Abstract
Point mutations of the fibroblast growth factor receptor (FGFR)2 receptor in intrahepatic cholangiocarcinoma (iCC) are mainly of unknown functional significance compared to FGFR2 fusions. Pemigatinib, a tyrosine kinase inhibitor, is approved for the treatment of cholangiocarcinoma with FGFR2 fusion/rearrangement. Although it is hypothesized that FGFR2 mutations may cause uncontrolled activation of the signaling pathway, the data for targeted therapies for FGFR2 mutations remain unclear. In vitro analyses demonstrated the importance of the p.C382R mutation for ligand-independent constitutive activation of FGFR2 with transforming potential. The following report describes the clinical case of a patient diagnosed with an iCC carrying a FGFR2 p.C382R point mutation which was detected in liquid, as well as in tissue-based biopsies. The patient was treated with pemigatinib, resulting in a sustained complete functional remission in fluorodeoxyglucose-positron emission tomography/computed tomography over 10 months to date. The reported case is the first description of a complete functional remission under the treatment with pemigatinib in a patient with p.C383R mutation.
Keywords: cholangiocarcinoma, FGFR2, mixed-all-nominated-in-one method, next-generation sequencing, targeted therapy, tyrosine kinase inhibitor
Introduction
Cholangiocarcinoma (CCA) accounts for 3% of upper gastrointestinal carcinomas. 1 The prognosis of the disease is poor, as most patients are diagnosed in an unresectable late stage. Clinically, extrahepatic CCA, which includes perihilar CCA and distal CCA is distinguished from intrahepatic CCA (iCC), which can be characterized by a different molecular profile and clinical presentation. With the increasing efforts of molecular diagnostics in the context of personalized medicine, CCAs could be further classified. It was shown that oncogenic and targetable gene alterations occur in about 50% of CCAs. 2 Alterations in IDH1 and fibroblast growth factor receptor (FGFR)2 genes are found nearly exclusively in iCC. 1
Based on findings of the FIGHT 202 trial, pemigatinib was recently approved in the United States and Europe for the treatment of patients with FGFR2 fusions and rearrangements.3,4
The FGFR belongs to the tyrosine kinase family and is a single-pass membrane protein consisting of three N-terminal immunoglobulin-like extracellular domains (D1–3), a transmembrane alpha helix domain and an intracellular tyrosine kinase domain (Figure 1). 5 Ligand binding of the fibroblast growth factor and a cofactor (heparin) is required for dimerization of FGFR and thus the activation of the receptor interaction.6,7 Ligand binding to FGFR2 triggers the dimerization of the receptor and results in a tight conjunction of both transmembrane alpha helices followed by an activation of the intracellular proteinkinase domain. This results in the autophosphorylation of the receptor. Subsequently, downstream pathways are activated (phospholipase C cascade and phospholipase C γ, RAS-RAF-MAPK, and PI3K-AKT-mTOR) resulting in proliferation, activation, and transforming signals. 8 PTEN is the main inhibitor of AKT in the PIK-AKT-mTOR pathway (Figure 2).8,9
In the case of FGFR2 fusion/rearrangement, ligand-independent continuous receptor activation occurs with a sustained proliferation signal to the cell. 7 The FGFR2 has a gatekeeper function at location V564. After the binding of pemigatinib N549, K641 and E565 form a hydrogen bond with an inactivating conformation of the activation loop. K659 is essential for stabilizing this inactivating confirmation. 1 Pemigatinib can inhibit both wild-type receptor and ligand-independent activations of FGFR2 by receptor fusion/rearrangement. In contrast, there are only few data available on the functional relevance of FGFR2 mutations.
An in vitro study from 1997 shows that the p.C382R mutation leads to ligand-independent, constitutive activation of the intracellular tyrosine kinase. It was hypothesized that the point mutation results in a permanent tight junction of intracellular alpha helices with subsequent ligand-independent activation of receptor and downstream pathways (Figure 3). 10 Moreover, it was reported that the mutation as such is oncogenic. 11
Clinical case
A 74-year-old male was diagnosed with an advanced iCC of both liver lobes and pulmonary metastases (Figures 5 and 6). First-line chemotherapy with gemcitabine 800 mg/m2, cisplatin 25 mg/m2, and nab-paclitaxel 100 mg/m2 days 1 and 8 qd22 was initiated. After the fifth treatment cycle, progressive disease was observed. Biopsies of the liver lesions as well as a liquid biopsy were taken and a hybrid capture-based next-generation sequencing (NGS; FoundationOne CDx, Penzberg, Germany) was performed for the tissue biopsies and FoundationOne Liquide CDx for the blood samples. 9 The test is based on the examination of 324 genes as well as introns of 34 genes known to be involved in rearrangements. In addition, tumor mutation burden and microsatellite instability were evaluated.12–14 Sequencing revealed a p.C382R mutation located in the transmembrane receptor domain. The p.C382R was detected in both specimens with a variant allele frequency of 76.48% in the tissue biopsy and 8.1% in the blood sample. Regarding downstream signaling pathways, we detected a PTEN alteration (loss of exons 7–9 in tissue samples and loss of exons 3–8 in the liquid biopsy). The sequencing results are shown in Table 1. The results of the histologic examination are shown in Figure 4.
Table 1.
Antibody | Antigen | Provider | Dilution | Epitope retrieval | Incubation |
---|---|---|---|---|---|
Mouse IgG1, kappa | CK 7, clone: OV-TL | DCS GmbH | 1:500 | Pressure cooker in citrate buffer 20 min | Overnight RT |
Mouse IgG1 | Ki-67, clone: K-2 | Zytomed GmbH | 1:500 | Pressure cooker in citrate buffer 20 min | Overnight RT |
rabbit IgG1 | Her-2, clone: SP3 | Zytomed GmbH | 1:75 | Pressure cooker in citrate buffer 20 min | Overnight RT |
rabbit IgG1 | PD-L1, clone: Cal10 | Zytomed GmbH | 1:50 | Pressure cooker in citrate buffer 20 min | Overnight RT |
NGS results of liquid biopsy | |
---|---|
Blood tumor mutational burden | 3 Muts/Mb |
Microsatellite status | MSS |
PIK3CA (H1047R) | 0.16% |
ARID1A (A45fs*6) | 6.6% |
FGFR2 (C382R) | 8.1% |
MTOR (S2013G) | 0.14% |
PTEN (deletion exons 3–8) | 0.57% |
NGS results of tissue biopsy | |||
---|---|---|---|
Gene | Protein effect | CNA | VAF (%) |
FGFR2 | C382R | – | 76.48 |
ARIA1A | A45fs*6 | – | 67.81 |
MYC | Amplification – equivocal | 6 | – |
PARP1 | Amplification – equivocal | 6 | – |
CNA, Copy Number Alteration; NGS, next-generation sequencing; VAF, variant allele frequency.
Furthermore, we performed an in silico study to understand the potential mode of p.C382R action. To modulate and visualize the protein structure results from p.C382R, we used AlphaFold2 (https://alphafold.ebi.ac.uk/entry/P21802). The evaluation revealed that the p.C382R mutation is located in the transmembrane domain at a position that is crucial for the oncogenic tyrosine kinase activation but does not interfere with the binding ability and inhibition of autophosphorylation of FGFR by pemigatinib.
Following the promising in vitro data for pemigatinib-targeted treatment and the reported results of three patients carrying FGFR2 p.C382R mutations who were treated with pemigatinib in the FIGHT 202 trial, we decided to initiate therapy after a discussion of the clinical case in the molecular tumor board (MTB). 10
Pemigatinib of 13.5 mg was administered once daily for 14 days, followed by 7 days of therapy-free interval. The treatment outcome was evaluated by magnetic resonance imaging (MRI) and fluorodeoxyglucose-positron emission tomography/computed tomography (FDG-PET/CT) before pemigatinib administration and after 3 months (Figures 5 and 6). In the pre- and post-MRI image data, an interactive tumor delineation method previously applied to liver CT data was used to determine the change in tumor size. 15 The images show a significant decrease in the tumor lesion volume from 453.9 to 133.7 ml after 3 months of treatment (Figure 5).
The FDG-PET/CT revealed a complete functional remission of the iCC and lung metastases after 3 months of treatment (Figure 6). The therapy is well tolerated without any side effects. The patient continues to be treated with 13.5 mg as described above. Over the course of the treatment, tumor markers that were elevated at baseline measurement decreased to a plateau (Figure 7).
Discussion
The results of the FIGHT 202 phase II trial led to the approval of the tyrosine kinase inhibitor (TKI) pemigatinib for the treatment of patients with CCA and fusion/rearrangement of FGFR2. In the FIGHT 202 trail, potentially targetable oncogenic driver alterations were detected in 44.5% of the enrolled patients. 4 Of 20 patients without fusion/rearrangement of FGFR2, four carried a p.C382R mutation. 10 Of these, three patients had co-occurrence of a BAP1 variant and responded to treatment. The best overall response of these three patients was stable disease with tumor shrinkage of −26% and −30.6% and a reported progression-free survival (PFS) of 6.9; 4.0 and 9.0 months. 10 Our case report is the first study that showed a complete clinical remission of a patient treated with pemigatinib who carried a FGFR2 point mutation. The p.C382R mutation results in replacement of cysteine by arginine at amino acid position 382 of the transmembrane domain of FGFR2. The mutation results in an uncontrolled and prolonged downstream activation of FGFR2 pathways. In vitro experiments demonstrated that p.C383R leads to a constitutive ligand-independent activation of the intracellular tyrosine kinase domain of FGFR2, resulting in autophosphorylation of the receptor with subsequent activation of the downstream pathway. 12
The clinical response under treatment with pemigatinib is further supported by in vitro data of Nakamura and colleagues. 7 They investigated the transformation activity and drug sensitivity of 110 FGFR variants using the so-called mixed-all-nominated-in-one (MANO) method. 7 The MANO method is a functional assay using Ba/F3 cells colony stimulating factor or interleukin 3-dependent, murine pro-B-cells and mouse fibroblast cell line 3T3 which was previously described. 7 An IC50 < 10 nM was determined for growth inhibition of 3T3 cells bearing a FGFR2 p.C382R mutation treated in vitro with pemigatinib. In the last decades, several in vitro and in vivo models have been developed to explore and increase their complexity and reliability to investigate treatment response in CCA. 16 However, especially in CCA, the tumor microenvironment needs to be further investigated to evaluate additional therapies and biomarkers, which was recently discussed in a review by Massa et al. 16
In our patient, we simultaneously detected a loss of PTEN exons 7–9 in tissue biopsy and loss of exons 3–8 in the liquid biopsy of this patient, leading to a functional loss of PTEN. PTEN is known to be a key modulator of the PIK3CA-AKT pathway. As a functional tumor suppressor, the loss of PTEN may additionally activate the downstream pathway of mutated receptor tyrosine kinases FGFR2.
These findings are also supported by data from a study published in 2015, which describes the mechanism of FGFR dimerization and activation. 9 The authors investigated the effect of different mutations in the transmembrane domain of FGFRs and showed that the FGFR3 A391E mutation, that occurred analogously in the transmembrane domain of FGFR3, led to stabilization of the receptor dimer, which ultimately mimics the effect of the ligand binding and explains the activation of FGFR2 due to p.C382R. 9
Although the expansion of treatment options has been broadened by the approval of pemigatinib, studies have shown that the efficacy can be significantly limited by the emergence of acquired resistance.4,17 Secondary polyclonal mutations, in particular, can lead to these resistance mechanisms and underscore the importance of further research efforts to optimize the use of these molecularly targeted therapies.3,17,18 Pemigatinib is a reversible ATP-competitive FGFR inhibitor. As shown in studies, the occurrence of drug resistance due to mutations within the binding site is relatively common with reversible ATP-competitive kinase inhibitors.19–22 In contrast, kinase inhibitors that inhibit kinase activity irreversibly through covalent binding can achieve a longer treatment response compared to ATP-competitive inhibitors.22–24 Futibatinib is an irreversible FGFR1-4 inhibitor and covalently binds to the FGFR kinase domain, inhibiting FGFR phosphorylation and thus downstream signaling. A study by Sootome et al. 25 showed that the frequency of occurrence of drug-resistant clones was lower with futibatinib than with a reversible ATP-competitive FGFR inhibitor.22,24 Futibatinib inhibited multiple drug-resistant FGFR2 mutants, including FGFR2 V565I/L gatekeeper mutants, with greater efficacy than all reversible FGFR inhibitors tested (IC50, 1.3–50.6 nmol/L). 24 Irreversible and reversible FGFR inhibitors differ in their binding region. Futibatinib binds covalently to a highly conserved P-loop cysteine residue in the ATP pocket of FGFR (C492 in the FGFR2-IIIb isoform). Reversible binding of inhibitors occurs mainly in the hinge region of the ATP-binding pocket of FGFR, where drug-resistant mutations are frequently detected. Thus, the use of futibatinib is particularly suggested in patients with resistance to prior TKI therapies. 24
Acquired resistance in the form of polyclonal FGFR2 kinase domain mutations may shorten response. This underscores the utility of serial biopsy and circulating tumor DNA (ctDNA) analyses to identify resistance mechanisms and to guide the selection of FGFR inhibitors, as discussed in a study by Goyal et al. 17 The study showed that polyclonal mutations in the FGFR2 kinase domain could be detected when cell-free DNA biopsy samples were taken at different stages (at baseline and after progression) reflecting the state of resistance. 17 ctDNA analysis can detect a greater extent of resistance mechanisms than tumor biopsy alone which, in terms of tumor heterogeneity, may play a role in resistance mechanisms and the frequently observed mixed response to FGFR inhibitors. 3 Thus, serial ctDNA analysis represents a method that can provide complementary information on FGFR resistance mechanisms. 25
Nevertheless, tissue biopsy is necessary to confirm the histologic diagnosis of CCA. In addition, multigene analysis should be performed in all patients with iCC, as targeted oncogenic alterations are detectable in approximately 50% of cases.2,26 In tissue-based molecular analysis, the quality of the extracted DNA is often insufficient for multigene analysis. Combined diagnostics with ctDNA and proteins using liquid biopsies (omics) will gain importance in the future, both for initial diagnosis and for monitoring during treatment. Our case report also shows that liquid biopsy provides a reliable result compared with tissue-based NGS analysis. Nevertheless, the diagnosis of CCA at an early stage remains a clinical challenge, especially in patients with primary sclerosing cholangitis and biliary strictures.
Another FGFR inhibitor currently being investigated in clinical trials is derazantinib.27,28 The FIDES-01 phase II trial is evaluating the efficacy of derazantinib, which is directed against FGFR1-3 and the CSF1Rkinase in patients with advanced iCC.27,29 The fact that the FIDES-01 study includes patients with FGFR2 mutation or amplification (FGFRM/A) in addition to patients with FGFR2 fusion is of particular interest. 29 The results of an interim analysis of 23 patients were presented at ASCO 2022. 29 In the interim analysis, two patients (8.7%) showed partial remission and 15 patients (65.2%) presented with stable disease, resulting in a disease control rate of 73.9%. The median PFS was 7.3 months. The authors describe a clinical response in all molecular subtypes enrolled in the FGFRM/A group. 29 These data underline that not only FGFR2 fusions, but also mutations or amplifications are actionable targets in iCC. These results should be discussed in MTBs and be considered when recommending therapy. Given that derazantinib also inhibits CSF1Rkinase, it is hypothesized that the inhibition of CSF1R leads to reactivation of exhausted T cells, can reverse tumor-induced immunosuppression, and supports macrophage activity. Therefore, it is hypothesized that the combination of derazantinib and atezolizumab (anti-programmed death-ligand 1) may result in improved efficacy due to inhibition of immunosuppressive stromal cells by the TKI via CSF1R and checkpoint inhibition via PD-L1. 28 This combination is being investigated in the ADVANCE phase II trial (NCT05174650) led by Arndt Vogel and colleagues, which is currently recruiting and aims to enroll 37 patients.
Conclusion
The reported clinical case in conjunction with the three reported patients in the FIGHT 202 trial demonstrates the oncogenic relevance of the p.C382R mutation of the FGFR2 receptor. The gene variant is clinically relevant as it is sensitive to targeted therapy with pemigatinib. To our knowledge, the reported case is the first description of complete functional remission with pemigatinib in a patient with p.C382R mutation. This report highlights the importance of the transforming activity and drug sensitivity of p.C382R mutations based on in vitro assay as described by Nakamura for in vivo clinical application. 7 Furthermore, the comparison of tissue-based NGS in conjunction with liquid biopsy demonstrates that also blood samples are suitable to detect potentially druggable actionable driver mutations in patients with metastatic iCC. Sequencing of serial biopsies and ctDNA could prolong the duration of response to targeted treatments and become a fundamental tool in the daily management of these patients. 30 Multigene sequencing should be performed in every patient with advanced iCC, as not only FGFR fusions/rearrangements but also other alterations have oncogenic potency and may respond to targeted treatment. However, many challenges remain, such as the management of secondary polyclonal mutations, the ideally timed use of liquid biopsies, and the identification of biomarkers predictive of response to FGFR inhibitors.
Acknowledgments
None.
Footnotes
ORCID iDs: Louisa Hempel https://orcid.org/0000-0001-5145-4705
Patrick Philipp https://orcid.org/0000-0002-7968-9757
Contributor Information
Louisa Hempel, Medical School, Sigmund Freud University, Freudplatz 3, Vienna 1020, Austria.
Constantin Lapa, University Hospital Augsburg, Department of Nuclear Medicine, Augsburg, Germany.
Alexander Dierks, University Hospital Augsburg, Department of Nuclear Medicine, Augsburg, Germany.
Andreas Gaumann, Molekularpathologie Suedbayern, Kaufbeuren, Germany.
Josef Scheiber, BioVariance GmbH, Waldsassen, Germany.
Julia Veloso de Oliveira, Fraunhofer Institute of Optronics, System Technologies and Image Exploitation IOSB, Karlsruhe, Germany.
Patrick Philipp, Fraunhofer Institute of Optronics, System Technologies and Image Exploitation IOSB, Karlsruhe, Germany.
Cristina Oyarzun Laura, Fraunhofer Institute for Computer Graphics Research IGD, Darmstadt, Germany.
Stefan Wesarg, Fraunhofer Institute for Computer Graphics Research IGD, Darmstadt, Germany.
Sebastian Robert, Rosenheim Technical University of Applied Sciences, Rosenheim, Germany.
Dirk Hempel, Institute of Translational Molecular Tumor Research, Freising, Germany.
Declarations
Ethics approval and consent to participate: Patients were informed by an oncological as well as a genetics specialist before they provided written informed consent for the collection of tumor samples and NGS analysis as well as the publication of the case report. The clinical case was presented in the MTB which consisted of an interdisciplinary team coordinated by the Cancer Center Dachau and includes experts in clinical and translational oncology, pathology, bioinformatics, molecular biology, radiology, and human genetics. The study was conducted according to the guidelines of the Declaration of Helsinki of 1964 and its later amendments.
Consent for publication: The patient consented to the publication of the data in the present anonymized form as well as to the publication of the image material and the description of the clinical course.
Author contribution(s): Louisa Hempel: Investigation; Validation; Visualization; Writing – original draft; Writing – review & editing.
Constantin Lapa: Resources; Visualization; Writing – review & editing.
Alexander Dierks: Writing – original draft.
Andreas Gaumann: Resources; Supervision; Validation; Writing – review & editing.
Josef Scheiber: Software; Validation; Visualization; Writing – review & editing.
Julia Veloso de Oliveira: Software; Visualization; Writing – review & editing.
Patrick Philipp: Software; Validation; Visualization; Writing – review & editing.
Cristina Oyarzun Laura: Resources; Software; Writing – review & editing.
Stefan Wesarg: Conceptualization; Software; Writing – review & editing.
Sebastian Robert: Resources; Software; Writing – original draft; Writing – review & editing.
Dirk Hempel: Supervision; Validation; Writing – original draft; Writing – review & editing.
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
Competing interests: The authors declare that there is no conflict of interest.
Availability of data and materials: The NGS panel sequencing dataset generated during the current study is not publicly available as these are patient samples with potentially identifiable genetic information and there is no patient consent for depositing this sequencing data in a public repository. However, the data are available from the corresponding author on reasonable request.
References
- 1. Lowery MA, Ptashkin R, Jordan E, et al. Comprehensive molecular profiling of intrahepatic and extrahepatic cholangiocarcinomas: potential targets for intervention. Clin Cancer Res 2018; 24: 4154–4161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Lamarca A, Edeline J, Goyal L. How I treat biliary tract cancer. ESMO Open 2022; 7:100378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Rizzo A, Ricci AD, Brandi G. Pemigatinib: hot topics behind the first approval of a targeted therapy in cholangiocarcinoma. Cancer Treat Res Commun 2021; 27: 100337. [DOI] [PubMed] [Google Scholar]
- 4. Abou-Alfa GK, Sahai V, Hollebecque A, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol 2020; 21: 671–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Li F, Peiris MN, Donoghue DJ. Functions of FGFR2 corrupted by translocations in intrahepatic cholangiocarcinoma. Cytokine Growth Factor Rev 2020; 52: 56–67. [DOI] [PubMed] [Google Scholar]
- 6. Krook MA, Reeser JW, Ernst G, et al. Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance. Br J Cancer 2021; 124: 880–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Nakamura IT, Kohsaka S, Ikegami M, et al. Comprehensive functional evaluation of variants of fibroblast growth factor receptor genes in cancer. NPJ Precis Oncol 2021; 5: 66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 2010; 141: 1117–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Sarabipour S, Hristova K. Mechanism of FGF receptor dimerization and activation. Nat Commun 2016; 7: 10262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Silverman IM, Hollebecque A, Friboulet L, et al. Clinicogenomic analysis of FGFR2–rearranged cholangiocarcinoma identifies correlates of response and mechanisms of resistance to pemigatinib. Cancer Discov 2021; 11: 326–339. [DOI] [PubMed] [Google Scholar]
- 11. Li Y, Mangasarian K, Mansukhani A, et al. Activation of FGF receptors by mutations in the transmembrane domain. Oncogene 1997; 14: 1397–1406. [DOI] [PubMed] [Google Scholar]
- 12. Allegretti M, Fabi A, Buglioni S, et al. Tearing down the walls: FDA approves next generation sequencing (NGS) assays for actionable cancer genomic aberrations. J Exp Clin Cancer Res 2018; 37: 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017; 9: 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Frampton GM, Fichtenholtz A, Otto GA, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol 2013; 31: 1023–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Oyarzun Laura C, Drechsler K, Wesarg S, et al. Accurate physics-based registration for the outcome validation of minimal invasive interventions and open liver surgeries. IEEE Trans Biomed Eng 2017; 64: 362–371. [DOI] [PubMed] [Google Scholar]
- 16. Massa A, Varamo C, Vita F, et al. Evolution of the experimental models of cholangiocarcinoma. Cancers (Basel) 2020; 12: 2308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Goyal L, Saha SK, Liu LY, et al. Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion–positive cholangiocarcinoma. Cancer Discov 2017; 7: 252–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Smyth EC, Babina IS, Turner NC. Gatekeeper mutations and intratumoral heterogeneity in FGFR2–translocated cholangiocarcinoma. Cancer Discov 2017; 7: 248–249. [DOI] [PubMed] [Google Scholar]
- 19. Nakamura H, Arai Y, Totoki Y, et al. Genomic spectra of biliary tract cancer. Nat Genet 2015; 47: 1003–1010. [DOI] [PubMed] [Google Scholar]
- 20. Borad MJ, Champion MD, Egan JB, et al. Integrated genomic characterization reveals novel, therapeutically relevant drug targets in FGFR and EGFR pathways in sporadic intrahepatic cholangiocarcinoma. PLoS Genetics 2014; 10: e1004135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Borger DR, Tanabe KK, Fan KC, et al. Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. Oncologist 2012; 17: 72–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Goyal L, Kongpetch S, Crolley VE, et al. Targeting FGFR inhibition in cholangiocarcinoma. Cancer Treat Rev 2021; 95: 102170. [DOI] [PubMed] [Google Scholar]
- 23. Ross JS, Wang K, Gay L, et al. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist 2014; 19: 235–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Sootome H, Fujita H, Ito K, et al. Futibatinib is a novel irreversible FGFR 1–4 inhibitor that shows selective antitumor activity against FGFR-deregulated tumors. Cancer Res 2020; 80: 4986–4997. [DOI] [PubMed] [Google Scholar]
- 25. Rizzo A, Ricci AD, Tavolari S, et al. Circulating tumor DNA in biliary tract cancer: current evidence and future perspectives. Cancer Genomics Proteomics 2020; 17: 441–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Bekaii-Saab TS, Bridgewater J, Normanno N. Practical considerations in screening for genetic alterations in cholangiocarcinoma. Ann Oncol 2021; 32: 1111–1126. [DOI] [PubMed] [Google Scholar]
- 27. Braun S, McSheehy P, Litherland K, et al. Derazantinib: an investigational drug for the treatment of cholangiocarcinoma. Expert Opin Investig Drugs 2021; 30: 1071–1080. [DOI] [PubMed] [Google Scholar]
- 28. Droz Dit Busset M, Braun S, El-Rayes B, et al. Efficacy of derazantinib (DZB) in patients (pts) with intrahepatic cholangiocarcinoma (iCCA) expressing FGFR2-fusion or FGFR2 mutations/amplifications. Ann Oncol 2019; 30: v276–v277. [Google Scholar]
- 29. Javle MM, Abou-Alfa GK, Macarulla T, et al. Efficacy of derazantinib in intrahepatic cholangiocarcinoma patients with FGFR2 mutations or amplifications: interim results from the phase 2 study FIDES-01. J Clin Oncol 2022; 40: 427.34793250 [Google Scholar]
- 30. Rizzo A, Ricci AD, Tober N, et al. Second-line treatment in advanced biliary tract cancer: today and tomorrow. Anticancer Res 2020; 40: 3013–3030. [DOI] [PubMed] [Google Scholar]