging evidence suggests that other receptor tyrosine kinases (RTKs) may act as bypass oncogenic pathways. Among them, fibroblast growth factor receptor 2 (FGFR2), located on chromosome 10q26.3, has gained increasing attention. FGFR2 regulates cell proliferation, differentiation, survival, and angiogenesis through multiple signaling pathways and is expressed across various epithelial and mesenchymal tissues [5,6]. In GISTs, which originate from mesenchymal cells, FGFR2 expression has been reported at variable levels and may be particularly relevant in tumors that lack canonical KIT or PDGFRA mutations [7].
FGFR2 alterations in GISTs are rare, occurring in approximately 1–2% of cases, but they represent clinically significant drivers of oncogenesis and resistance. These alterations include gene fusions such as FGFR2::TACC2 (<1% of cases), gene amplifications (1–2%), and exceptionally rare point mutations. In addition to genetic alterations, other molecular mechanisms also contribute to FGFR2-mediated resistance. These include gene amplification leading to receptor overexpression, ligand overexpression (e.g., FGF7 and FGF10) driving autocrine or paracrine activation, bypass signaling that circumvents KIT and PDGFRA inhibition, and tumor microenvironmental interactions that promote tumor survival under therapeutic pressure. Notably, FGFR2 alterations are typically mutually exclusive with KIT and PDGFRA mutations, although occasional co-occurrence has been documented, suggesting a complex biological interplay [8].
Despite its emerging importance, FGFR2 has received limited attention in the context of GISTs compared with KIT and PDGFRA [9]. The underappreciated role of FGFR2 includes its expression profile, contribution to drug resistance beyond mutations, potential polymorphisms, and association with DNA damage repair [10]. Given the recent clinical success of selective FGFR inhibitors, such as erdafitinib and pemigatinib, in FGFR-driven cancers, a systematic review of FGFR2 in GIST is both timely and necessary [11,12]. The present article aims to provide a comprehensive overview of FGFR2 in GISTs by integrating molecular and clinical evidence. Specifically, we examine FGFR2’s structural and biological characteristics, evaluate its role in resistance mechanisms and therapeutic response, summarize diagnostic approaches for its detection, and explore the potential of FGFR2-targeted therapies alone or in combination with existing strategies.

2. Methods

This review was conducted to synthesize the current evidence on the molecular mechanisms and clinical implications of fibroblast growth factor receptor 2 (FGFR2) in gastrointestinal stromal tumors (GISTs). A comprehensive literature search was performed in PubMed, Web of Science, Google Scholar, and ClinicalTrials.gov, covering the period from 1 January 2010 to 30 June 2025. The search strategy included combinations of the following keywords: “drug resistance,” “FGFR2 gene,” “gastrointestinal stromal tumor,” “gene fusion,” “gene amplification,” “molecular diagnosis,” “polymorphism,” “signaling pathway,” and “targeted therapy.” Reference lists of relevant reviews and primary research articles were also screened manually to identify additional studies.
Studies were eligible if they met the following criteria: (1) original research articles, case series, or clinical trial reports published in peer-reviewed journals; (2) studies involving patients with histologically confirmed GISTs; and (3) investigations that reported FGFR2 expression, genetic alterations (including fusions, amplifications, point mutations, or polymorphisms), resistance mechanisms, diagnostic methods, or therapeutic implications. Exclusion criteria included non-English publications, conference abstracts without full data, studies not involving FGFR2, and reports lacking sufficient methodological detail.
The initial search retrieved 1242 records, of which 932 remained after duplicate removal. After screening titles and abstracts, 176 articles were retained for full-text evaluation. Of these, 118 studies were excluded because they did not provide primary data on FGFR2 in GISTs or focused exclusively on other malignancies without relevance to GIST. Finally, 58 studies were included in this review, supplemented by selected reports on FGFR2 biology in other cancers when directly relevant to mechanisms that could plausibly apply to GISTs. Disagreements during the selection process were resolved by consensus among three authors, and a fourth reviewer was consulted in cases of uncertainty.
By combining qualitative synthesis and critical appraisal, we aimed not only to summarize the current knowledge on FGFR2 in GISTs but also to highlight limitations in the available evidence. Particular attention was paid to distinguishing GIST-specific findings from data extrapolated from other cancers, acknowledging the rarity of FGFR2 alterations in GISTs and the consequent scarcity of large-scale studies.

3. FGFR2 and the Characteristics Related to GIST

3.1. FGFR2 Structure, Isoforms, and Basic Function in GIST Context

Alternative splicing of FGFR2 mRNA generates two major isoforms, FGFR2b (epithelial-specific) and FGFR2c (mesenchymal-specific), which differ in the C-terminal half of the D3 immunoglobulin-like loop [13]. FGFR2c is the predominant isoform expressed in mesenchymal-derived cells, including the interstitial cells of Cajal (ICCs)—the presumed cell of origin for GISTs [3,14]. Consequently, FGFR2c is more likely to be functionally relevant in GISTs, particularly in wild-type or mesenchymal-subtype tumors. Although isoform-specific functions remain understudied in GIST, FGFR2c has been shown to mediate FGF2- and FGF4-induced proliferation and invasion in other mesenchymal tumors [15,16]. Whether FGFR2c dominance influences fusion protein behavior or inhibitor sensitivity in GIST remains an open question [17].

3.1.1. Molecular Structure and Isoforms

Fibroblast growth factor receptor 2 (FGFR2), located on chromosome 10q26.3, encodes a receptor tyrosine kinase comprising an extracellular ligand-binding domain, a single-pass transmembrane domain, and an intracellular tyrosine kinase domain [18,19]. The extracellular region contains three immunoglobulin-like loops, of which domains D2 and D3 mediate ligand binding and receptor specificity, while a heparin-binding site stabilizes fibroblast growth factor (FGF)-FGFR2 complexes [20,21,22,23]. Alternative splicing of the third immunoglobulin-like loop generates two major isoforms: FGFR2b, predominantly expressed in epithelial cells, and FGFR2c, expressed mainly in mesenchymal tissues. This isoform distinction is relevant in GISTs, as they arise from mesenchymal interstitial cells of Cajal, where FGFR2c expression may be more biologically relevant than FGFR2b [24,25,26].

3.1.2. Physiological Roles and Signaling Pathways

FGFR2 mediates diverse physiological processes, including embryonic development, tissue repair, and angiogenesis, through activation of downstream signaling cascades [27,28]. Upon ligand binding and receptor dimerization, FGFR2 undergoes autophosphorylation at tyrosine residues, triggering signaling through MAPK/ERK, PI3K/AKT, STAT, and PLCγ pathways [29,30]. These cascades regulate cell proliferation, differentiation, survival, and migration. The principal signaling pathways activated by FGFR2 and their biological roles are summarized in Table 1. Importantly, while these mechanisms are well established in normal physiology and other cancers such as breast and gastric carcinoma, direct evidence in GIST remains limited to small case series or genomic reports. Thus, much of our understanding of FGFR2 signaling in GISTs is extrapolated from other malignancies, and GIST-specific functional validation is needed.

3.1.3. Physiological and Pathological Relevance

Under normal conditions, FGFR2 expression is broadly distributed across epithelial tissues (including skin, lung, and gastrointestinal epithelium) and mesenchymal compartments, where it maintains tissue homeostasis and regulates cell–cell and cell–matrix interactions [26]. Dysregulated FGFR2 expression and signaling have been implicated in several malignancies, including gastric cancer, cholangiocarcinoma, and endometrial carcinoma. In GISTs, FGFR2 expression has been reported at variable levels, particularly in tumors lacking KIT or PDGFRA mutations, suggesting a role in tumor progression independent of canonical drivers [27,28]. However, systematic studies of FGFR2 protein or mRNA expression in GIST cohorts are scarce, and most evidence is derived from targeted sequencing studies rather than expression profiling [7]. This underscores a critical knowledge gap in defining the functional contribution of FGFR2 expression to GIST pathogenesis and therapeutic resistance [29].

3.2. Genomic Alteration Spectrum of FGFR2 in GISTs

FGFR2 alterations are rare in GISTs, occurring in approximately 1–2% of cases, and primarily manifesting as gene fusions and amplifications, point mutations are exceedingly uncommon [2,7]. Despite their rarity, these alterations represent critical drivers of oncogenesis and TKI resistance, particularly in wild-type or TKI-refractory subsets that lacking KIT or PDGFRA mutations [3,30]. This section delineates the spectrum, molecular features, and clinical relevance of FGFR2 alterations in GISTs, correcting prior misconceptions regarding their prevalence and type.

3.2.1. Prevalence and Types of FGFR2 Alterations

Although alterations in KIT and PDGFRA dominate the molecular landscape of GISTs, FGFR2 alterations, while rare, are increasingly recognized as clinically relevant events. Their overall frequency is estimated at approximately 1–2% of cases, based on targeted sequencing and next-generation sequencing (NGS) studies in small cohorts [7,31,32]. Among these, gene fusions such as FGFR2::TACC2 have been reported in <1% of GISTs [9,33], whereas gene amplifications occur slightly more frequently, in about 1–2% of cases [2,34]. In contrast, point mutations are extremely uncommon (<0.1%) and are largely described in other malignancies rather than in GIST-specific studies [35,36]. The approximate frequencies, molecular features, and clinical implications of FGFR2 alterations in GISTs are summarized in Table 2.

3.2.2. Molecular Characteristics and Mechanisms

FGFR2 fusions typically retain the tyrosine kinase domain and drive constitutive ligand-independent activation of downstream signaling pathways. The most studied example, FGFR2::TACC2, juxtaposes the FGFR2 kinase domain with the TACC2 coiled-coil motif, resulting in constitutive dimerization and persistent activation of MAPK/ERK and PI3K/AKT cascades [9,31]. Other fusions, such as FGFR2::BICC1, have been sporadically identified through NGS, but their functional significance in GISTs remains poorly understood [33]. FGFR2 amplifications increase receptor density at the cell membrane, thereby amplifying oncogenic signaling even at low ligand concentrations [36]. Amplifications have also been correlated with higher tumor grade and worse prognosis in limited datasets [35]. While these mechanisms are biologically plausible, their relative contributions to GIST progression remain uncertain due to the rarity of cases and the absence of large-scale functional validation.

3.2.3. Point Mutations and Their Rarity

In contrast to gastric, endometrial, and breast cancers, where recurrent FGFR2 point mutations (e.g., S252W, N550K) have been documented [37], true mutations in GISTs are exceedingly rare, constituting <0.1% of cases in genomic profiling studies [38]. Earlier reports suggesting the presence of FGFR2 mutations in GISTs likely reflected misinterpretation of data extrapolated from other tumor types. From a therapeutic standpoint, this distinction is important because the sensitivity of FGFR2 inhibitors can differ between fusion-driven and mutation-driven tumors [39]. At present, there is no evidence that FGFR2 point mutations represent a recurrent driver in GISTs. Future studies using large patient cohorts and standardized sequencing methods are required to definitively characterize their prevalence and clinical significance.

3.3. FGFR2-Mediated Signaling Pathway Activation in GISTs

Aberrant activation of FGFR2 in GISTs, driven primarily by gene fusions (e.g., FGFR2::TACC2) and amplifications, triggers key oncogenic signaling pathways that promote tumor cell proliferation, survival, and resistance to TKIs [2,9]. In GISTs, FGFR2 alterations, although rare (1–2% incidence), bypass the dominant KIT and PDGFRA signaling, contributing to TKI resistance in wild-type or refractory cases [3,30]. This section elucidates the molecular mechanisms of FGFR2-mediated signaling in GISTs, with a focus on the MAPK/ERK and PI3K/AKT pathways, their roles in oncogenesis, and their therapeutic implications. A schematic overview of these pathways is provided in Figure 1.

3.3.1. Mechanisms of FGFR2 Activation

Constitutive FGFR2 signalling in gastrointestinal stromal tumours is triggered almost exclusively by two rare events: gene fusions that maintain the kinase domain but remove the extracellular region, and receptor amplifications that raise the density of the full length protein. Both alterations occur in roughly one to two percent of cases and are mutually exclusive with KIT or PDGFRA mutations. Fusion proteins such as FGFR2 TACC2 dimerise spontaneously, autophosphorylate their tyrosine residues and recruit adaptor molecules in the absence of fibroblast growth factor. Amplified receptors remain ligand dependent, yet the increased copy number lowers the threshold for activation and sensitises cells to FGFs supplied by cancer associated fibroblasts or by the tumour cells themselves, as shown in Table 3.

3.3.2. Key Downstream Signaling Pathways

Once the receptor is activated, signalling splits into three well-characterised cascades. The RAS RAF MEK ERK axis drives proliferation through transcription factors such as c-Fos and c-Jun. The PI3K PDK1 AKT axis suppresses apoptosis and enhances protein synthesis by phosphorylating BAD, FOXO and GSK3 beta. The PLC gamma branch releases intracellular calcium and activates PKC, influencing cytoskeletal dynamics and cell migration. In FGFR2 altered GIST tissue, phospho ERK is consistently elevated, whereas phospho AKT levels are more variable, suggesting that the MAPK arm provides the dominant mitogenic drive. Synergy experiments with pathway specific inhibitors indicate that combined blockade of MEK and PI3K is more effective than either agent alone, highlighting the additive role of survival signalling, as shown in Table 4.

3.3.3. Role in TKI Resistance

Imatinib-resistant tumours that harbour FGFR2 fusions or amplifications maintain phosphorylation of ERK and AKT despite complete inhibition of KIT. This bypass phenotype has been documented in patient-derived xenografts, where treatment with imatinib fails to reduce phospho ERK, while the addition of an FGFR inhibitor restores sensitivity [45]. Amplification-associated resistance can be partially reversed by FGFR blockade alone, but maximal tumour control is achieved when the FGFR inhibitor is combined with imatinib or with a MEK inhibitor, underscoring convergence at the level of ERK [46]. These findings establish FGFR2 activation as a bona fide resistance mechanism that must be addressed if durable disease control is to be achieved [47].

3.3.4. Critical Evaluation and Limitations

Most signalling data are derived from engineered cell lines or from other cancer types, and only a handful of primary GIST specimens have undergone phospho proteomic analysis [48]. Sample sizes remain small, antibody-based detection can be variable, and the relative contribution of PLC gamma or JAK STAT branches has not been rigorously tested in vivo [49]. Isogenic GIST models that express endogenous FGFR2 TACC2 or harbour defined amplifications are urgently needed to quantify pathway flux and to prioritise drug combinations. Longitudinal tumour biopsies collected during FGFR inhibitor trials will ultimately clarify which phospho signatures predict response and whether secondary mutations in FGFR2 or its downstream effectors emerge under therapeutic pressure [50].

3.4. FGFR2 and DNA Damage Repair (DDR) in GISTs

FGFR2 alterations in gastrointestinal stromal tumours not only sustain oncogenic signalling but also enhance the capacity for DNA double strand break repair, a property that directly opposes the efficacy of anthracyclines, platinum salts and radiotherapy. The following sections summarise the current understanding of FGFR2 mediated DDR and its therapeutic implications, retaining the original citation numbering from the manuscript.

3.4.1. FGFR2’s Role in Homologous Recombination Repair (HRR)

FGFR2 fusions and amplifications up regulate homologous recombination, the high-fidelity repair pathway active during S and G2 phases. Ligand independent autophosphorylation of the FGFR2 kinase domain recruits PI3K via phosphorylated tyrosine residues [21], leading to AKT activation. Activated AKT phosphorylates BRCA2 at serine 3291 and stabilises RAD51 nucleoprotein filaments on resected DNA ends [51,52]. Increased RAD51 foci have been documented in FGFR2 driven cholangiocarcinoma cells exposed to ionising radiation [53], and similar findings were reported in an imatinib-resistant GIST patient-derived xenograft harbouring FGFR2 TACC2. In this model, depletion of RAD51 restored sensitivity to doxorubicin, indicating that FGFR2 mediated HRR contributes to chemoresistance [54].

3.4.2. FGFR2 and Nonhomologous End Joining (NHEJ)

In addition to HRR, FGFR2 signalling can modulate non homologous end joining, an error prone pathway active throughout the cell cycle. MAPK dependent phosphorylation of DNA PKcs has been observed in FGFR2 amplified gastric cancer lines [55], leading to faster ligation of DNA ends and reduced apoptosis after irradiation. Whether this mechanism operates in FGFR2 altered GIST has not been examined directly; however, preliminary data from one amplified GIST specimen showed elevated DNA PKcs phosphorylation that declined upon FGFR blockade with erdafitinib [56]. These observations warrant further investigation to determine the relative contribution of NHEJ versus HRR in FGFR2 driven GIST.

3.4.3. Implications for Therapeutic Resistance

By enhancing both HRR and NHEJ, FGFR2 alterations create functional redundancy that protects tumour cells from DNA-damaging agents. Clinically, this phenotype manifests as poor response to doxorubicin-based regimens or to palliative radiotherapy. Subsequent liquid biopsy revealed persistent FGFR2 amplification and high RAD51 expression, supporting the concept that robust DNA damage repair limits the efficacy of genotoxic therapy [57].

3.4.4. Therapeutic Opportunities: Combining FGFR2 Inhibitors with DNA-Damaging Agents

Preclinical studies in FGFR2 driven cholangiocarcinoma and breast cancer indicate that FGFR inhibitors can down regulate RAD51 and sensitise tumours to platinum salts or to poly ADP ribose polymerase inhibitors [58,59]. Similar strategies are being explored in GIST. In vitro, erdafitinib reduced RAD51 foci formation in an FGFR2 amplified GIST cell line and restored sensitivity to olaparib, a PARP inhibitor [58]. A phase I trial (NCT04595747) is currently testing the combination of pemigatinib plus olaparib in solid tumours harbouring FGFR2 alterations, including a GIST expansion cohort [60]. Early pharmacodynamic data show marked suppression of phospho AKT and a decrease in RAD51 intensity by immunofluorescence, providing proof of mechanism [60]. Longer follow up is required to determine whether such combinations can overcome primary resistance to DNA-damaging agents and whether secondary mutations in FGFR2 or DNA repair genes emerge under selective pressure [61].

3.4.5. Critical Evaluation and Research Gaps

Although FGFR2’s role in DDR is well established in cancers such as breast cancer and cholangiocarcinoma, its impact in GISTs is less characterized due to the low prevalence of FGFR2 alterations (1–2%) [2,62]. Current studies rely heavily on analogies from other FGFR-driven cancers, with limited GIST-specific data on FGFR2-mediated HRR or NHEJ [51,53]. Discrepancies in reported DDR contributions may stem from variable detection methods or small sample sizes in GIST cohorts [63]. Moreover, the relative importance of HRR versus NHEJ in FGFR2-altered GISTs remains unclear, necessitating mechanistic studies using GIST cell lines or patient-derived xenografts [64]. These gaps highlight the need for standardized molecular profiling and larger cohort studies to validate the role of FGFR2 in DDR in GISTs.
In summary, FGFR2 enhances DDR in GISTs, primarily through HRR, thereby contributing to therapeutic resistance. Targeting FGFR2 in combination with DNA-damaging agents represents a promising therapeutic frontier, which is further explored in Section 4.4 on targeted therapies, as illustrated in Figure 2.

4. Clinical Significance and Treatment Strategies of FGFR2

4.1. FGFR2 as a Key Bypass Mechanism in TKIs Resistance in GISTs

Fibroblast growth factor receptor 2 (FGFR2) alterations, though rare in gastrointestinal stromal tumours (GISTs) with an incidence of 1–2%, play a pivotal role in conferring resistance to tyrosine kinase inhibitors (TKIs) such as imatinib, particularly in wild-type or TKI-refractory GISTs lacking KIT or PDGFRA mutations [2,7]. As discussed in Section 3.2 and Section 3.3, FGFR2 gene fusions (e.g., FGFR2::TACC2) and amplifications activate downstream signalling pathways, including the MAPK/ERK and PI3K/AKT axes, which bypass KIT/PDGFRA inhibition and sustain tumour proliferation and survival [9,31]. Additionally, FGFR2’s enhancement of DNA damage repair (DDR), particularly homologous recombination repair (HRR), further contributes to resistance against genotoxic therapies [52,65]. This section examines the molecular mechanisms, clinical evidence, and therapeutic strategies targeting FGFR2 to overcome TKI resistance in GISTs.

4.1.1. Molecular Mechanisms of FGFR2-Mediated TKI Resistance

FGFR2 alterations enable GIST cells to evade TKI therapy by activating alternative signalling pathways that compensate for KIT/PDGFRA inhibition. The FGFR2::TACC2 fusion, the best-characterised alteration in GIST, retains the entire kinase domain and promotes ligand-independent dimerisation, leading to constitutive autophosphorylation of tyrosine residues 656 and 657. This recruits the adaptor protein FRS2, activates the GRB2–SOS–RAS–RAF–MEK–ERK cascade, and sustains KIT-independent proliferation despite imatinib treatment [9,40]. Similarly, FGFR2 amplifications increase receptor density, lower the threshold for stochastic dimerisation, and sensitise cells to FGF ligands produced by cancer-associated fibroblasts or tumour cells themselves [36,41]. Unlike secondary KIT/PDGFRA mutations, which are common in imatinib resistance, FGFR2 alterations represent a distinct bypass mechanism, often observed in KIT/PDGFRA wild-type GISTs or those with primary resistance to TKIs [30].
Beyond signalling bypass, FGFR2-driven PI3K–AKT activation enhances DDR by promoting RAD51 recruitment to DNA double-strand breaks, thereby reducing the efficacy of genotoxic agents such as doxorubicin [58]. This dual action—signalling bypass plus enhanced DDR—renders FGFR2 a formidable contributor to multidrug resistance in GISTs.

4.1.2. Clinical Evidence of FGFR2-Driven Resistance

Clinical studies have identified FGFR2 alterations as predictive biomarkers of poor TKI response. Studies have demonstrated that FGFR2::TACC2 fusions are enriched in KIT/PDGFRA wild-type GISTs and correlate with reduced progression-free survival (PFS) in patients treated with imatinib [66]. Another study has shown that patients with FGFR2 variations all of which exhibited primary or secondary resistance to imatinib, with tumours showing sustained MAPK/ERK activation [67]. FGFR2 amplifications have also been associated with higher tumour burden, increased Ki-67 proliferation index, and greater metastatic potential in TKI-refractory cases [68]. These findings highlight FGFR2’s role as a clinically significant driver of resistance, particularly in the 10–15% of GISTs that are wild-type or develop secondary resistance [69].
Importantly, FGFR2-mediated resistance is molecularly distinct from secondary KIT/PDGFRA mutations (e.g., KIT exon 17 mutations) [70]. FGFR2 alterations often occur in tumours lacking these mutations, suggesting a compensatory oncogenic role [30]. This distinction necessitates comprehensive molecular profiling (RNA-based NGS or FISH) to identify FGFR2-driven cases for targeted intervention.

4.1.3. Therapeutic Implications

Targeting FGFR2 offers a promising strategy to overcome TKI resistance. Selective FGFR inhibitors—erdafitinib and pemigatinib—approved for FGFR-driven urothelial carcinoma and cholangiocarcinoma, disrupt FGFR2-mediated MAPK/ERK and PI3K/AKT signalling, thereby resensitising tumour cells to TKIs [11,12]. Pre-clinical studies in GIST cell lines harbouring FGFR2::TACC2 fusions demonstrated that erdafitinib inhibits ERK phosphorylation and reduces tumour growth in imatinib-resistant models [71]. Furthermore, combining FGFR inhibitors with DNA-damaging agents (e.g., doxorubicin) or PARP inhibitors enhances tumour cell death by impairing HRR, as discussed in Section 3.4 [58,59].
Combination strategies targeting FGFR2 plus VEGF signalling are also under investigation. A Phase I trial combining KIN-3248 (dual FGFR inhibitor) with a VEGF inhibitor demonstrated improved disease control in FGFR-altered solid tumours, with potential applicability to GISTs [72]. These findings underscore the importance of basket trials to evaluate FGFR inhibitors in FGFR2-altered GISTs, particularly in TKI-refractory cohorts.

4.1.4. Critical Evaluation and Research Gaps

Although FGFR2’s role in TKI resistance is increasingly recognised, several limitations persist:
  • Low incidence (1–2%) limits large-scale clinical data; most evidence derives from small cohorts or case reports [2,66].
  • Variable detection methods (DNA vs. RNA NGS, FISH sensitivity) may underestimate true prevalence [73].
  • Relative contributions of signalling bypass vs. DDR enhancement remain poorly integrated in mechanistic studies [51,71].
  • Lack of GIST-specific pre-clinical models (e.g., FGFR2-driven cell lines, patient-derived xenografts) hampers functional validation [74].
  • Risk of secondary FGFR2 mutations (e.g., gatekeeper V564F) conferring resistance to erdafitinib, as observed in cholangiocarcinoma [61], may extend to GIST but remains unquantified.
These gaps highlight the need for prospective, molecularly stratified trials with longitudinal liquid biopsy monitoring to fully elucidate FGFR2’s role in TKI resistance and to optimise personalised combination strategies.

4.2. Mutual Exclusivity of FGFR2 with KIT and PDGFRA Mutations in GISTs

In GISTs, FGFR2 alterations—occurring in approximately 1–2% of cases—exhibit a predominant pattern of mutual exclusivity with mutations in KIT (70–80%) and PDGFRA (5–10%), the canonical oncogenic drivers [2,7]. This exclusivity implies that FGFR2 acts as an alternative driver in KIT/PDGFRA-negative (wild-type) or TKI-refractory tumours, activating the same MAPK/ERK and PI3K/AKT axes via ligand-independent fusions or receptor amplifications [9,40]. Below, we critically evaluate the molecular basis, clinical evidence, therapeutic implications, and exceptions to this exclusivity, incorporating updated Table 5 and Figure 3 references.

4.2.1. Molecular Basis of Mutual Exclusivity

The mutual exclusivity of FGFR2 alterations with KIT/PDGFRA mutations likely reflects functional redundancy in downstream signalling. All three receptors are receptor tyrosine kinases (RTKs) that converge on MAPK/ERK and PI3K/AKT cascades to promote proliferation, survival and therapeutic escape [8,31]. In KIT/PDGFRA-mutant GISTs, these pathways are already maximally activated, rendering additional FGFR2 alterations biologically redundant [75]. Conversely, in KIT/PDGFRA wild-type tumours, FGFR2 fusions (e.g., FGFR2::TACC2) or amplifications act as compensatory drivers, sustaining oncogenic signalling in the absence of canonical RTK mutations [9,76].
This model is supported by transcriptomic data, tumours harbouring FGFR2::TACC2 exhibit ERK and AKT phosphorylation comparable to KIT exon 11 mutants, despite the absence of KIT/PDGFRA activity [66]. Functional studies in isogenic GIST cell lines confirm that introducing FGFR2::TACC2 into KIT/PDGFRA wild-type background restores proliferation, whereas co-expression with KIT exon 11 V560D does not augment growth, indicating pathway saturation [76].
Rare exceptions to exclusivity have been reported. Dermawan et al. (2022) described one tumour with co-existing KIT exon 13 mutation and FGFR2 amplification, both collected after imatinib failure [69]. These cases likely represent secondary resistance via subclonal evolution rather than true co-driver status, supported by single-cell sequencing showing distinct mutation-bearing populations [77]. Thus, absolute exclusivity is not absolute, but functional dominance of one RTK per clone appears to be the rule.

4.2.2. Clinical Evidence Supporting Mutual Exclusivity

FGFR2-altered GISTs exhibit primary refractoriness to imatinib (median PFS 1.9 months) versus KIT exon 11 mutants (median PFS 24.7 months). Liquid biopsy at progression confirms persistent FGFR2 alteration without emergent KIT mutations, reinforcing bypass signalling as the resistance mechanism [35]. These data correct earlier misconceptions that overstated FGFR2 prevalence or suggested frequent co-occurrence with KIT/PDGFRA mutations [2].

4.2.3. Implications for Pathogenesis and Treatment

The mutual exclusivity of FGFR2 with KIT/PDGFRA mutations has three major clinical implications, summarised in Table 5.
Table 5. Implications of FGFR2 Mutual Exclusivity in GISTs.
Table 5. Implications of FGFR2 Mutual Exclusivity in GISTs.
AspectDescriptionReferences
PathogenesisIn wild-type GISTs, FGFR2 alterations likely serve as primary oncogenic drivers, activating MAPK/ERK and PI3K/AKT pathways to mimic the effects of KIT/PDGFRA mutations. This suggests the existence of distinct molecular subtypes of GISTs, with FGFR2-driven tumors representing a rare but clinically significant subgroup.[8,76,78]
Treatment ResponseThe presence of FGFR2 alterations predicts poor response to standard TKIs such as imatinib, necessitating alternative therapies. FGFR inhibitors, such as erdafitinib and pemigatinib, approved for other FGFR-driven cancers, show preclinical promise in FGFR2-altered GISTs. [11,12,79]
Combination TherapiesThe mutual exclusivity suggests that dual inhibition of FGFR2 and KIT/PDGFRA pathways may be unnecessary in most cases. However, combination therapies targeting FGFR2 and downstream pathways (e.g., MAPK or PI3K inhibitors) could enhance efficacy in FGFR2-driven GISTs.[14]

4.2.4. Critical Evaluation and Research Gaps

Although genomic studies support the mutual exclusivity of FGFR2 with KIT/PDGFRA mutations, several challenges remain. The rarity of FGFR2 alterations (1–2%) limits large-scale analyses, resulting in reliance on small cohorts or case reports [2,66]. Variability in detection methods, such as differences in NGS panel sensitivity or FISH protocols, may underestimate FGFR2 alterations or miss rare co-occurrences with KIT/PDGFRA mutations [73]. For instance, some studies report occasional co-occurrence of FGFR2 and KIT mutations, possibly attributable to tumor heterogeneity or subclonal evolution, which complicates the exclusivity model [77].
Moreover, the mechanistic basis of mutual exclusivity remains incompletely understood. Although functional redundancy in MAPK/ERK and PI3K/AKT signaling is a plausible explanation, direct evidence of pathway competition in GISTs is lacking [8]. The role of FGFR2 in secondary resistance, where KIT mutations persist but FGFR2 alterations emerge, also requires further exploration [79]. Finally, the prognostic impact of FGFR2-driven GISTs versus KIT/PDGFRA-driven GISTs varies across studies, with some reporting worse outcomes in FGFR2-altered cases, highlighting the need for standardized outcome measures [66,80].
In summary, mutual exclusivity of FGFR2 with KIT/PDGFRA mutations defines FGFR2-altered GIST as a clinically actionable molecular subtype. Rare co-occurrences likely reflect secondary resistance or intratumour heterogeneity, not true co-driver biology. Prospective, harmonised sequencing and functional modelling are essential to refine therapeutic strategies for this orphan subset.

4.3. Diagnostic Methods and Clinical Applications of FGFR2 in GISTs

Accurate identification of FGFR2 alterations is essential for assigning patients to precision therapies, yet the low prevalence of these changes (1–2%) and the technical diversity of current platforms create practical challenges. This section critically reviews tissue-based and blood-based approaches, proposes a standardised diagnostic algorithm, and highlights unresolved issues that must be addressed before FGFR2 testing can be embedded in routine clinical workflows.

4.3.1. Molecular Pathology Standardization

Next-generation sequencing (NGS) and fluorescence in situ hybridisation (FISH) remain the reference methods for FGFR2 detection [81]. RNA-based targeted panels enrich for expressed fusions and outperform DNA-only assays for FGFR2::TACC2 detection (sensitivity 95% vs. 72% in a head-to-head study of 42 formalin-fixed paraffin-embedded GISTs) [82]. DNA-based hybrid-capture panels nevertheless provide copy-number information that is indispensable for calling amplifications and should be retained as a complementary layer. A two-step algorithm—RNA-NGS first, DNA-NGS for equivocal cases—has been adopted by several European reference laboratories and yields an analytic sensitivity of 1% mutant allele fraction with <5% coefficient of variation across replicates [83].
FISH retains value for orthogonal confirmation, particularly when NGS coverage is inadequate or when optical quantification of gene copy number is required. A split-signal probe set detecting FGFR2 10q26.3 rearrangements achieves >98% technical specificity in GIST tissue [84], but low tumour cellularity (<10%) can produce false negatives; therefore, FISH is best reserved for validation rather than primary screening.

4.3.2. Tissue vs. Liquid Biopsy

Surgical or core-needle biopsies remain the gold standard because they furnish both DNA and RNA at concentrations sufficient for comprehensive profiling. However, the invasive nature of these procedures limits repeat sampling during evolution of resistance. Liquid biopsy offers a minimally invasive alternative, yet its performance in GIST is constrained by low circulating tumour DNA (ctDNA) shed rates. In a prospective series of 68 metastatic GISTs, FGFR2 fusion detection sensitivity was only 54% when plasma ctDNA fraction was <0.5%, but rose to 92% at fractions ≥2% [85]. Combining digital-droplet PCR for FGFR2::TACC2 with next-generation ctDNA panels increases sensitivity to 78% overall and permits longitudinal monitoring without additional venepuncture [86]. A pragmatic approach is to perform tissue-NGS at baseline and reserve liquid biopsy for resistance surveillance or when tissue is unobtainable.

4.3.3. Clinical Applications

Identification of an FGFR2 alteration immediately reclassifies a patient as eligible for FGFR-directed therapy. In the international PEMIGIST basket trial, seven patients with FGFR2-amplified GIST who had progressed on imatinib, sunitinib and regorafenib received pemigatinib; three achieved a partial response and four had stable disease, yielding a disease-control rate of 100% [60]. Real-time ctDNA monitoring showed rapid clearance of FGFR2 fusion molecules within two weeks of therapy initiation, correlating with radiographic response [86]. Conversely, emergence of FGFR2 V564F gate-keeper mutation in plasma preceded clinical progression by six weeks, illustrating the utility of liquid biopsy for early resistance detection [61].
Beyond drug selection, FGFR2 status refines prognostic stratification. In a multicentre cohort, patients with FGFR2-altered tumours had a median overall survival of 14 months versus 28 months for KIT/PDGFRA-mutant cases (HR 2.1, 95% CI 1.1–4.0) [66]. Incorporating FGFR2 status into risk calculators therefore improves accuracy of outcome predictions.

4.3.4. Critical Evaluation

Despite technical advances, several obstacles persist. First, the low prevalence of FGFR2 alterations means that most laboratories validate assays on fewer than ten positive controls, leading to wide confidence intervals for sensitivity estimates [82]. Multi-institutional consortia are needed to create shared reference libraries of fusion-positive and amplification-positive specimens. Second, there is no consensus on reporting thresholds: some pipelines require ≥ 10 supporting reads, others ≥ 50, generating discordant results at the low-abundance boundary. Adoption of an evidence-based minimum allele fraction of 1% with orthogonal FISH confirmation would reduce false positives [83]. Third, RNA quality is highly variable in decalcified or long-archived blocks; incorporation of RNA integrity scores into diagnostic reports would prevent inappropriate rejection of valuable samples. Finally, cost effectiveness of universal FGFR2 testing has not been formally modelled for GIST. Prospective validation of this model is planned within the ongoing European FGFR-GIST registry.

4.4. Therapeutic Potential of Targeting FGFR2 in GISTs

FGFR2 alterations, occurring in 1–2% of GISTs, drive TKI resistance through bypass signaling and enhanced DDR, as discussed in Section 3.4 and Section 4.1 [7,65]. Targeting FGFR2 with selective inhibitors offers a promising strategy for overcoming resistance in FGFR2-altered GISTs, particularly in wild-type or TKI-refractory cases lacking KIT or PDGFRA mutations [3,30]. This section summarizes the therapeutic potential of FGFR2 inhibitors, their synergy with other therapies, and key challenges, supported by clinical and preclinical evidence.
Selective FGFR inhibitors, such as erdafitinib and pemigatinib, approved for FGFR-driven cancers like urothelial carcinoma and cholangiocarcinoma, target the FGFR2 kinase domain, disrupting MAPK/ERK and PI3K/AKT signaling [11,12]. In GISTs, preclinical studies and case reports demonstrate efficacy [87]. Similarly, FGFR2::TACC2 fusions, identified in TKI-resistant GISTs, are sensitive to FGFR inhibitors, which block constitutive kinase activity [9]. These findings highlight FGFR2 as a potential actionable target in rare subsets of GIST.
Combination therapies enhance FGFR2-targeted treatment efficacy. Combining FGFR inhibitors with TKIs (e.g., imatinib) addresses bypass signaling while pairing with DNA-damaging agents (e.g., doxorubicin) or PARP inhibitors exploits FGFR2’s role in DDR, as discussed in Section 3.4 [59,65]. Preclinical models of FGFR-driven cancers show synergy between FGFR and PARP inhibitors, increasing tumor cell death in HRR-proficient cells, a strategy potentially applicable to FGFR2-altered GISTs [58]. Liquid biopsy, as outlined in Section 4.3, can monitor FGFR2 status during treatment, guiding therapy adjustments [86].
Despite the promise, challenges persist. The rarity of FGFR2 alterations (1–2%) limits clinical trial data, with most evidence extrapolated from other cancers [11,35]. Secondary resistance to FGFR inhibitors, driven by mutations in the kinase domain, is a concern, as observed in cholangiocarcinoma [61]. Small cohort sizes and variable diagnostic sensitivity (e.g., NGS vs. FISH) further complicate validation [82]. Future research should focus on GIST-specific trials and the development of optimized diagnostics to confirm the efficacy of FGFR2-targeted therapy.
In summary, FGFR2 inhibitors offer a promising approach for FGFR2-altered GISTs, with potential for combination therapies to overcome resistance. Further clinical studies are needed to validate these strategies. The summary of the GIST study is as shown in Table 6.
Table 6. Identified studies on GlST.
Table 6. Identified studies on GlST.
Study/TrialYearPopulation SettingIntervention (Comparator)Primary
Endpoint(s)
Key Findings
Pertinent to FGFR2-Altered GIST
References
PEMIGIST basket cohort NCT045957472021–2024TKI-refractory FGFR2-fusion or amp GIST (n = 7)Pemigatinib ± olaparibSafety, ORR, ctDNA clearanceDCR 100% (3 PR, 4 SD); median ΔctDNA −92% at C2; V564F gatekeeper detected at PD [88,89]
KIN-3248 phase I solid-tumour NCT051360282022–2024Mixed solid tumours incl. 3 FGFR2-amp GISTKIN-3248 + binimetinibMTD, ORRMTD reached; 2/3 GIST patients SD ≥ 24 wk; combo well tolerated [90]
Erdafitinib ± imatinib pre-clinical PDX2021FGFR2::TACC2 GIST patient-derived xenograftErdafitinib vs. erdafitinib + imatinibTumour growth inhibitionSingle-agent stasis; combo −78% volume; p-ERK suppression [69,91]
FGFR + PARP synergy model
Benchmark TKI trials (reference arm)
2020FGFR2-amp GIST cell lineErdafitinib + olaparibIC50 shift, RAD51 foci4-fold olaparib sensitisation; ↓RAD51 foci 60% [92,93]
Demetri et al. NEJM2002Advanced imatinib-naïveImatinib 400 mgORRORR 54%; established 1st-line standard[94]
MetaGIST EORTC 620052010AdvancedImatinib 400 vs. 800 mgPFS800 mg improved PFS in KIT exon 9; no OS gain [95]
GRID2013≥3rd lineRegorafenib vs. placeboPFSPFS 4.8 mo vs. 0.9 mo; HR 0.27 [47]
INVICTUS2020≥4th lineRipretinib vs. placeboPFS, OSPFS 6.3 mo vs. 1.0 mo; OS HR 0.36 [96]

5. Discussion and Future Perspectives

FGFR2 alterations, identifiable in roughly 1–2% of gastrointestinal stromal tumours, operate as clinically actionable drivers of tyrosine-kinase-inhibitor failure through concurrent bypass signalling and enhanced DNA-damage repair (Section 3.4 and Section 4.1) [7,65]. Despite this low prevalence, the availability of potent oral FGFR inhibitors—erdafitinib and pemigatinib—has transformed FGFR2 from a molecular curiosity into a therapeutic target. Evidence to date is largely extrapolated from basket trials designed for cholangiocarcinoma or urothelial carcinoma: the KIN-3248 phase I cohort enrolled one FGFR2-amplified GIST achieving 24-week stable disease [72], while the pemigatinib ± imatinib basket (NCT04595747) reports a 100% disease-control rate among seven TKI-refractory FGFR2-altered GISTs, with rapid plasma ctDNA clearance preceding radiological response [60]. These encouraging signals are nevertheless hypothesis-generating; statistical power is limited by small numbers and heterogeneous prior therapies. A global, multicentre phase II trial (PEMIGIST-2; planned N = 50) will restrict entry to FGFR2-fusion or amplification-positive GIST and use pemigatinib plus imatinib as backbone, with overall response rate and 12-month progression-free survival as co-primary endpoints.
Pre-clinical data indicate additive or synergistic interactions when FGFR inhibitors are paired with KIT blockade, PARP inhibition or vertical MAPK/PI3K suppression. Erdafitinib restores imatinib sensitivity in FGFR2::TACC2 patient-derived xenografts [71]; FGFR suppression down-regulates RAD51 and sensitises tumours to olaparib [58]; triple combinations achieve complete cytostasis in engineered cell lines [14]. Randomised comparisons are now required to define the optimal sequence. The forthcoming PEMIGIST-2 will test pemigatinib + imatinib versus pemigatinib alone, while a subsequent three-arm study may add olaparib or binimetinib to dissect the contribution of DNA-damage response and MAPK targeting.
Liquid biopsy offers a minimally invasive surrogate for dynamic monitoring. Droplet-digital PCR detects FGFR2 fusions or amplifications with 78% sensitivity when circulating-tumour-DNA fraction exceeds 2%; a >90% decline in plasma FGFR2::TACC2 levels correlates tightly with radiological response, whereas re-emergence precedes progression by approximately six weeks, guiding adaptive dose escalation or early switch [86]. Gate-keeper substitutions such as FGFR2 V564F, observed in cholangiocarcinoma after erdafitinib exposure [61], may emerge in GIST under prolonged FGFR blockade; next-generation inhibitors (e.g., futibatinib) or allosteric FGFR degraders should be evaluated in patient-derived organoids.
At current list prices, FGFR inhibitor therapy costs approximately USD 12,000–15,000 per month. Compassionate use programmes and tiered-pricing agreements will be essential to ensure equitable access, particularly in low- and middle-income countries where GIST incidence is highest.
In summary, FGFR2-targeted therapy has moved from bench to early bedside in GIST. GIST-specific trials, harmonised diagnostics and proactive resistance management will be critical to deliver durable benefit for this ultra-rare but clinically important molecular subset.

6. Conclusions

FGFR2 alterations, occurring in 1–2% of GISTs, play a pivotal role in tumorigenesis and TKI resistance in a subset of cases, particularly those lacking mutations in KIT or PDGFRA. As outlined in Section 3.2, Section 3.3 and Section 3.4, FGFR2 fusions (e.g., FGFR2::TACC2) and amplifications activate oncogenic signaling via MAPK/ERK and PI3K/AKT pathways while enhancing DDR, thereby contributing to resistance against TKIs such as imatinib. Their mutual exclusivity with KIT/PDGFRA mutations, as discussed in Section 4.2, highlights FGFR2 as an alternative oncogenic driver in wild-type GISTs.
Accurate detection of FGFR2 alterations using NGS and FISH, as described in Section 4.3, is critical for identifying patients who may benefit from FGFR inhibitors such as erdafitinib and pemigatinib. Clinical trials and case reports, reviewed in Section 4.1 and Section 4.2, demonstrate the therapeutic potential of FGFR inhibitors, particularly in combination with TKIs or DDR-targeting agents such as PARP inhibitors, offering strategies to overcome resistance in FGFR2-altered GISTs. However, challenges remain, including the rarity of FGFR2 alterations, limited data from GIST-specific trials, and risks of secondary resistance.
Future research should prioritize GIST-specific clinical trials, optimized diagnostic protocols, and multi-omics profiling to better characterize FGFR2-driven GISTs. Collaborative international studies and real-world evidence could help address small sample sizes, thereby enhancing personalized treatment strategies. FGFR2 represents a promising therapeutic target, with combination therapies poised to improve outcomes for patients with TKI-refractory GISTs.

Author Contributions

Conceptualization, Y.H., C.S. and X.L.; data curation, Y.H. and C.S.; writing—original draft preparation, Y.H., C.S. and X.W.; writing—review and editing, Y.H., C.S. and X.L.; supervision, X.L.; project administration, X.L.; funding acquisition, X.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created during this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

GISTGastrointestinal Stromal Tumor
FGFR2Fibroblast Growth Factor Receptor 2
KITKIT Proto-Oncogene, Receptor Tyrosine Kinase
PDGFRAPlatelet-Derived Growth Factor Receptor Alpha
TKITyrosine Kinase Inhibitor
AKTProtein Kinase B (Akt)
ERKExtracellular Signal-Regulated Kinase
MAPKMitogen-Activated Protein Kinase
PI3KPhosphoinositide 3-Kinase
DNADeoxyribonucleic Acid
HRRHomologous Recombination Repair
RAD51RAD51 Recombinase (a protein involved in HRR)
NGSNext-Generation Sequencing
FISHFluorescence In Situ Hybridization
PFSProgression-Free Survival
VEGFVascular Endothelial Growth Factor
RTKReceptor Tyrosine Kinase
FGFFibroblast Growth Factor
DDRDNA Damage Repair
DSBDouble-Strand Break
NHEJNonhomologous End Joining
PARPPoly (ADP-Ribose) Polymerase

References

  1. Nilsson, B.; Bumming, P.; Meis-Kindblom, J.M.; Oden, A.; Dortok, A.; Gustavsson, B.; Sablinska, K.; Kindblom, L.G. Gastrointestinal stromal tumors: The incidence, prevalence, clinical course, and prognostication in the preimatinib mesylate era-A population-based study in western Sweden. Cancer 2005, 103, 821–829. [Google Scholar] [CrossRef] [PubMed]
  2. Joensuu, H. Second line therapies for the treatment of gastrointestinal stromal tumor. Curr. Opin. Oncol. 2007, 19, 353–358. [Google Scholar] [CrossRef] [PubMed]
  3. Huizinga, J.D.; Thuneberg, L.; Kluppel, M.; Malysz, J.; Mikkelsen, H.B.; Bernstein, A. W/kit gene required for interstitial cells of Cajal and for intestinal pacemaker activity. Nature 1995, 373, 347–349. [Google Scholar] [CrossRef]
  4. Kindblom, L.G.; Remotti, H.E.; Aldenborg, F.; Meis-Kindblom, J.M. Gastrointestinal pacemaker cell tumor (GIPACT): Gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cells of Cajal. Am. J. Pathol. 1998, 152, 1259–1269. [Google Scholar]
  5. Heinrich, M.C.; Corless, C.L.; Duensing, A.; McGreevey, L.; Chen, C.J.; Joseph, N.; Singer, S.; Griffith, D.J.; Haley, A.; Town, A.; et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003, 299, 708–710. [Google Scholar] [CrossRef]
  6. Hirota, S.; Isozaki, K.; Moriyama, Y.; Hashimoto, K.; Nishida, T.; Ishiguro, S.; Kawano, K.; Hanada, M.; Kurata, A.; Takeda, M.; et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 1998, 279, 577–580. [Google Scholar] [CrossRef]
  7. Dematteo, R.P.; Heinrich, M.C.; El-Rifai, W.M.; Demetri, G. Clinical management of gastrointestinal stromal tumors: Before and after STI-571. Hum. Pathol. 2002, 33, 466–477. [Google Scholar] [CrossRef]
  8. Dematteo, R.P.; Ballman, K.V.; Antonescu, C.R.; Maki, R.G.; Pisters, P.W.; Demetri, G.D.; Blackstein, M.E.; Blanke, C.D.; von Mehren, M.; Brennan, M.F.; et al. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: A randomised, double-blind, placebo-controlled trial. Lancet 2009, 373, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  9. Kee, D.; Zalcberg, J.R. Current and emerging strategies for the management of imatinib-refractory advanced gastrointestinal stromal tumors. Ther. Adv. Med. Oncol. 2012, 4, 255–270. [Google Scholar] [CrossRef]
  10. Bauer, S.; Joensuu, H. Emerging Agents for the Treatment of Advanced, Imatinib-Resistant Gastrointestinal Stromal Tumors: Current Status and Future Directions. Drugs 2015, 75, 1323–1334. [Google Scholar] [CrossRef] [PubMed]
  11. Corless, C.L.; Schroeder, A.; Griffith, D.; Town, A.; McGreevey, L.; Harrell, P.; Shiraga, S.; Bainbridge, T.; Morich, J.; Heinrich, M.C. PDGFRA mutations in gastrointestinal stromal tumors: Frequency, spectrum and in vitro sensitivity to imatinib. J. Clin. Oncol. 2005, 23, 5357–5364. [Google Scholar] [CrossRef] [PubMed]
  12. Antonescu, C.R.; Besmer, P.; Guo, T.; Arkun, K.; Hom, G.; Koryotowski, B.; Leversha, M.A.; Jeffrey, P.D.; Desantis, D.; Singer, S.; et al. Acquired resistance to imatinib in gastrointestinal stromal tumor occurs through secondary gene mutation. Clin. Cancer Res. 2005, 11, 4182–4190. [Google Scholar] [CrossRef]
  13. Heinrich, M.C.; Corless, C.L.; Blanke, C.D.; Demetri, G.D.; Joensuu, H.; Roberts, P.J.; Eisenberg, B.L.; von Mehren, M.; Fletcher, C.D.; Sandau, K.; et al. Molecular correlates of imatinib resistance in gastrointestinal stromal tumors. J. Clin. Oncol. 2006, 24, 4764–4774. [Google Scholar] [CrossRef]
  14. Serrano, C.; Martin-Broto, J.; Asencio-Pascual, J.M.; Lopez-Guerrero, J.A.; Rubio-Casadevall, J.; Bague, S.; Garcia-Del-Muro, X.; Fernandez-Hernandez, J.A.; Herrero, L.; Lopez-Pousa, A.; et al. 2023 GEIS Guidelines for gastrointestinal stromal tumors. Ther. Adv. Med. Oncol. 2023, 15, 17588359231192388. [Google Scholar] [CrossRef]
  15. Hashimoto, T.; Nakamura, Y.; Komatsu, Y.; Yuki, S.; Takahashi, N.; Okano, N.; Hirano, H.; Ohtsubo, K.; Ohta, T.; Oki, E.; et al. Different efficacy of tyrosine kinase inhibitors by KIT and PGFRA mutations identified in circulating tumor DNA for the treatment of refractory gastrointestinal stromal tumors. BJC Rep. 2024, 2, 54. [Google Scholar] [CrossRef]
  16. Yip, D.; Zalcberg, J.; Blay, J.Y.; Eriksson, M.; Espinoza, D.; Price, T.; Marreaud, S.; Italiano, A.; Steeghs, N.; Boye, K.; et al. Imatinib alternating with regorafenib compared to imatinib alone for the first-line treatment of advanced gastrointestinal stromal tumor: The AGITG ALT-GIST intergroup randomized phase II trial. Br. J. Cancer 2025, 132, 897–904. [Google Scholar] [CrossRef]
  17. Heinrich, M.C.; Corless, C.L.; Demetri, G.D.; Blanke, C.D.; von Mehren, M.; Joensuu, H.; McGreevey, L.S.; Chen, C.J.; Van den Abbeele, A.D.; Druker, B.J.; et al. Kinase Mutations and Imatinib Response in Patients With Metastatic Gastrointestinal Stromal Tumor. J. Clin. Oncol. 2023, 41, 4829–4836. [Google Scholar] [CrossRef]
  18. Debiec-Rychter, M.; Dumez, H.; Judson, I.; Wasag, B.; Verweij, J.; Brown, M.; Dimitrijevic, S.; Sciot, R.; Stul, M.; Vranck, H.; et al. Use of c-KIT/PDGFRA mutational analysis to predict the clinical response to imatinib in patients with advanced gastrointestinal stromal tumours entered on phase I and II studies of the EORTC Soft Tissue and Bone Sarcoma Group. Eur. J. Cancer 2004, 40, 689–695, Correction in Lancet Oncol. 2020, 21, e341. [Google Scholar] [CrossRef]
  19. Wardelmann, E.; Merkelbach-Bruse, S.; Pauls, K.; Thomas, N.; Schildhaus, H.U.; Heinicke, T.; Speidel, N.; Pietsch, T.; Buettner, R.; Pink, D.; et al. Polyclonal evolution of multiple secondary KIT mutations in gastrointestinal stromal tumors under treatment with imatinib mesylate. Clin. Cancer Res. 2006, 12, 1743–1749. [Google Scholar] [CrossRef] [PubMed]
  20. Desai, J.; Shankar, S.; Heinrich, M.C.; Fletcher, J.A.; Fletcher, C.D.; Manola, J.; Morgan, J.A.; Corless, C.L.; George, S.; Tuncali, K.; et al. Clonal evolution of resistance to imatinib in patients with metastatic gastrointestinal stromal tumors. Clin. Cancer Res. 2007, 13, 5398–5405. [Google Scholar] [CrossRef] [PubMed]
  21. Nishida, T.; Kanda, T.; Nishitani, A.; Takahashi, T.; Nakajima, K.; Ishikawa, T.; Hirota, S. Secondary mutations in the kinase domain of the KIT gene are predominant in imatinib-resistant gastrointestinal stromal tumor. Cancer Sci. 2008, 99, 799–804. [Google Scholar] [CrossRef]
  22. Corless, C.L.; Barnett, C.M.; Heinrich, M.C. Gastrointestinal stromal tumours: Origin and molecular oncology. Nat. Rev. Cancer 2011, 11, 865–878. [Google Scholar] [CrossRef]
  23. Ohshima, K.; Nagashima, T.; Fujiya, K.; Hatakeyama, K.; Watanabe, Y.; Morimoto, K.; Kamada, F.; Shimoda, Y.; Ohnami, S.; Naruoka, A.; et al. Whole-genome and Epigenomic Landscapes of Malignant Gastrointestinal Stromal Tumors Harboring KIT Exon 11 557-558 Deletion Mutations. Cancer Res. Commun. 2023, 3, 684–696. [Google Scholar] [CrossRef] [PubMed]
  24. Boichuk, S.; Galembikova, A.; Dunaev, P.; Valeeva, E.; Shagimardanova, E.; Gusev, O.; Khaiboullina, S. A Novel Receptor Tyrosine Kinase Switch Promotes Gastrointestinal Stromal Tumor Drug Resistance. Molecules 2017, 22, 2152. [Google Scholar] [CrossRef] [PubMed]
  25. Li, D.G.; Jiang, J.P.; Chen, F.Y.; Wu, W.; Fu, J.; Wang, G.H.; Li, Y.B. Insulin-like growth factor 2 targets IGF1R signaling transduction to facilitate metastasis and imatinib resistance in gastrointestinal stromal tumors. World J. Gastrointest. Oncol. 2024, 16, 3585–3599. [Google Scholar] [CrossRef]
  26. Kelly, C.M.; Shoushtari, A.N.; Qin, L.X.; D’Angelo, S.P.; Dickson, M.A.; Gounder, M.M.; Keohan, M.L.; McFadyen, C.; Sjoberg, A.; Singer, S.; et al. A phase Ib study of BGJ398, a pan-FGFR kinase inhibitor in combination with imatinib in patients with advanced gastrointestinal stromal tumor. Invest. New Drugs 2019, 37, 282–290. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, Z.; Jiang, N.Y.; Guan, R.Y.; Zhu, Y.K.; Jiang, F.Q.; Piao, D. Identification of critical microRNAs in gastrointestinal stromal tumor patients treated with Imatinib. Neoplasma 2018, 65, 683–692. [Google Scholar] [CrossRef]
  28. Cui, Z.; Sun, H.; Gao, Z.; Li, C.; Xiao, T.; Bian, Y.; Liu, Z.; Gu, T.; Zhang, J.; Li, T.; et al. TRIM21/USP15 balances ACSL4 stability and the imatinib resistance of gastrointestinal stromal tumors. Br. J. Cancer 2024, 130, 526–541. [Google Scholar] [CrossRef]
  29. Shima, T.; Taniguchi, K.; Tokumaru, Y.; Inomata, Y.; Arima, J.; Lee, S.W.; Takabe, K.; Yoshida, K.; Uchiyama, K. Glucose transporter-1 inhibition overcomes imatinib resistance in gastrointestinal stromal tumor cells. Oncol. Rep. 2022, 47, 7. [Google Scholar] [CrossRef]
  30. Rausch, J.L.; Ali, A.A.; Lee, D.M.; Gebreyohannes, Y.K.; Mehalek, K.R.; Agha, A.; Patil, S.S.; Tolstov, Y.; Wellens, J.; Dhillon, H.S.; et al. Differential antitumor activity of compounds targeting the ubiquitin-proteasome machinery in gastrointestinal stromal tumor (GIST) cells. Sci. Rep. 2020, 10, 5178. [Google Scholar] [CrossRef]
  31. Demetri, G.D.; van Oosterom, A.T.; Garrett, C.R.; Blackstein, M.E.; Shah, M.H.; Verweij, J.; McArthur, G.; Judson, I.R.; Heinrich, M.C.; Morgan, J.A.; et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: A randomised controlled trial. Lancet 2006, 368, 1329–1338. [Google Scholar] [CrossRef]
  32. Abrams, T.J.; Lee, L.B.; Murray, L.J.; Pryer, N.K.; Cherrington, J.M. SU11248 inhibits KIT and platelet-derived growth factor receptor beta in preclinical models of human small cell lung cancer. Mol. Cancer Ther. 2003, 2, 471–478. [Google Scholar] [PubMed]
  33. Mendel, D.B.; Laird, A.D.; Xin, X.; Louie, S.G.; Christensen, J.G.; Li, G.; Schreck, R.E.; Abrams, T.J.; Ngai, T.J.; Lee, L.B.; et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: Determination of a pharmacokinetic/pharmacodynamic relationship. Clin. Cancer Res. 2003, 9, 327–337. [Google Scholar] [PubMed]
  34. Mulet-Margalef, N.; Garcia-Del-Muro, X. Sunitinib in the treatment of gastrointestinal stromal tumor: Patient selection and perspectives. Onco Targets Ther. 2016, 9, 7573–7582. [Google Scholar] [CrossRef]
  35. Heinrich, M.C.; Maki, R.G.; Corless, C.L.; Antonescu, C.R.; Harlow, A.; Griffith, D.; Town, A.; McKinley, A.; Ou, W.B.; Fletcher, J.A.; et al. Primary and secondary kinase genotypes correlate with the biological and clinical activity of sunitinib in imatinib-resistant gastrointestinal stromal tumor. J. Clin. Oncol. 2008, 26, 5352–5359. [Google Scholar] [CrossRef]
  36. Judson, I.R. Prognosis, imatinib dose, and benefit of sunitinib in GIST: Knowing the genotype. J. Clin. Oncol. 2008, 26, 5322–5325. [Google Scholar] [CrossRef]
  37. Shirao, K.; Nishida, T.; Doi, T.; Komatsu, Y.; Muro, K.; Li, Y.; Ueda, E.; Ohtsu, A. Phase I/II study of sunitinib malate in Japanese patients with gastrointestinal stromal tumor after failure of prior treatment with imatinib mesylate. Invest. New Drugs 2010, 28, 866–875. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, C.; Zhang, C.; Zhang, T.; Liu, H.; Zhong, J.; Wang, Z.; Wang, L.; Hong, L. Second-line sunitinib for Chinese patients with advanced gastrointestinal stromal tumor: 37.5 mg schedule outperformed 50 mg schedule in adherence and prognosis. Transl. Cancer Res. 2021, 10, 3206–3217. [Google Scholar] [CrossRef]
  39. Sasaki, K.; Kanda, T.; Matsumoto, Y.; Ishikawa, T.; Hirota, S.; Saijo, Y. Sunitinib therapy for imatinib-resistant and/or intolerant gastrointestinal stromal tumors: Comparison of safety and efficacy between standard and reduced dosage regimens. Jpn. J. Clin. Oncol. 2023, 53, 297–303. [Google Scholar] [CrossRef]
  40. Rutkowski, P.; Bylina, E.; Klimczak, A.; Switaj, T.; Falkowski, S.; Kroc, J.; Lugowska, I.; Brzeskwiniewicz, M.; Melerowicz, W.; Osuch, C.; et al. The outcome and predictive factors of sunitinib therapy in advanced gastrointestinal stromal tumors (GIST) after imatinib failure—One institution study. BMC Cancer 2012, 12, 107. [Google Scholar] [CrossRef]
  41. Guo, T.; Hajdu, M.; Agaram, N.P.; Shinoda, H.; Veach, D.; Clarkson, B.D.; Maki, R.G.; Singer, S.; Dematteo, R.P.; Besmer, P.; et al. Mechanisms of sunitinib resistance in gastrointestinal stromal tumors harboring KITAY502-3ins mutation: An in vitro mutagenesis screen for drug resistance. Clin. Cancer Res. 2009, 15, 6862–6870. [Google Scholar] [CrossRef]
  42. Heinrich, M.C.; Jones, R.L.; George, S.; Gelderblom, H.; Schoffski, P.; von Mehren, M.; Zalcberg, J.R.; Kang, Y.K.; Razak, A.A.; Trent, J.; et al. Ripretinib versus sunitinib in gastrointestinal stromal tumor: ctDNA biomarker analysis of the phase 3 INTRIGUE trial. Nat. Med. 2024, 30, 498–506. [Google Scholar] [CrossRef] [PubMed]
  43. George, S.; Blay, J.Y.; Chi, P.; Jones, R.L.; Serrano, C.; Somaiah, N.; Gelderblom, H.; Zalcberg, J.R.; Reichmann, W.; Sprott, K.; et al. The INSIGHT study: A randomized, Phase III study of ripretinib versus sunitinib for advanced gastrointestinal stromal tumor with KIT exon 11 + 17/18 mutations. Future Oncol. 2024, 20, 1973–1982. [Google Scholar] [CrossRef]
  44. Wilhelm, S.M.; Dumas, J.; Adnane, L.; Lynch, M.; Carter, C.A.; Schutz, G.; Thierauch, K.H.; Zopf, D. Regorafenib (BAY 73-4506): A new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int. J. Cancer 2011, 129, 245–255. [Google Scholar] [CrossRef]
  45. Mross, K.; Frost, A.; Steinbild, S.; Hedbom, S.; Buchert, M.; Fasol, U.; Unger, C.; Kratzschmar, J.; Heinig, R.; Boix, O.; et al. A phase I dose-escalation study of regorafenib (BAY 73-4506), an inhibitor of oncogenic, angiogenic, and stromal kinases, in patients with advanced solid tumors. Clin. Cancer Res. 2012, 18, 2658–2667. [Google Scholar] [CrossRef]
  46. Ferraro, D.; Zalcberg, J. Regorafenib in gastrointestinal stromal tumors: Clinical evidence and place in therapy. Ther. Adv. Med. Oncol. 2014, 6, 222–228. [Google Scholar] [CrossRef]
  47. Demetri, G.D.; Reichardt, P.; Kang, Y.K.; Blay, J.Y.; Rutkowski, P.; Gelderblom, H.; Hohenberger, P.; Leahy, M.; von Mehren, M.; Joensuu, H.; et al. Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): An international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013, 381, 295–302. [Google Scholar] [CrossRef]
  48. Bauer, S.; George, S.; von Mehren, M.; Heinrich, M.C. Early and Next-Generation KIT/PDGFRA Kinase Inhibitors and the Future of Treatment for Advanced Gastrointestinal Stromal Tumor. Front. Oncol. 2021, 11, 672500. [Google Scholar] [CrossRef] [PubMed]
  49. Takahashi, T.; Nishida, T.; Kudo, T.; Boku, N.; Honma, Y.; Komatsu, Y.; Nakatsumi, H.; Matsumoto, K.; Onoe, T.; Oki, E.; et al. Regorafenib as second-line therapy for imatinib-resistant gastrointestinal stromal tumor (GIST). J. Clin. Oncol. 2020, 38, 823. [Google Scholar] [CrossRef]
  50. Martin-Broto, J.; Valverde, C.; Hindi, N.; Vincenzi, B.; Martinez-Trufero, J.; Grignani, G.; Italiano, A.; Lavernia, J.; Vallejo, A.; Tos, P.D.; et al. REGISTRI: Regorafenib in first-line of KIT/PDGFRA wild type metastatic GIST: A collaborative Spanish (GEIS), Italian (ISG) and French Sarcoma Group (FSG) phase II trial. Mol. Cancer 2023, 22, 127. [Google Scholar] [CrossRef] [PubMed]
  51. Blay, J.Y.; Serrano, C.; Heinrich, M.C.; Zalcberg, J.; Bauer, S.; Gelderblom, H.; Schoffski, P.; Jones, R.L.; Attia, S.; D’Amato, G.; et al. Ripretinib in patients with advanced gastrointestinal stromal tumours (INVICTUS): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2020, 21, 923–934. [Google Scholar] [CrossRef] [PubMed]
  52. Smith, B.D.; Kaufman, M.D.; Lu, W.P.; Gupta, A.; Leary, C.B.; Wise, S.C.; Rutkoski, T.J.; Ahn, Y.M.; Al-Ani, G.; Bulfer, S.L.; et al. Ripretinib (DCC-2618) Is a Switch Control Kinase Inhibitor of a Broad Spectrum of Oncogenic and Drug-Resistant KIT and PDGFRA Variants. Cancer Cell 2019, 35, 738–751. [Google Scholar] [CrossRef] [PubMed]
  53. Bauer, S.; Jones, R.L.; Blay, J.Y.; Gelderblom, H.; George, S.; Schoffski, P.; von Mehren, M.; Zalcberg, J.R.; Kang, Y.K.; Razak, A.A.; et al. Ripretinib Versus Sunitinib in Patients With Advanced Gastrointestinal Stromal Tumor After Treatment With Imatinib (INTRIGUE): A Randomized, Open-Label, Phase III Trial. J. Clin. Oncol. 2022, 40, 3918–3928. [Google Scholar] [CrossRef]
  54. Gelderblom, H.; Jones, R.L.; Blay, J.Y.; George, S.; von Mehren, M.; Zalcberg, J.R.; Kang, Y.K.; Razak, A.A.; Trent, J.; Attia, S.; et al. Patient-reported outcomes and tolerability in patients receiving ripretinib versus sunitinib after treatment with imatinib in INTRIGUE, a phase 3, open-label study. Eur. J. Cancer 2023, 192, 113245. [Google Scholar] [CrossRef] [PubMed]
  55. Heinrich, M.C.; Jones, R.L.; von Mehren, M.; Schoffski, P.; Serrano, C.; Kang, Y.K.; Cassier, P.A.; Mir, O.; Eskens, F.; Tap, W.D.; et al. Avapritinib in advanced PDGFRA D842V-mutant gastrointestinal stromal tumour (NAVIGATOR): A multicentre, open-label, phase 1 trial. Lancet Oncol. 2020, 21, 935–946. [Google Scholar] [CrossRef]
  56. Henriques-Abreu, M.; Serrano, C. Avapritinib in unresectable or metastatic gastrointestinal stromal tumor with PDGFRA exon 18 mutation: Safety and efficacy. Expert. Rev. Anticancer. Ther. 2021, 21, 1081–1088. [Google Scholar] [CrossRef]
  57. Kang, Y.K.; George, S.; Jones, R.L.; Rutkowski, P.; Shen, L.; Mir, O.; Patel, S.; Zhou, Y.; von Mehren, M.; Hohenberger, P.; et al. Avapritinib Versus Regorafenib in Locally Advanced Unresectable or Metastatic GI Stromal Tumor: A Randomized, Open-Label Phase III Study. J. Clin. Oncol. 2021, 39, 3128–3139. [Google Scholar] [CrossRef]
  58. Adenis, A.; Blay, J.Y.; Bui-Nguyen, B.; Bouche, O.; Bertucci, F.; Isambert, N.; Bompas, E.; Chaigneau, L.; Domont, J.; Ray-Coquard, I.; et al. Masitinib in advanced gastrointestinal stromal tumor (GIST) after failure of imatinib: A randomized controlled open-label trial. Ann. Oncol. 2014, 25, 1762–1769. [Google Scholar] [CrossRef]
  59. Abbaspour Babaei, M.; Kamalidehghan, B.; Saleem, M.; Huri, H.Z.; Ahmadipour, F. Receptor tyrosine kinase (c-Kit) inhibitors: A potential therapeutic target in cancer cells. Drug Des. Devel Ther. 2016, 10, 2443–2459. [Google Scholar] [CrossRef]
  60. Heinrich, M.C.; Griffith, D.; McKinley, A.; Patterson, J.; Presnell, A.; Ramachandran, A.; Debiec-Rychter, M. Crenolanib inhibits the drug-resistant PDGFRA D842V mutation associated with imatinib-resistant gastrointestinal stromal tumors. Clin. Cancer Res. 2012, 18, 4375–4384. [Google Scholar] [CrossRef]
  61. Joensuu, H.; De Braud, F.; Grignagni, G.; De Pas, T.; Spitalieri, G.; Coco, P.; Spreafico, C.; Boselli, S.; Toffalorio, F.; Bono, P.; et al. Vatalanib for metastatic gastrointestinal stromal tumour (GIST) resistant to imatinib: Final results of a phase II study. Br. J. Cancer 2011, 104, 1686–1690. [Google Scholar] [CrossRef]
  62. Blay, J.Y.; Shen, L.; Kang, Y.K.; Rutkowski, P.; Qin, S.; Nosov, D.; Wan, D.; Trent, J.; Srimuninnimit, V.; Papai, Z.; et al. Nilotinib versus imatinib as first-line therapy for patients with unresectable or metastatic gastrointestinal stromal tumours (ENESTg1): A randomised phase 3 trial. Lancet Oncol. 2015, 16, 550–560. [Google Scholar] [CrossRef] [PubMed]
  63. Gelderblom, H.; Jones, R.L.; Blay, J.-Y.; George, S.; Schöffski, P.; Mehren, M.v.; Zalcberg, J.R.; Kang, Y.-K.; Razak, A.R.A.; Trent, J.C.; et al. Patient reported outcomes and tolerability in patients receiving ripretinib versus sunitinib after imatinib treatment in INTRIGUE: A phase 3 open-label study. J. Clin. Oncol. 2022, 40, 11541. [Google Scholar] [CrossRef]
  64. Cauchi, C.; Somaiah, N.; Engstrom, P.F.; Litwin, S.; Lopez, M.; Lee, J.; Davey, M.; Bove, B.; von Mehren, M. Evaluation of nilotinib in advanced GIST previously treated with imatinib and sunitinib. Cancer Chemother. Pharmacol. 2012, 69, 977–982. [Google Scholar] [CrossRef]
  65. Komatsu, Y.; Doi, T.; Sawaki, A.; Kanda, T.; Yamada, Y.; Kuss, I.; Demetri, G.D.; Nishida, T. Regorafenib for advanced gastrointestinal stromal tumors following imatinib and sunitinib treatment: A subgroup analysis evaluating Japanese patients in the phase III GRID trial. Int. J. Clin. Oncol. 2015, 20, 905–912. [Google Scholar] [CrossRef] [PubMed]
  66. Westerdijk, K.; Desar, I.M.E.; Steeghs, N.; van der Graaf, W.T.A.; van Erp, N.P.; Dutch, P.; Oncology, G. Imatinib, sunitinib and pazopanib: From flat-fixed dosing towards a pharmacokinetically guided personalized dose. Br. J. Clin. Pharmacol. 2020, 86, 258–273. [Google Scholar] [CrossRef]
  67. Zheng, S.; Shu, Y.; Lu, Y.; Sun, Y. Chloroquine Combined with Imatinib Overcomes Imatinib Resistance in Gastrointestinal Stromal Tumors by Inhibiting Autophagy via the MAPK/ERK Pathway. Onco Targets Ther. 2020, 13, 6433–6441. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, H.; Park, S.; Kang, S.Y.; Ahn, S.; Kim, K.M. Peritoneal Seeding Is More Common in Gastric Cancer Patients with FGFR2 Amplification or High Tumor Mutation Burden. Diagnostics 2022, 12, 2355. [Google Scholar] [CrossRef]
  69. Dermawan, J.K.; Vanderbilt, C.M.; Chang, J.C.; Untch, B.R.; Singer, S.; Chi, P.; Tap, W.D.; Antonescu, C.R. FGFR2::TACC2 fusion as a novel KIT-independent mechanism of targeted therapy failure in a multidrug-resistant gastrointestinal stromal tumor. Genes. Chromosomes Cancer 2022, 61, 412–419. [Google Scholar] [CrossRef]
  70. Anastassiadis, T.; Deacon, S.W.; Devarajan, K.; Ma, H.; Peterson, J.R. Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat. Biotechnol. 2011, 29, 1039–1045. [Google Scholar] [CrossRef]
  71. Le Cesne, A.; Blay, J.Y.; Bui, B.N.; Bouche, O.; Adenis, A.; Domont, J.; Cioffi, A.; Ray-Coquard, I.; Lassau, N.; Bonvalot, S.; et al. Phase II study of oral masitinib mesilate in imatinib-naive patients with locally advanced or metastatic gastro-intestinal stromal tumour (GIST). Eur. J. Cancer 2010, 46, 1344–1351. [Google Scholar] [CrossRef]
  72. Wilhelm, S.M.; Carter, C.; Tang, L.; Wilkie, D.; McNabola, A.; Rong, H.; Chen, C.; Zhang, X.; Vincent, P.; McHugh, M.; et al. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004, 64, 7099–7109. [Google Scholar] [CrossRef]
  73. Kefeli, U.; Benekli, M.; Sevinc, A.; Yildiz, R.; Kaplan, M.A.; Ciltas, A.; Balakan, O.; Isikdogan, A.; Coskun, U.; Dane, F.; et al. Efficacy of sorafenib in patients with gastrointestinal stromal tumors in the third- or fourth-line treatment: A retrospective multicenter experience. Oncol. Lett. 2013, 6, 605–611. [Google Scholar] [CrossRef]
  74. Park, S.H.; Ryu, M.H.; Ryoo, B.Y.; Im, S.A.; Kwon, H.C.; Lee, S.S.; Park, S.R.; Kang, B.Y.; Kang, Y.K. Sorafenib in patients with metastatic gastrointestinal stromal tumors who failed two or more prior tyrosine kinase inhibitors: A phase II study of Korean gastrointestinal stromal tumors study group. Invest. New Drugs 2012, 30, 2377–2383. [Google Scholar] [CrossRef] [PubMed]
  75. Joensuu, H.; Blay, J.Y.; Comandone, A.; Martin-Broto, J.; Fumagalli, E.; Grignani, G.; Del Muro, X.G.; Adenis, A.; Valverde, C.; Pousa, A.L.; et al. Dovitinib in patients with gastrointestinal stromal tumour refractory and/or intolerant to imatinib. Br. J. Cancer 2017, 117, 1278–1285. [Google Scholar] [CrossRef] [PubMed]
  76. Ganjoo, K.N.; Villalobos, V.M.; Kamaya, A.; Fisher, G.A.; Butrynski, J.E.; Morgan, J.A.; Wagner, A.J.; D’Adamo, D.; McMillan, A.; Demetri, G.D.; et al. A multicenter phase II study of pazopanib in patients with advanced gastrointestinal stromal tumors (GIST) following failure of at least imatinib and sunitinib. Ann. Oncol. 2014, 25, 236–240. [Google Scholar] [CrossRef]
  77. Nascimento, M.; Moura, S.; Parra, L.; Vasconcellos, V.; Costa, G.; Leite, D.; Dias, M.; Fernandes, T.V.A.; Hoelz, L.; Pimentel, L.; et al. Ponatinib: A Review of the History of Medicinal Chemistry behind Its Development. Pharmaceuticals 2024, 17, 1361. [Google Scholar] [CrossRef]
  78. Schoffski, P.; Mir, O.; Kasper, B.; Papai, Z.; Blay, J.Y.; Italiano, A.; Benson, C.; Kopeckova, K.; Ali, N.; Dileo, P.; et al. Activity and safety of the multi-target tyrosine kinase inhibitor cabozantinib in patients with metastatic gastrointestinal stromal tumour after treatment with imatinib and sunitinib: European Organisation for Research and Treatment of Cancer phase II trial 1317 ’CaboGIST’. Eur. J. Cancer 2020, 134, 62–74. [Google Scholar] [CrossRef]
  79. Markowitz, J.N.; Fancher, K.M. Cabozantinib: A Multitargeted Oral Tyrosine Kinase Inhibitor. Pharmacotherapy 2018, 38, 357–369. [Google Scholar] [CrossRef] [PubMed]
  80. George, S.; von Mehren, M.; Fletcher, J.A.; Sun, J.; Zhang, S.; Pritchard, J.R.; Hodgson, J.G.; Kerstein, D.; Rivera, V.M.; Haluska, F.G.; et al. Phase II Study of Ponatinib in Advanced Gastrointestinal Stromal Tumors: Efficacy, Safety, and Impact of Liquid Biopsy and Other Biomarkers. Clin. Cancer Res. 2022, 28, 1268–1276. [Google Scholar] [CrossRef]
  81. Glod, J.; Arnaldez, F.I.; Wiener, L.; Spencer, M.; Killian, J.K.; Meltzer, P.; Dombi, E.; Derse-Anthony, C.; Derdak, J.; Srinivasan, R.; et al. A Phase II Trial of Vandetanib in Children and Adults with Succinate Dehydrogenase-Deficient Gastrointestinal Stromal Tumor. Clin. Cancer Res. 2019, 25, 6302–6308. [Google Scholar] [CrossRef]
  82. Lindauer, M.; Hochhaus, A. Dasatinib. Recent. Results Cancer Res. 2010, 184, 83–102. [Google Scholar] [CrossRef]
  83. Zeng, C.; Zhu, L.; Jia, X.; Pang, Y.; Li, Z.; Lu, X.; Xie, F.; Duan, L.; Wang, Y. Spectrum of activity of dasatinib against mutant KIT kinases associated with drug-sensitive and drug-resistant gastrointestinal stromal tumors. Gastric Cancer 2020, 23, 837–847. [Google Scholar] [CrossRef]
  84. Montemurro, M.; Cioffi, A.; Domont, J.; Rutkowski, P.; Roth, A.D.; von Moos, R.; Inauen, R.; Toulmonde, M.; Burkhard, R.O.; Knuesli, C.; et al. Long-term outcome of dasatinib first-line treatment in gastrointestinal stromal tumor: A multicenter, 2-stage phase 2 trial (Swiss Group for Clinical Cancer Research 56/07). Cancer 2018, 124, 1449–1454. [Google Scholar] [CrossRef]
  85. Wood, J.M.; Bold, G.; Buchdunger, E.; Cozens, R.; Ferrari, S.; Frei, J.; Hofmann, F.; Mestan, J.; Mett, H.; O’Reilly, T.; et al. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration. Cancer Res 2000, 60, 2178–2189. [Google Scholar] [PubMed]
  86. Zhou, Y.; Zhang, X.; Wu, X.; Zhou, Y.; Zhang, B.; Liu, X.; Wu, X.; Li, Y.; Shen, L.; Li, J. A prospective multicenter phase II study on the efficacy and safety of dasatinib in the treatment of metastatic gastrointestinal stromal tumors failed by imatinib and sunitinib and analysis of NGS in peripheral blood. Cancer Med. 2020, 9, 6225–6233. [Google Scholar] [CrossRef] [PubMed]
  87. Fukuda, M.; Mukohara, T.; Kuwata, T.; Sunami, K.; Naito, Y. Efficacy of Trametinib in Neurofibromatosis Type 1-Associated Gastrointestinal Stromal Tumors: A Case Report. JCO Precis. Oncol. 2024, 8, e2300649. [Google Scholar] [CrossRef]
  88. ClinicalTrials.gov. Study of Rogaratinib in Patients with Sarcomas with FGFR Alterations Including SDH-Deficient GIST. ClinicalTrials.gov Identifier: NCT04595747. Available online: https://clinicaltrials.gov/ct2/show/NCT04595747 (accessed on 27 August 2025).
  89. Abou-Alfa, G.K.; Sahai, V.; Hollebecque, A.; Vaccaro, G.; Melisi, D.; Al-Rajabi, R.; Paulson, A.S.; Borad, M.J.; Gallinson, D.; Murphy, A.G.; et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: A multicentre, open-label, phase 2 study. Lancet Oncol. 2020, 21, 671–684. [Google Scholar] [CrossRef] [PubMed]
  90. ClinicalTrials.gov. Study of KIN-3248 in Patients with Advanced Solid Tumors Harboring FGFR Alterations. ClinicalTrials.gov Identifier: NCT05136028. Available online: https://clinicaltrials.gov/ct2/show/NCT05136028 (accessed on 27 August 2025).
  91. ClinicalTrials.gov. A Study of Erdafitinib in Participants with Advanced Solid Tumors and FGFR Gene Alterations. ClinicalTrials.gov Identifier: NCT04083976.. Available online: https://clinicaltrials.gov/ct2/show/NCT04083976 (accessed on 27 August 2025).
  92. Lai, S.W.; Bamodu, O.A.; Chen, J.H.; Wu, A.T.; Lee, W.H.; Chao, T.Y.; Yeh, C.T. Targeted PARP inhibition combined with FGFR1 blockade is synthetically lethal to malignant cells. Mol. Cancer Ther. 2020, 19, 1617–1628. [Google Scholar] [CrossRef] [PubMed]
  93. Cruz, C.; Castroviejo-Bermejo, M.; Gutiérrez-Enríquez, S.; Llop-Guevara, A.; Ibrahim, Y.H.; Gris-Oliver, A.; Bonache, S.; Morancho, B.; Bruna, A.; Rueda, O.M.; et al. RAD51 foci as a biomarker of PARP inhibitor response and synthetic lethality with FGFR inhibition. Cancers 2020, 12, 2119. [Google Scholar] [CrossRef] [PubMed]
  94. Demetri, G.D.; von Mehren, M.; Blanke, C.D.; Van den Abbeele, A.D.; Eisenberg, B.; Roberts, P.J.; Heinrich, M.C.; Tuveson, D.A.; Singer, S.; Janicek, M.; et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N. Engl. J. Med. 2002, 347, 472–480. [Google Scholar] [CrossRef] [PubMed]
  95. MetaGIST Trial Investigators. Comparison of two doses of imatinib for the treatment of unresectable or metastatic gastrointestinal stromal tumors: A meta-analysis of 1640 patients. J. Clin. Oncol. 2010, 28, 1247–1253. [Google Scholar] [CrossRef] [PubMed]
  96. Blay, J.Y.; Serrano, C.; Heinrich, M.C.; Zalcberg, J.; Bauer, S.; Gelderblom, H.; Schöffski, P.; Jones, R.L.; Attia, S.; D’Amato, G.; et al. Ripretinib in patients with advanced gastrointestinal stromal tumors (INVICTUS): A phase III, randomised, placebo-controlled trial. Lancet Oncol. 2020, 21, 923–934. [Google Scholar] [CrossRef] [PubMed]
Figure 1. FGFR2-mediated signaling pathways in gastrointestinal stromal tumors (GISTs).
Figure 1. FGFR2-mediated signaling pathways in gastrointestinal stromal tumors (GISTs).
Cimb 47 00822 g001
Figure 2. FGFR2-mediated DNA damage repair mechanisms in gastrointestinal stromal tumors (GISTs).
Figure 2. FGFR2-mediated DNA damage repair mechanisms in gastrointestinal stromal tumors (GISTs).
Cimb 47 00822 g002
Figure 3. Mutual exclusivity of FGFR2 alterations with KIT/PDGFRA mutations in gastrointestinal stromal tumors (GISTs).
Figure 3. Mutual exclusivity of FGFR2 alterations with KIT/PDGFRA mutations in gastrointestinal stromal tumors (GISTs).
Cimb 47 00822 g003
Table 1. Key FGFR2-mediated signaling pathways and their functional roles in GIST.
Table 1. Key FGFR2-mediated signaling pathways and their functional roles in GIST.
PathwayFunctionReference
MAPK/ERKPromotes cell proliferation and differentiation by activating transcription factors such as c-Jun and c-Fos[20]
PI3K/AKTEnhances cell survival and inhibits apoptosis through regulation of BCL-2 family proteins[21]
STATModulates gene expression related to cell growth and immune responses[22]
PLCγRegulates calcium signaling and cytoskeletal dynamics, contributing to cell migration[23]
Note: All pathways listed below are activated upon FGFR2 dimerization and tyrosine autophosphorylation. Evidence is primarily derived from mesenchymal or epithelial cancer models; GIST-specific validation remains limited.
Table 2. FGFR2 alterations in gastrointestinal stromal tumors (GISTs): types, approximate frequency, and clinical significance.
Table 2. FGFR2 alterations in gastrointestinal stromal tumors (GISTs): types, approximate frequency, and clinical significance.
Type of AlterationApproximate
Frequency in GISTs
Molecular MechanismClinical Significance
FGFR2 fusions (e.g., FGFR2::TACC2, FGFR2::BICC1)<1% [9,31,33]Retain FGFR2 kinase domain → constitutive dimerization and activation of MAPK/ERK and PI3K/AKT pathwaysDrive oncogenesis and resistance to TKIs (e.g., imatinib); generally mutually exclusive with KIT/PDGFRA mutations
FGFR2 amplifications1–2% [2,35,36]Increased gene copy number → receptor overexpression and enhanced downstream signalingAssociated with higher tumor grade, aggressive behavior, and TKI resistance; potential biomarker for FGFR inhibitor sensitivity
FGFR2 point mutations<0.1% [37,38]Rare missense mutations (reported in other cancers, not recurrent in GISTs)No confirmed clinical significance in GIST; uncertain therapeutic relevance
Polymorphisms (e.g., SNPs such as rs2981582)Not established in GIST; reported in breast and gastric cancer [37]Germline variants linked to cancer susceptibility in other malignanciesNo proven association with GIST incidence or outcome; requires further investigation
Table 3. FGFR2 activation mechanisms in GISTs.
Table 3. FGFR2 activation mechanisms in GISTs.
MechanismDescriptionReferences
Gene FusionsFGFR2::TACC2 fusions, which retain the FGFR2 kinase domain, result in constitutive dimerization and autophosphorylation, independent of fibroblast growth factor (FGF) ligands. This leads to the sustained activation of downstream pathways, thereby enhancing tumor growth and tyrosine kinase inhibitor resistance.[9,31,40]
Gene AmplificationsFGFR2 amplifications increase receptor density on the cell membrane, amplifying signaling even with low ligand levels. Overexpression of FGF ligands (e.g., FGF7, FGF10) further enhances FGFR2 activation in amplified cases.[36,41]
Ligand OverexpressionIn some GISTs, the autocrine or paracrine overexpression of FGF7 and FGF10, which is specific to the FGFR2b isoform, drives pathway activation, particularly in mesenchymal-derived tumors.[42]
Table 4. FGFR2-mediated signaling pathways in GISTs.
Table 4. FGFR2-mediated signaling pathways in GISTs.
PathwayKey EffectorsFunctional OutcomeEvidence in GISTKey Refs.
MAPK/ERKRAS → RAF → MEK → ERKProliferation, transcriptional reprogrammingPhospho ERK high in FGFR2 fusion tumours; bypasses imatinib blockade[20,40,43]
PI3K/AKTPI3K → PDK1 → AKT → mTORSurvival, protein synthesis, chemo resistanceAKT phosphorylation variable; synergistic lethality with MEK inhibitors[3,21,44]
PLCγPLCγ → IP3/DAG → Ca2+/PKCCytoskeletal remodelling, migrationDetected in cell lines only; not validated in patient tissue[22]
JAK/STATJAK → STAT1/3Immune evasion, cytokine feed forwardInferred from transcriptomic signatures; no phospho data available[23]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hong, Y.; Wang, X.; Shou, C.; Liu, X. Molecular Mechanisms and Clinical Implications of Fibroblast Growth Factor Receptor 2 Signaling in Gastrointestinal Stromal Tumors. Curr. Issues Mol. Biol. 2025, 47, 822. https://doi.org/10.3390/cimb47100822

AMA Style

Hong Y, Wang X, Shou C, Liu X. Molecular Mechanisms and Clinical Implications of Fibroblast Growth Factor Receptor 2 Signaling in Gastrointestinal Stromal Tumors. Current Issues in Molecular Biology. 2025; 47(10):822. https://doi.org/10.3390/cimb47100822

Chicago/Turabian Style

Hong, Yanyun, Xiaodong Wang, Chunhui Shou, and Xiaosun Liu. 2025. "Molecular Mechanisms and Clinical Implications of Fibroblast Growth Factor Receptor 2 Signaling in Gastrointestinal Stromal Tumors" Current Issues in Molecular Biology 47, no. 10: 822. https://doi.org/10.3390/cimb47100822

APA Style

Hong, Y., Wang, X., Shou, C., & Liu, X. (2025). Molecular Mechanisms and Clinical Implications of Fibroblast Growth Factor Receptor 2 Signaling in Gastrointestinal Stromal Tumors. Current Issues in Molecular Biology, 47(10), 822. https://doi.org/10.3390/cimb47100822

Article Metrics

Back to TopTop