Precision oncology relies on the identification of molecular alterations, responsible for tumor initiation and growth, which are suitable targets of specific inhibitors. The development of FGFR inhibitors represents an edifying example of the rapid evolution in the field of targeted oncology, with 10 different FGFR tyrosine kinase inhibitors actually under clinical investigation. In parallel, the discovery of FGFR activating molecular alterations (mainly FGFR3 mutations and FGFR2 fusions) across many tumor types, especially urothelial carcinomas and intrahepatic cholangiocarcinomas, widens the selection of patients that might benefit from selective FGFR inhibitors. The ongoing concomitant clinical evaluation of selective FGFR inhibitors in molecularly selected solid tumors brings new hopes for patients with metastatic cancer, for tumors so far excluded from molecularly guided treatments. Matching molecularly selected tumors with selective FGFR inhibitors has indeed led to promising results in phase I and II trials, justifying their registration to be expected in a near future, such as the recent accelerated approval of erdafitinib granted by the FDA for urothelial cancer. Widening our knowledge of the activity, efficacy, and toxicities relative to the selective FGFR tyrosine kinase inhibitors under clinical investigation, according to the exact FGFR molecular alteration, will be crucial to determine the optimal therapeutic strategy for patients suffering from FGFR-driven tumors. Similarly, identifying with appropriate molecular diagnostic, every single tumor harboring targetable FGFR alterations will be of utmost importance to attain the best outcomes for patients with FGFR-driven cancer.

The evolving spectrum of oncogene alterations accompanied by the increasing availability of specific inhibitors both concur to produce previously unachievable clinical results. The recent approval of dabrafenib/trametinib in BRAF-mutated cancers and larotrectinib for NTRK-rearranged tumors, regardless of tumor histology, is an emblematic example demonstrating the relevancy of molecular diagnosis that, coupled with specific inhibitors, can lead to an impressive improvement of patients' outcome (1, 2).

The development of FGFR1–4 tyrosine kinase inhibitors (TKI) is similarly being concomitantly evaluated in multiple tumor types. Since the identification of FGFR as a relevant player in cancer, abundant efforts have been dedicated toward its efficient inhibition, leading to a nearly simultaneous release of competing drug candidates, by contrast to the slower but classical stepwise investigation of generations of inhibitors (e.g., EGFR and ALK-TKI in lung cancer).

Previous strategies for FGFR inhibition in clinical trials have been reviewed elsewhere (3–5). This review discusses the recent improvements in selective FGFR targeting, coupled with the detection of functionally validated FGFR molecular drivers.

The role of the FGFR tyrosine kinase family as oncogenic drivers is more heterogeneous than the classical BRAF, ALK, or EGFR ones. Four different FGFR genes (1–4) can be affected by mutation, rearrangement, or amplification across multiple tumor types. The location of mutations, mainly affecting FGFR3 gene, differ from the ones activating the kinase domain of other oncogenes (e.g., EGFR, HER2, and BRAF). Indeed, mutations such as the most common FGFR3R248C/S249C occur in the ligand-binding domain, mimicking a constitutive extracellular signaling. FGFR2/3 rearrangements lead to increased signaling by means of protein dimerization promoted by partner genes involved. For FGFR amplification, only in the setting of high level gene amplification (i.e., ≥16 copies), leading to mRNA and protein overexpression, FGFR represents a selective marker of drug efficacy, as in the case of FGFR2-amplified breast and gastric cancers (6). Aberrant signaling in FGFR-activated tumors is mediated by the intracellular pathways shared with other oncogenes (i.e., RAS–MAPK and PI3K–AKT–mTOR). These promote the proliferation, survival, and aggressiveness of FGFR-dependent tumor cells (see section “Selective FGFR Inhibitors”).

FGFR1 is frequently amplified in squamous non–small cell lung cancer (NSCLC, 20%–25%) and breast cancer (15%; refs. 7, 8) but mutated in 18% of midline gliomas (9). FGFR2 is mainly activated by gene fusions in intrahepatic cholangiocarcinomas (iCCA, 15%) but by mutations in 10% of endometrial tumors (10, 11). FGFR3 is affected by mutations in urothelial carcinomas (up to 20% in the metastatic setting; refs. 12, 13); gene fusions (mainly FGFR3-TACC3) are present in glioblastomas and gliomas (3%; refs. 14, 15), as well as in bladder cancer (2%–3%; ref. 16).

Although approximately 7% of all cancers harbor an FGFR aberration (17), molecular screening to identify patients suitable for treatment with selective FGFR inhibitors is currently seeking for histotypes in which FGFR inhibition would provide major clinical benefit (i.e., cases with gene fusions and mutations; ref. 18). Indeed, if the majority of alterations are FGFR amplifications (66%), the lack of robust efficacy data of FGFR inhibition in FGFR-amplified cancers has hampered the interest in this setting. FGFR amplification may represent an imperfect biomarker for FGFR-TKI for several reasons. First, FGFR gene amplification may not be accompanied by FGFR overexpression; second, amplification may not represent the pivotal event for cancer cells growth and invasion; third, FISH cutoffs for a definition of FGFR amplification are still lacking. Definitions of FGFR amplification included the threshold of 6 to 10 gene copies per cell, with a variable cutoff between studies (7, 19–21), the ratio FGFR gene/centromere probes, ranging from 2 to 3 (22–27), and the notion of gene clusters in populations of tumor cells (23, 24). In addition, FGFR amplifications in squamous NSCLC, breast, and gastric malignancies may be accompanied by the concomitant presence of molecular alterations involving cyclins or cyclin-dependent kinases, PI3K signaling, and amplification with high expression of IGF1R, HER2, and EGFR (22, 27), thus questioning the FGFR dependency of those tumor cells.

First-generation FGFR-TKI (e.g., anlotinib, ponatinib, dovitinib, lucitanib, lenvatinib, and nintedanib) operate as multi-target inhibitors, including FGFR among their wide range of hits (VEGFR1/3, KIT, and RET among others). This led to the lack of a profound anti-FGFR inhibition and to the occurrence of deleterious adverse events (e.g., the disappointing results of dovitinib in FGFR2-mutated endometrial cancer; ref. 11).

The refinement in the molecular selection of patients with FGFR-driven cancer has been accompanied by the recent development of inhibitors specifically and selectively acting on the tyrosine kinase domain of FGFR family members, counteracting their phosphorylation at nanomolar concentrations (Table 1). Namely, this review focuses on erdafitinib (JNJ-42756493; Janssen-Johnson & Johnson), infigratinib (BGJ398; BridgeBio/QED Therapeutics), pemigatinib (INCB054828; Incyte), rogaratinib (BAY1163877; Bayer), derazantinib (ARQ 087; Basilea Pharmaceutica), futibatinib (TAS-120; Tahio), LY2874455 (Eli Lilly), AZD4547 (AstraZeneca), Debio 1347 (Debiopharm Group), and fisogatinib (BLU-554; Blueprint Medicines). These inhibitors are being developed almost simultaneously through clinical trials. Schematically, erdafitinib and rogaratinib have been mainly developed in urothelial cancer, infigratinib and futibatinib in cholangiocarcinoma, and pemigatinib in both tumor types. With regard to the other inhibitors, the initial results from clinical trials are too preliminary to allow direct comparisons between individual drugs and specific tumor types. With the exception of futibatinib all the inhibitors are characterized by a reversible, ATP-competitive binding to FGFR kinase domain. The irreversible, covalent binding of futibatinib is likely to account for its activity in patients with FGFR2-rearranged cholangiocarcinoma after progression to reversible FGFR-TKI (see section “Resistance to FGFR Inhibitors”). Of note, due to its promising clinical activity and efficacy (see section “Urothelial Cancer”), FDA has recently granted accelerated approval to erdafitinib for urothelial carcinomas harboring FGFR2–3 alterations (28).

Table 1.

Selective FGFR inhibitors in clinical development.

DrugTargetsInhibition typeDose adopted in ongoing trialsPhase of ongoing trials
Erdafitinib (JNJ-42756493) FGFR1–4 Reversible 8 mg dailya III 
Infigratinib (BGJ398) FGFR1–3 Reversible 125 mg daily 3-week on/1-week off III 
Pemigatinib (INCB054828) FGFR1–3 Reversible 13.5 mg daily III 
Rogaratinib (BAY1163877) FGFR1–3 Reversible 800 mg twice daily III 
Futibatinib (TAS-120) FGFR1–4 Irreversible 20 mg daily II 
Derazantinib (ARQ 087) FGFR1–4 Reversible 300 mg daily II 
LY2874455 FGFR1-4 Reversible 16 mg twice daily 
AZD4547 FGFR1–3 Reversible 80 mg twice daily II 
Debio 1347 (CH5183284) FGFR1–3 Reversible 80 mg daily II 
Fisogatinib (BLU-554) FGFR4 Irreversible 600 mg daily I extension 
DrugTargetsInhibition typeDose adopted in ongoing trialsPhase of ongoing trials
Erdafitinib (JNJ-42756493) FGFR1–4 Reversible 8 mg dailya III 
Infigratinib (BGJ398) FGFR1–3 Reversible 125 mg daily 3-week on/1-week off III 
Pemigatinib (INCB054828) FGFR1–3 Reversible 13.5 mg daily III 
Rogaratinib (BAY1163877) FGFR1–3 Reversible 800 mg twice daily III 
Futibatinib (TAS-120) FGFR1–4 Irreversible 20 mg daily II 
Derazantinib (ARQ 087) FGFR1–4 Reversible 300 mg daily II 
LY2874455 FGFR1-4 Reversible 16 mg twice daily 
AZD4547 FGFR1–3 Reversible 80 mg twice daily II 
Debio 1347 (CH5183284) FGFR1–3 Reversible 80 mg daily II 
Fisogatinib (BLU-554) FGFR4 Irreversible 600 mg daily I extension 

aTo be escalated to 9 mg daily at day 14 if plasma phosphate levels < 5.5 mg/dL (pharmacodynamically guided dose escalation).

In addition to the mentioned TKI, FGFR3-directed mAbs have also been developed, with promising signs of activity of vofatmab (B-701) combined with docetaxel in pretreated urothelial tumors (29), a setting in which the association of vofatmab and pembrolizumab is also envisaged (NCT03123055).

Activity data in early studies

The initial evidence of the activity and safety profile of selective FGFR inhibitors from phase I studies are summarized in Supplementary Table S1. In the dose-escalating phases, pretreated patients for whom no further standard therapies could be recommended were included, regardless of tumor type and molecular status, in line with an “all-comers” way. For all those selective inhibitors, almost no responses were observed in patients without any FGFR molecular alteration (erdafitinib, refs. 30–32; infigratinib, ref. 24; derazantinib, ref. 33; LY2874455, ref. 25; AZD4547, refs. 26, 27, 34, 35; pemigatinib, ref. 36; and rogaratinib, ref. 37).

In contrast, in the phase I dose-finding study of Debio 1374, patient enrollment was molecularly oriented toward the selection of diseases harboring FGFR1–3 alterations regardless of tumor type (38). Of the 58 patients exposed to increasing doses of the drug, 6 experienced partial responses (PR) and 16 stable diseases (SD), providing evidence of Debio 1374 activity in molecularly selected patients across histologies (Table 2), with a predilection for FGFR mutations or fusions (38). Importantly, 9 of 43 centrally rescreened cases turned out to be negative after local screening for FGFR activations, underlying the importance of a reliable molecular diagnostic process to select accurately the patient population (38).

Table 2.

Activity and efficacy data of selective FGFR inhibitors in urothelial tumors.

mPFS (mo)mDOR (mo)mOS (mo)
ReferencePatients treatedObjective responsesSDs(95% CI)(95% CI)(95% CI)
Erdafitinib phase I patients with UC 27 FGFRpos 12/30 (40%) among all UC FGFRposFGFRmut/fused: 5.1 5.6 — 
Balheda and colleagues, Clin Cancer Res 2019 (31) (17 FGFR3mut, 11 FGFRfused, 1 FGFRmut+fused) of 30 UC 12/26 (46%) among FGFRpos 4/26 (15%)    
NCT01703481       
Erdafitinib phase II FGFRmut/fused A: 8 (24%) A: 16 (49%) A: 4 A: 12.6 1-year survival: 
Loriot and colleagues, ASCO GU 2018 (13) A: 10 mg intermittent: 33 B: 27 (35%) B: 30 (39%) B: 5.1 B: 4.9 A: 31% 
NCT02365597 B: 6 mg daily: 78     B: 32% 
Erdafitinib phase II 8 mg dailya 99 FGFRpos 40 (40%) 39 (39%) 5.5 (4.2–6) 5.6 (4.2–7.2) 13.8 (9.8-NR) 
Loriot and colleagues, New Engl J Med 2019 (39) 74 (75%) FGFR3mut      
NCT02365597 25 (25%) FGFR2/3fused      
Infigratinib phase I expansion cohort 67 FGFR3pos 17 (25%) 26 (39%) 3.7 (3.1–5.4) — 7.7 (5.6–11.6) 
Pal and colleagues, Cancer Discov 2018 (40)       
NCT01004224       
Pemigatinib phase II Cohort A: FGFR3mut/fused: 61 A: 13 (21%) A: 22 (36%) A: 4.1 (3.0–5.6) A: 6.2 (4.2–8.4) — 
Necchi and colleagues, ESMO 2018 (41) Cohort B: other FGFR/FGF driver: 42 B: 1 (2%) FGF10ampli B: 10 (24%) B: 2.0 (1.9–2.1)   
NCT02872714       
Rogaratinib phase I expansion cohort 52 high FGFR1-3 mRNAb 12 (24%) 25 (49%) 100 days (57–143) — — 
Schuler and colleagues, Lancet Oncol 2019 (37) (48 high FGFR3; among these, 16 FGFR3mut and 2 FGFR3fused7 without FGFR genetic alterations     
NCT01976741       
Futibatinib phase I patients with UC 20 FGF/FGFR altered UC 3 (15%) 9 (45%) — — — 
Meric-Bernstam and colleagues, AACR 2019 (71)       
NCT02052778       
mPFS (mo)mDOR (mo)mOS (mo)
ReferencePatients treatedObjective responsesSDs(95% CI)(95% CI)(95% CI)
Erdafitinib phase I patients with UC 27 FGFRpos 12/30 (40%) among all UC FGFRposFGFRmut/fused: 5.1 5.6 — 
Balheda and colleagues, Clin Cancer Res 2019 (31) (17 FGFR3mut, 11 FGFRfused, 1 FGFRmut+fused) of 30 UC 12/26 (46%) among FGFRpos 4/26 (15%)    
NCT01703481       
Erdafitinib phase II FGFRmut/fused A: 8 (24%) A: 16 (49%) A: 4 A: 12.6 1-year survival: 
Loriot and colleagues, ASCO GU 2018 (13) A: 10 mg intermittent: 33 B: 27 (35%) B: 30 (39%) B: 5.1 B: 4.9 A: 31% 
NCT02365597 B: 6 mg daily: 78     B: 32% 
Erdafitinib phase II 8 mg dailya 99 FGFRpos 40 (40%) 39 (39%) 5.5 (4.2–6) 5.6 (4.2–7.2) 13.8 (9.8-NR) 
Loriot and colleagues, New Engl J Med 2019 (39) 74 (75%) FGFR3mut      
NCT02365597 25 (25%) FGFR2/3fused      
Infigratinib phase I expansion cohort 67 FGFR3pos 17 (25%) 26 (39%) 3.7 (3.1–5.4) — 7.7 (5.6–11.6) 
Pal and colleagues, Cancer Discov 2018 (40)       
NCT01004224       
Pemigatinib phase II Cohort A: FGFR3mut/fused: 61 A: 13 (21%) A: 22 (36%) A: 4.1 (3.0–5.6) A: 6.2 (4.2–8.4) — 
Necchi and colleagues, ESMO 2018 (41) Cohort B: other FGFR/FGF driver: 42 B: 1 (2%) FGF10ampli B: 10 (24%) B: 2.0 (1.9–2.1)   
NCT02872714       
Rogaratinib phase I expansion cohort 52 high FGFR1-3 mRNAb 12 (24%) 25 (49%) 100 days (57–143) — — 
Schuler and colleagues, Lancet Oncol 2019 (37) (48 high FGFR3; among these, 16 FGFR3mut and 2 FGFR3fused7 without FGFR genetic alterations     
NCT01976741       
Futibatinib phase I patients with UC 20 FGF/FGFR altered UC 3 (15%) 9 (45%) — — — 
Meric-Bernstam and colleagues, AACR 2019 (71)       
NCT02052778       

Abbreviations: 95% CI, 95% confidence interval; mDOR, median duration of response; mo, months; mOS, median OS; mPFS, median PFS; NR, not reached; UC, urothelial carcinomas.

aDose pharmacodynamically escalated to 9 mg daily if target phosphate levels < 5.5 mg/dL after 14 days of treatment.

bRNAscope and NanoString analysis on fresh or archival specimens.

Altogether, these early-phase studies highlighted the importance of an accurate molecular selection of patients in targeted therapy trials, revealing FGFR mutations and fusions as strong predictors of FGFR-TKI clinical activity, compared with FGFR amplification.

Histology-oriented trials

Expansion cohorts of phase I and II trials of selective FGFR inhibitors included specific histologies with mandatory FGFR molecular selection. As in early-stage studies, patients enrolled were mainly heavily pretreated (≥third line in about 50% of the cases).

Urothelial cancer

Five FGFR-TKI (erdafitinib, infigratinib, pemigatinib, rogaratinib, and futibatinib) showed activity in patients with urothelial carcinomas (Table 2). After the promising results of the phase I expansion cohort (31), the phase II study of erdafitinib included 99 patients with FGFR-altered urothelial carcinomas (74 with FGFR3 mutations and 25 with a FGFR2 or 3 fusion, evaluated in a central laboratory) treated at the “selected regimen” of 8 mg daily. Responses and SD occurred in 40% and 39% of the cases, respectively; of interest, 49% of patients harboring FGFR-mutated tumors achieved disease response, compared with 16% among the FGFR-fusion group. A median progression-free survival (PFS) of 5.5 months lead to a remarkable median overall survival of 13.8 months, taking into account the pretreatment setting of these patients with urothelial carcinomas (39). These data therefore prompted the FDA-accelerated approval of erdafitinib for urothelial carcinomas (28).

With the limitation of a reduced number of patients treated, slightly inferior results of activity and efficacy were observed for infigratinib [objective response rate (ORR) 25%, median PFS 3.7 months] and pemigatinib (ORR 21%, median PFS 4.1 months) in FGFR3-altered cases (40, 41). Similar activity results were observed with rogaratinib in patients selected with high expression of FGFR1–3 mRNA (37).

The best results in terms of activity and efficacy have been observed when the detection of specific FGFR alterations, such as FGFR3 mutations and FGFR2 or 3 fusions (39), was required to receive targeted treatment. Inclusion of less defined FGFR abnormalities (FGFR-positive or -altered tumors) led to inferior results (Table 2).

Cholangiocarcinoma

Infigratinib, pemigatinib, derazantinib, futibatinib, and erdafitinib were investigated in patients with cholangiocarcinoma (Table 3). Disease responses were almost exclusively observed in FGFR2-rearranged tumors, with ORR reaching 21% (derazantinib; ref. 42), 30% (infigratinib; refs. 43, 44), 40% (pemigatinib and futibatinib; refs. 45–47), with the outlier 67% for erdafitinib in the Asian trial [with 100% of Disease Control Rate (DCR)] requiring however validation with a wider patient population (only 9 patients evaluated; ref. 48). Even more marked than for urothelial carcinomas, tumor shrinkage was obtained in the large majority of patients with cholangiocarcinoma. While benefiting from FGFR-TKI, those cases did not always reach the threshold for PR, which is taken into account when evaluating drug activity in cholangiocarcinoma using RECIST criteria.

Table 3.

Activity and efficacy data of selective FGFR inhibitors in cholangiocarcinoma.

mPFS (mo)mDOR (mo)mDDC (mo)mOS (mo)
ReferencePatients treatedObjective responsesSDs(95% CI)(95% CI)(95% CI)(95% CI)
Infigratinib phase II 61 FGFRpos9 (15%) 37 (61%) 5.8 (4.3–7.6) 5.1 (3.9–7.4) 7.5 (5.6–7.6) — 
Javle and colleagues, J Clin Oncol 48 FGFR2fused All in FGFR2fused (19%) 31 (65%)     
2018 a (43) 8 FGFR2mut       
NCT02150967 8 FGFR1-3ampl       
Infigratinib phase II 71 FGFR2fused 22 (31%) 41 (58%) 6.8 (5.3–7.6) 5.4 (3.7–7.4)  12.5 (9.9–16.6) 
Javle and colleagues, ESMO 2018b (44)        
NCT02150967        
Erdafitinib phase I patients with CCA 11 FGFRpos3/11 (27%) 3/11 (27%) ≈5 11.4   
Balheda and colleagues, Clin Cancer Res 2019 (31) 8 FGFRfused       
NCT01703481 3 FGFRmut       
Derazantinib phase II 29 FGFR2fused 6 (21%) 18 (62%) 5.7 (4.0–9.2) 4.6 (2.3–8.9) 5.8 (5.3–8.4) Not reached (mFU 20 mo) 
Mazzaferro and colleagues, Br J Cancer 2018 (42)        
NCT03230318        
Pemigatinib phase II Cohort A: FGFR2fused: 47 A: 19 (40%) A: 21 (45%) A: 9.2 (6.44–NE) A: NE (6.9–NE)  A: 15.8 
Hollebecque and colleagues, ESMO 2018 (45) Cohort B: Other FGF/FGFR driver: 22 B: 0 B: 10 (45%) B: 2.1 (1.2–6.80)   B: 6.8 
NCT02924376 Cohort C: No FGF/FGFR: 18 C: 0 C: 4 (22%) C: 1.7 (1.4–1.8)   C: 4 
Futibatinib phase I CCA data 45 FGFRpos (41 iCCC): FGFR2fused: 7/24 (25%) FGFR2fused: 15 (54%) FGFR2fused— — — 
(1) Tran and colleagues, 8 FGFR2fused FGFR2fusionneg: 3/17 (18%) FGFR2fusionneg7.4 (4.8–NC)c    
ESMO Asia 2018 (46) 13 (29%) FGFR-TKI pretreated All in FGFR2“rearranged” 10 (59%) FGFR2fusionneg   
(2) Meric-Bernstam and colleagues,  4/13 (31%) in FGFR-TKI pretreated (3 fused, 1 ampli)  6.8 (1.9–NC)c    
ESMO GI 2018 (47)        
NCT02052778        
Erdafitinib phase IIa Asian 34: 7/15 (47%) 5/15 (33%) 5.59 (1.91–12.65) 7.06 (3.6–12.2) — — 
Park and colleagues, ASCO 2019 (48) 15 FGFR2fused, 9 FGFR2mut, 7 FGFR3mut, FGFR1mut FGFRfused: 6/9 (67%) FGFRfused: 3/9 (33%) FGFRfused: 12.65 (3.15–19.38) FGFRfused: 7.29 (3.9–12.2)   
NCT02699606 out of 157 molecularly evaluable       
mPFS (mo)mDOR (mo)mDDC (mo)mOS (mo)
ReferencePatients treatedObjective responsesSDs(95% CI)(95% CI)(95% CI)(95% CI)
Infigratinib phase II 61 FGFRpos9 (15%) 37 (61%) 5.8 (4.3–7.6) 5.1 (3.9–7.4) 7.5 (5.6–7.6) — 
Javle and colleagues, J Clin Oncol 48 FGFR2fused All in FGFR2fused (19%) 31 (65%)     
2018 a (43) 8 FGFR2mut       
NCT02150967 8 FGFR1-3ampl       
Infigratinib phase II 71 FGFR2fused 22 (31%) 41 (58%) 6.8 (5.3–7.6) 5.4 (3.7–7.4)  12.5 (9.9–16.6) 
Javle and colleagues, ESMO 2018b (44)        
NCT02150967        
Erdafitinib phase I patients with CCA 11 FGFRpos3/11 (27%) 3/11 (27%) ≈5 11.4   
Balheda and colleagues, Clin Cancer Res 2019 (31) 8 FGFRfused       
NCT01703481 3 FGFRmut       
Derazantinib phase II 29 FGFR2fused 6 (21%) 18 (62%) 5.7 (4.0–9.2) 4.6 (2.3–8.9) 5.8 (5.3–8.4) Not reached (mFU 20 mo) 
Mazzaferro and colleagues, Br J Cancer 2018 (42)        
NCT03230318        
Pemigatinib phase II Cohort A: FGFR2fused: 47 A: 19 (40%) A: 21 (45%) A: 9.2 (6.44–NE) A: NE (6.9–NE)  A: 15.8 
Hollebecque and colleagues, ESMO 2018 (45) Cohort B: Other FGF/FGFR driver: 22 B: 0 B: 10 (45%) B: 2.1 (1.2–6.80)   B: 6.8 
NCT02924376 Cohort C: No FGF/FGFR: 18 C: 0 C: 4 (22%) C: 1.7 (1.4–1.8)   C: 4 
Futibatinib phase I CCA data 45 FGFRpos (41 iCCC): FGFR2fused: 7/24 (25%) FGFR2fused: 15 (54%) FGFR2fused— — — 
(1) Tran and colleagues, 8 FGFR2fused FGFR2fusionneg: 3/17 (18%) FGFR2fusionneg7.4 (4.8–NC)c    
ESMO Asia 2018 (46) 13 (29%) FGFR-TKI pretreated All in FGFR2“rearranged” 10 (59%) FGFR2fusionneg   
(2) Meric-Bernstam and colleagues,  4/13 (31%) in FGFR-TKI pretreated (3 fused, 1 ampli)  6.8 (1.9–NC)c    
ESMO GI 2018 (47)        
NCT02052778        
Erdafitinib phase IIa Asian 34: 7/15 (47%) 5/15 (33%) 5.59 (1.91–12.65) 7.06 (3.6–12.2) — — 
Park and colleagues, ASCO 2019 (48) 15 FGFR2fused, 9 FGFR2mut, 7 FGFR3mut, FGFR1mut FGFRfused: 6/9 (67%) FGFRfused: 3/9 (33%) FGFRfused: 12.65 (3.15–19.38) FGFRfused: 7.29 (3.9–12.2)   
NCT02699606 out of 157 molecularly evaluable       

Abbreviations: 95% CI, 95% confidence interval; CCA, cholangiocarcinoma; mDDC, median duration of disease control; mDOR, median duration of response; mFU, median follow-up; mo, months; mOS, median OS; mPFS, median PFS; NE, not evaluable.

aProtocol was then modified to limit enrollment only to cases harboring FGFR2 fusions.

bThis update includes patients with cholangiocarcinoma with FGFR2 fusions already presented in the published study in the upper line.

cMedian treatment time.

In the FGFR2-rearranged population, an extremely large variety of FGFR2 fusion partners were identified: beside BICC1 (30% of the cases), 30 additional partner genes were detected (45, 47). Of note, among this group of advanced cholangiocarcinoma, pemigatinib achieved the longest median PFS of 9.2 months (45).

The phase I trial of the covalent inhibitor futibatinib in patients with cholangiocarcinoma allowed the enrollment of patients who had already progressed on a reversible FGFR inhibitor. Notably, 4 of 13 patients (3 FGFR2-rearranged and 1 FGFR2-amplified) previously exposed to reversible inhibitors achieved a PR, supporting the utility of futibatinib in overcoming drug-specific resistance (see section “Ongoing Trials,” for second-line FGFR-TKI), owing likely to its covalent binding (see section “Resistance to FGFR Inhibitors;” refs. 46, 47).

Globally, the most relevant results were observed when the molecular screening focused on FGFR2 rearrangements, toward which current trials are now being addressed (see section “Ongoing Trials”).

Other tumor types

AZD4547 phase II trials have been dedicated to cancers known to harbor FGFR gene abnormalities, namely squamous NSCLC, breast, and gastric tumors (refs. 6, 20–23, 26, 49; Supplementary Table S2). Nevertheless, when selecting for FGFR amplification (the most frequent FGFR alteration present in the cited malignancies), ORR rarely exceeded 10% across the reports, as seen also in FGFR1-amplified squamous NSCLC treated in the dedicated expansion cohort of infigratinib phase I study (ref. 24; Supplementary Table S2). These disappointing results confirmed the weakness of FGFR amplification as a biomarker for FGFR inhibitors sensitivity. As elegantly pointed out by Pearson and colleagues in their translational work, AZD4547 activity relies on the level of FGFR amplification (6).

Rogaratinib revealed promising efficacy data in patients with head and neck cancer overexpressing FGFR1–3 at the mRNA level but will require a larger number of patients and a longer follow-up for validation (50). In NSCLC nevertheless, only 2 responses out of 36 evaluable patients with high FGFR1–3 mRNA were observed, suggesting that more precise biomarkers are required (ref. 51; Supplementary Table S2).

FGFR4 is emerging as a potential target in hepatocellular carcinoma (HCC), through FGF19 overexpression (30% of HCC; refs. 52, 53). In the phase I trial of the highly selective FGFR4 inhibitor fisogatinib no responses were observed among the 29 HCC cases not tested or negative for FGF19 IHC. Six of 38 (16%) IHC-positive patients experienced disease response and additional 20 patients (53%) remained stable, achieving a median PFS of 3.7 months (ref. 54; Supplementary Table S2).

Altogether, these histology-oriented trials validated the mandatory aspect of FGFR molecular selection of patients and the urgent need for a better definition of the FGFR amplification or overexpression status in cases without mutation of fusion.

Safety and toxicity

The spectrum of adverse events accompanying selective FGFR inhibitors is thought to be due to their specific mechanism of action, counteracting the physiologic role of the FGF/FGFR axis. Table 4 reports the incidence of the most frequent events occurring in FGFR-TKI clinical trials but, considering the limited number of patients exposed, updated results will be required to characterize inhibitor-specific toxicities.

Table 4.

Toxicity rates across trials with selective FGFR inhibitors.

Toxicity
Drug and diseaseAll toxicitiesHyperPhHypoPhStomatitisDiarrheaDigestive tract eventsNail eventsHand–foot syndromeOcular events
ReferencePatients numberAny GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3
Erdafitinib UC 99 100% 46% 77% 2% NR NR 58% 10% 51% 4% 30–40% 1% 18% 2% 23% 5% 19% 3% 
Loriot and colleagues, New Engl J Med 2019 (39) 
Infigratinib CCA 61 92% 41% 72% 16% 11% 5% 29% 7% 15% 3% 18% 1% 18% — 21% 5% 21% — 
Javle and colleagues, J Clin Oncol 2018 (44) 
Pemigatinib CCA/UC 89/108 93% NR 31% 1% 10% 6% 34% 7% 43% 3% 35% 3% NR NR 13% 1% 
Hollebecque/Necchi and colleagues, ESMO 2018 (41, 46) 
Rogaratinib UC 51 NR 45% — NR NR 61% 4% 29% 2% NR NR NR 
Joerger and colleagues, ASCO 2018 (42) 
Derazantinib CCA 29 93% 28% 76% 10% NR 7% 3% 21% — 45% 3% NR NR 41% 7% 
Mazzaferro and colleagues, Br J Cancer 2018 (43) 
AZD4547 NCI-MATCH Arm W 49 80% 49% NR NR 2% 22% 14% 20% — 24% 2% NR NR 6% NR 2% 
Chae and colleagues, ASCO 2018 (21) 
Toxicity
Drug and diseaseAll toxicitiesHyperPhHypoPhStomatitisDiarrheaDigestive tract eventsNail eventsHand–foot syndromeOcular events
ReferencePatients numberAny GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3Any GG ≥ 3
Erdafitinib UC 99 100% 46% 77% 2% NR NR 58% 10% 51% 4% 30–40% 1% 18% 2% 23% 5% 19% 3% 
Loriot and colleagues, New Engl J Med 2019 (39) 
Infigratinib CCA 61 92% 41% 72% 16% 11% 5% 29% 7% 15% 3% 18% 1% 18% — 21% 5% 21% — 
Javle and colleagues, J Clin Oncol 2018 (44) 
Pemigatinib CCA/UC 89/108 93% NR 31% 1% 10% 6% 34% 7% 43% 3% 35% 3% NR NR 13% 1% 
Hollebecque/Necchi and colleagues, ESMO 2018 (41, 46) 
Rogaratinib UC 51 NR 45% — NR NR 61% 4% 29% 2% NR NR NR 
Joerger and colleagues, ASCO 2018 (42) 
Derazantinib CCA 29 93% 28% 76% 10% NR 7% 3% 21% — 45% 3% NR NR 41% 7% 
Mazzaferro and colleagues, Br J Cancer 2018 (43) 
AZD4547 NCI-MATCH Arm W 49 80% 49% NR NR 2% 22% 14% 20% — 24% 2% NR NR 6% NR 2% 
Chae and colleagues, ASCO 2018 (21) 

Note: Futibatinib, LY2874455, Debio 1347, and fisogatinib only phase I dose-finding data available. Studies with the largest patient population were chosen for each inhibitor. Digestive tract events: constipation, nausea, vomiting, abdominal pain, and dyspepsia and ocular events: dry eye, vision blurred, conjunctivitis, visual acuity reduced, photophobia, corneal erosion, blepharitis, and serous retinal detachment.

Abbreviations: CCA, cholangiocarcinoma; G, grade; HyperPh, hyperphosphatemia; HypoPh, hypophosphatemia; NR, not reported; UC, urothelial carcinomas.

The most commonly observed toxicities included hyper-phosphoremia, fatigue, alopecia, dry skin and mouth with stomatitis, hand–foot syndrome (Fig. 1), diarrhea, and other gastrointestinal events (nausea, vomiting, abdominal pain, and constipation; Table 4). In particular, FGFR inhibition disrupts phosphate homeostasis physiologically maintained by FGF23 (secreted from the bone and acting on the kidney), whose high levels induced by FGFR-TKI administration lead to hyper-phosphoremia (55).

Figure 1.

Nail toxicity and hand–foot syndrome in a patient treated with erdafitinib. A and D, Baseline. B, C, and E, Onychodystrophy appearing few weeks from treatment beginning and persisting over time (more than 1 year). F and G, Progressive worsening of hand–foot syndrome.

Figure 1.

Nail toxicity and hand–foot syndrome in a patient treated with erdafitinib. A and D, Baseline. B, C, and E, Onychodystrophy appearing few weeks from treatment beginning and persisting over time (more than 1 year). F and G, Progressive worsening of hand–foot syndrome.

Close modal

Biological abnormalities, namely aspartate aminotransferase, alanine aminotransferase, and lipase increase, were often detected. Adverse events affecting nails (onychodystrophy and nail loss) and eyes (dryness and central serous retinopathy) were frequently reported (Table 4). Globally, limiting toxicities were represented by fatigue, stomatitis, and nail events (Fig. 1; ref. 56). In the clinical trials, toxicity was managed by dose interruption or reduction and supportive therapies (e.g., low-phosphate diet and administration of phosphate binders such as sevelamer for hyperphosphoremia) but rarely led to treatment discontinuation.

Supplementary Table S3 resumes the ongoing trials evaluating selective FGFR inhibitors in molecularly selected populations, in single-arm or randomized trials, either as monotherapy or in combination with other agents (cytotoxic treatments, targeted therapies, and immune checkpoint inhibitors). Information gathered in Supplementary Table S3 has been obtained from clinicaltrials.gov (checked last time on June 2019) and abstracts presented in international meetings. Of note, phase III clinical trials are currently ongoing to compare selective FGFR-TKI with standard-of-care chemotherapy or immunotherapy, both in second- and first-line setting in urothelial carcinomas and cholangiocarcinoma. Considering the recently established efficacy of immune checkpoint inhibitors in urothelial carcinomas, potential synergies are being envisaged. Anti-PD-1/PD-L1 antibodies are indeed associated with erdafitinib (NCT03473743, pretreated patients) or rogaratinib (first-line).

Clinical data on molecular mechanisms of resistance to specific FGFR inhibitors are limited thus far. Preclinical work has mostly been dedicated toward FGFR-amplified models and mainly defines the potential involvement of bypass tracks activation in both primary and acquired resistance (57–60). However, on-target mechanisms (e.g., the onset of FGFR mutations) can also confer acquired resistance to FGFR-TKI in patients. Of note, in several experiences the longitudinal evaluation of circulating tumor DNA (ctDNA) of patients receiving FGFR inhibitors was able to catch the multiplicity of FGFR mutations occurring in the tyrosine kinase domain at disease progression.

Goyal and colleagues analyzed longitudinal blood samples and tumor biopsies at progression of 3 patients with FGFR2-rearranged cholangiocarcinoma receiving infigratinib (61). Blood and biopsy sequencing revealed the acquisition of multiple resistant mutations in the FGFR2 kinase domain (N549H/K, V564F, E565A, L617V, K641R, and K659M), among which the gatekeeper V564F was shared by the 3 patients. Conversely, a limited number of FGFR2 mutations were detected in tumor tissue samples, thus corroborating the utility of liquid biopsies to capture the heterogeneity of resistance mechanisms (62). A broader autopsy analysis, besides confirming the presence of FGFR2 mutations, allowed the detection of a PTEN deletion and a PI3KCA-activating mutation (Q546R) suggesting the potential involvement of the PI3K/AKT pathway in FGFR resistance (63). In their related in vitro studies, the authors defined the functional role of each FGFR2 mutation observed in clinical samples and provided evidence that LY2874455 is an active inhibitor across a range of secondary mutations (61). This is in line with the biochemical and pharmacologic study provided by Wu and colleagues, determining LY2874455 as an active FGFR inhibitor in presence of gatekeeper mutations (i.e., FGFR1V561M, FGFR2V564F, FGFR3V555M, and FGFR4V550L; refs. 64, 65). Crystallographic studies of LY2874455 interaction with FGFR revealed that the inhibitor positions away from the gatekeeper residues, avoiding the steric clash precluding drug binding usually induced by gatekeeper mutations (64).

Goyal and colleagues later demonstrated futibatinib efficacy in 4 patients with FGFR2-rearranged iCCA after acquired resistance to infigratinib or Debio 1347. Using preclinical models, the authors demonstrated the futibatinib activity against multiple FGFR2 mutations conferring resistance to the two others FGFR-TKI (M537I, N549K/H, E565A, L617V, K659M, H682L, and K714R; ref. 66). Differently from LY2874455, futibatinib lacks activity against FGFR2 gatekeeper–mutant V564F, due to a steric clash. Futibatinib-irreversible interaction with the ATP-binding pocket is the basis for its activity in the occurrence of the cited mutations that shift FGFR2 tyrosine kinase domain conformation in a constitutively active state, only targetable by a covalent inhibitor (66, 67).

Similar detection of polyclonal FGFR mutations in ctDNA at progression, mainly in patients with FGFR2-rearanged cholangiocarcinoma and in patients with FGFR3-mutated urothelial carcinomas receiving infigratinib or erdafitinib were reported in other studies (12, 40, 68). Remarkably, futibatinib is thus far the only inhibitor able to resensitize in the clinic tumors resistance to reversible FGFR inhibitors, likely owing to its covalent binding. The following emergence of FGFR2N549K, FGFR2V652L, and FGFR2V564F/L mutations at progression on futibatinib nevertheless testifies the complexity of the mutations arising in FGFR kinase domain, responsible for resistance to differential inhibitors (69).

Since the initial targeting of FGFR with multi-target inhibitors evaluated in clinical trials lacking a strict molecular selection, meaningful improvements have been provided in this field. Specific FGFR inhibition represents a concrete reality in the field of precision medicine, thanks to the refinement of FGFR targetable alterations (i.e., fusions and mutations) and to the availability of selective inhibitors. Waiting for definitive results from randomized phase III trials, the benefit driven from FGFR inhibitors has been acknowledged for urothelial cancer (28) and it is expected to be fully proven for cholangiocarcinoma in a near future.

FGFR activations should be screened across a multitude of malignancies (17), with a particular mention for brain tumors given the frequency of these aberrations (9, 15), the initial signs of activity of selective inhibitors (30), the availability of specific clinical trials (NCT01975701; Supplementary Table S3), and the urgent need for effective treatment options. The narrowing of molecular selection, allowing only tumors harboring functionally validated FGFR activations to be included into clinical trial, has provided the better results and accelerated the approval of erdafitinib in urothelial carcinomas (28, 39). On the other hand, the recent widening of molecular next-generation sequencing (NGS) diagnostics in the clinical setting, should speed up the implementation of FGFR molecular screening on a daily basis, leading to treatment with selective FGFR inhibitors as a common clinical practice (39).

Phase I trial of Debio 1347 serve as an example of the importance of an accurate molecular diagnostics, with 21% of the cases, initially considered as FGFR altered according to local assessment, but turning out to lack FGFR aberration, therefore undermining the global results of the study (38). These discrepancies may be explained by intratumoral heterogeneity, sampling errors, or technical issues. Among the latter, analysis algorithms evolution during the trial could induce differences in annotating complex fusions and the different accuracy in detecting gene amplifications between FISH and NGS have been evoked by study investigators (38).

Molecular screening should identify cases harboring FGFR alterations with known functional significance. In addition, peculiar attention should be paid toward potential passenger events, especially in the case of hypermutated tumors, or benign germline polymorphisms not filtered from the somatic data.

In contrast to fusions and mutations, FGFR amplification, frequent in squamous NSCLC, breast, and gastric cancers, is yet to be defined as a reliable biomarker for response to FGFR inhibitors. A careful selection of patients whose tumors harbor biologically meaningful levels of FGFR amplifications (i.e., cases truly relying on FGFR for tumor growth), would potentially help to detect the cases suitable for specific FGFR inhibition (18). In this sense, the refinement of diagnostics approaches and of thresholds for defining significant levels of FGFR amplification should be accompanied by analysis of mRNA and/or protein quantification.

Conducting randomized phase III trials in specific histologies with FGFR aberration may be challenging, given the gap between the number of patients required for those studies and the relative rarity of precise molecular-altered tumor types. In this sense, phase II basket trials may provide sufficient evidence for activity and efficacy of specific FGFR-TKI with a limited number of patients included for each tumor type (70). Nevertheless, the differential signals of activity of FGFR inhibitors against diverse FGFR activations (i.e., mutations vs. fusions), as well as in different tumor types (Tables 2 and 3; Supplementary Table S2), should be mentioned. Each TKI may indeed retain specific activity toward precise targets, influenced in addition by the cellular context in which they arise.

Goyal and colleagues provided the clinical evidence that, guided by biopsies and ctDNA, the sequencing of FGFR inhibitors can prolong the duration of benefit from FGFR inhibition in patients with FGFR-positive cancer (61, 66, 69). Thus far, futibatinib is the only inhibitor to have shown the potential of overcoming resistance to other, reversible FGFR-TKI in patients with FGFR2-rearranged cholangiocarcinoma (46, 47). As futibatinib, LY2874455 has been proven to be active in vitro against FGFR mutants engendering resistance to other inhibitors (61, 64). Nevertheless, futibatinib development is ongoing (Table 3; Supplementary Table S3), LY2874455 lacks recent updates (Supplementary Table S3). A clear functional understanding of the activity of each of the 10 specific FGFR inhibitors against all activating alterations and acquired resistance mechanisms will be key to define the optimal treatment strategies with sequential FGFR-TKI to overcome resistance, likely to be accompanied by a combination approach in the case of bypass activation.

The current estimations of PFS and duration of response reported for selective FGFR inhibitors (Tables 2 and 3), still encouraging, are not yet overlapping to the prolonged disease control achieved with other targeted agents in different histologies (e.g., lung cancer and melanoma). Potentially due to an intrinsic aggressiveness of FGFR-driven malignancies, these suboptimal outcomes will be hopefully extended by a wider comprehension of drug resistance and the overcoming strategies. In addition, the molecularly driven tumors for which resistance mechanisms to TKI have already been widely studied (i.e., EGFR- and ALK-positive lung cancers) are characterized by the acquisition of single mutational events responsible for resistance, likely easing the efficacy of sequential inhibitors administration. In contrast, the initial reports regarding FGFR2-rearranged cancers (cholangiocarcinoma mainly) prove the development of multiple, polyclonal mutations arising from single tumors. In this sense, covering a multitude of diverse resistance mutations, with differential sensitivity to novel FGFR TKI, may turn out to be arduous. On the other hand, the upfront administration of more potent and widely active FGFR-TKI (once clearly identified) may prevent or delay the onset of polyclonal mutations responsible for disease progression. Ideally, the development of selective FGFR-TKI could retrace the generations of EGFR and ALK inhibitors in NSCLC, with drugs progressively more potent, active on a wide spectrum of resistance mutations and more specific. In this regard, the synthesis of new compounds should be accompanied by the design of optimized clinical trials.

In conclusion, specific FGFR inhibition is a relevant chapter in the field of precision medicine. The design of ongoing trials involving selective anti-FGFR drugs, as well as the recent approval of erdafitinib, will hopefully lead to a wide clinical availability. In this sense, it is essential to implement appropriate molecular screening tests for detecting FGFR alterations (mutations and fusions) or other means of activation (amplification accompanied by overexpression of the target) starting from tumors suitable for FGFR-targeted treatments (i.e., urothelial carcinomas and cholangiocarcinomas). Initiating basket clinical trials agnostic of tumor origin would hopefully fasten FGFR-TKI approvals for patients with FGFR-mutated/rearranged/overexpressed cancer (2).

A. Hollebecque is an employee/paid consultant for Incyte and Debiopharm. Y. Loriot is an employee/paid consultant for Janssen, Astellas, Roche, AstraZeneca, MSD, Bristol-Myers Squibb, and Seattle Genetics. No potential conflicts of interest were disclosed by the other authors.

The work of Francesco Facchinetti is supported by a grant from Philanthropia – Lombard Odier Foundation. The work of Luc Friboulet is supported by an ERC starting grant (agreement no. 717034).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Turski
ML
,
Vidwans
SJ
,
Rodon
J
,
Kurzrock
R
,
Munoz
J
,
Subbiah
V
, et al
Genomically driven tumors and actionability across histologies: BRAF-mutant cancers as a paradigm
.
Mol Cancer Ther
2016
;
15
:
533
47
.
2.
Drilon
A
,
Laetsch
TW
,
Kummar
S
,
DuBois
SG
,
Lassen
UN
,
Demetri
GD
, et al
Efficacy of larotrectinib in TRK fusion–positive cancers in adults and children
.
N Engl J Med
2018
;
378
:
731
9
.
3.
Dienstmann
R
,
Rodon
J
,
Prat
A
,
Perez-Garcia
J
,
Adamo
B
,
Felip
E
, et al
Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors
.
Ann Oncol
2014
;
25
:
552
63
.
4.
Touat
M
,
Ileana
E
,
Postel-Vinay
S
,
André
F
,
Soria
JC
. 
Targeting FGFR signaling in cancer
.
Clin Cancer Res
2015
;
21
:
2684
94
.
5.
Katoh
M
. 
Fibroblast growth factor receptors as treatment targets in clinical oncology
.
Nat Rev Clin Oncol
2019
;
16
:
105
22
.
6.
Pearson
A
,
Smyth
E
,
Babina
IS
,
Herrera-Abreu
MT
,
Tarazona
N
,
Peckitt
C
, et al
High-level clonal FGFR amplification and response to FGFR inhibition in a translational clinical trial
.
Cancer Discov
2016
;
6
:
838
51
.
7.
Weiss
J
,
Sos
ML
,
Seidel
D
,
Peifer
M
,
Zander
T
,
Heuckmann
JM
, et al
Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer
.
Sci Transl Med
2010
;
2
:
62ra93
.
8.
Andre
F
,
Job
B
,
Dessen
P
,
Tordai
A
,
Michiels
S
,
Liedtke
C
, et al
Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array
.
Clin Cancer Res
2009
;
15
:
441
51
.
9.
Di Stefano
AL
,
Picca
A
,
Saragoussi
E
,
Berzero
G
,
Alentorn
A
,
Touat
M
, et al
Actionable targets involving FGF receptors in gliomas: molecular specificities, spatial distribution, clinical outcome and radiological phenotype
.
J Clin Oncol
2018
;
36
:
2005
.
10.
Verlingue
L
,
Malka
D
,
Allorant
A
,
Massard
C
,
Ferté
C
,
Lacroix
L
, et al
Precision medicine for patients with advanced biliary tract cancers: an effective strategy within the prospective MOSCATO-01 trial
.
Eur J Cancer
2017
;
87
:
122
30
.
11.
Konecny
GE
,
Finkler
N
,
Garcia
AA
,
Lorusso
D
,
Lee
PS
,
Rocconi
RP
, et al
Second-line dovitinib (TKI258) in patients with FGFR2-mutated or FGFR2-non-mutated advanced or metastatic endometrial cancer: a non-randomised, open-label, two-group, two-stage, phase 2 study
.
Lancet Oncol
2015
;
16
:
686
94
.
12.
Chen
Y
,
Park
JO
,
Su
W
,
Oh
D
,
Kim
K
,
Feng
Y
, et al
Preliminary results of a Ph2a study to evaluate the clinical efficacy and safety of erdafitinib in Asian patients with biomarker-selected advanced cholangiocarcinoma (CCA)
.
Ann Oncol
2018
;
29
:
viii205
70
.
13.
Loriot
Y
,
Necchi
A
,
Park
SH
,
García-Donas
J
,
Huddart
RA
,
Burgess
EF
, et al
Erdafitinib (JNJ-42756493), a pan-fibroblast growth factor receptor inhibitor, in patients with metastatic or unresectable urothelial carcinoma and FGFR alterations
.
J Clin Oncol
2018
;
36
:
411
.
14.
Singh
D
,
Chan
JM
,
Zoppoli
P
,
Niola
F
,
Sullivan
R
,
Castano
A
, et al
Transforming fusions of FGFR and TACC genes in human glioblastoma
.
Science
2012
;
337
:
1231
5
.
15.
Di Stefano
AL
,
Fucci
A
,
Frattini
V
,
Labussiere
M
,
Mokhtari
K
,
Zoppoli
P
, et al
Detection, characterization, and inhibition of FGFR-TACC fusions in IDH wild-type glioma
.
Clin Cancer Res
2015
;
21
:
3307
17
.
16.
Robertson
AG
,
Kim
J
,
Al-Ahmadie
H
,
Bellmunt
J
,
Guo
G
,
Cherniack
AD
, et al
Comprehensive molecular characterization of muscle-invasive bladder cancer
.
Cell
2017
;
171
:
540
56
.
17.
Helsten
T
,
Elkin
S
,
Arthur
E
,
Tomson
BN
,
Carter
J
,
Kurzrock
R
. 
The FGFR landscape in cancer: analysis of 4,853 tumors by next-generation sequencing
.
Clin Cancer Res
2016
;
22
:
259
67
.
18.
Leroy
L
,
Cousin
S
,
Italiano
A
. 
Genetic determinants of response to fibroblast growth factor receptor inhibitors in solid tumours
.
Eur J Cancer
2017
;
81
:
102
5
.
19.
André
F
,
Bachelot
T
,
Campone
M
,
Dalenc
F
,
Perez-Garcia
JM
,
Hurvitz
SA
, et al
Targeting FGFR with dovitinib (TKI258): preclinical and clinical data in breast cancer
.
Clin Cancer Res
2013
;
19
:
3693
702
.
20.
Aggarwal
C
,
Redman
MW
,
Lara
PN
 Jr
,
Borghaei
H
,
Hoffman
P
,
Bradley
JD
, et al
SWOG S1400D (NCT02965378), a phase II study of the fibroblast growth factor receptor inhibitor AZD4547 in previously-treated patients with fibroblast growth factor pathway-activated stage IV squamous cell lung cancer (Lung-MAP Sub-Study)
.
J Thorac Oncol
2019
;
14
:
1847
52
.
21.
Chae
YK
,
Vaklavas
C
,
Cheng
HH
,
Hong
F
,
Harris
L
,
Mitchell
EP
, et al
Molecular analysis for therapy choice (MATCH) arm W: phase II study of AZD4547 in patients with tumors with aberrations in the FGFR pathway
.
J Clin Oncol
2018
;
36
:
2503
.
22.
Paik
PK
,
Shen
R
,
Berger
MF
,
Ferry
D
,
Soria
JC
,
Mathewson
A
, et al
A phase Ib open-label multicenter study of AZD4547 in patients with advanced squamous cell lung cancers
.
Clin Cancer Res
2017
;
23
:
5366
73
.
23.
Van Cutsem
E
,
Bang
YJ
,
Mansoor
W
,
Petty
RD
,
Chao
Y
,
Cunningham
D
, et al
A randomized, open-label study of the efficacy and safety of AZD4547 monotherapy versus paclitaxel for the treatment of advanced gastric adenocarcinoma with FGFR2 polysomy or gene amplification
.
Ann Oncol
2017
;
28
:
1316
24
.
24.
Nogova
L
,
Sequist
LV
,
Perez Garcia
JM
,
Andre
F
,
Delord
JP
,
Hidalgo
M
, et al
Evaluation of BGJ398, a fibroblast growth factor receptor 1-3 kinase inhibitor, in patients with advanced solid tumors harboring genetic alterations in fibroblast growth factor receptors: results of a global phase I, dose-escalation and dose-expansion study
.
J Clin Oncol
2017
;
35
:
157
65
.
25.
Michael
M
,
Bang
YJ
,
Park
YS
,
Kang
YK
,
Kim
TM
,
Hamid
O
, et al
A phase 1 study of LY2874455, an oral selective pan-FGFR inhibitor, in patients with advanced cancer
.
Target Oncol
2017
;
12
:
463
74
.
26.
Arkenau
HT
,
Saggese
M
,
Hollebecque
A
,
Mathewson
A
,
Lemech
CR
,
Landers
D
, et al
A phase 1 expansion cohort of the fibroblast growth factor receptor (FGFR) inhibitor AZD4547 in patients (pts) with advanced gastric (GC) and gastroesophageal (GOJ) cancer
.
J Clin Oncol
2014
;
32
:
2620
.
27.
Kilgour
E
,
Ferry
D
,
Saggese
M
,
Arkenau
H-T
,
Rooney
C
,
Smith
NR
, et al
Exploratory biomarker analysis of a phase I study of AZD4547, an inhibitor of fibroblast growth factor receptor (FGFR), in patients with advanced solid tumors
.
J Clin Oncol
2014
;
15
:
11010
.
28.
U.S. Food and Drug Administration
. Available from: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm635906.htm.
29.
Necchi
A
,
Castellano
DE
,
Mellado
B
,
Pang
S
,
Urun
Y
,
Park
SH
, et al
Fierce-21: phase II study of vofatmab (B-701), a selective inhibitor of FGFR3, as salvage therapy in metastatic urothelial carcinoma (mUC)
.
J Clin Oncol
37
:
7s
, 
2019
(
suppl; abstr 409
).
30.
Tabernero
J
,
Bahleda
R
,
Dienstmann
R
,
Infante
JR
,
Mita
A
,
Italiano
A
, et al
Phase I dose-escalation study of JNJ-42756493, an oral pan-fibroblast growth factor receptor inhibitor, in patients with advanced solid tumors
.
J Clin Oncol
2015
;
33
:
3401
8
.
31.
Bahleda
R
,
Italiano
A
,
Hierro
C
,
Mita
AC
,
Cervantes
A
,
Chan
N
, et al
Multicenter phase I study of erdafitinib (JNJ-42756493), oral pan-fibroblast growth factor receptor inhibitor, in patients with advanced or refractory solid tumors
.
Clin Cancer Res
2019
;
25
:
4888
97
.
32.
Nishina
T
,
Takahashi
S
,
Iwasawa
R
,
Noguchi
H
,
Aoki
M
,
Doi
T
. 
Safety, pharmacokinetic, and pharmacodynamics of erdafitinib, a pan-fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitor, in patients with advanced or refractory solid tumors
.
Invest New Drugs
2018
;
36
:
424
34
.
33.
Papadopoulos
KP
,
El-Rayes
BF
,
Tolcher
AW
,
Patnaik
A
,
Rasco
DW
,
Harvey
RD
, et al
A phase 1 study of ARQ 087, an oral pan-FGFR inhibitor in patients with advanced solid tumours
.
Br J Cancer
2017
;
117
:
1592
9
.
34.
Andre
F
,
Ranson
M
,
Dean
E
,
Varga
A
,
van der Noll
R
,
Stockma
PK
, et al
Results of a phase I study of AZD4547, an inhibitor of fibroblast growth factor receptor (FGFR), in patients with advanced solid tumors. [abstract]
.
In
:
Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6–10
;
Washington, DC. Philadelphia (PA)
:
AACR
; 
2013
.
Abstract nr LB-145
.
35.
Saka
H
,
Kitagawa
C
,
Kogure
Y
,
Takahashi
Y
,
Fujikawa
K
,
Sagawa
T
, et al
Safety, tolerability and pharmacokinetics of the fibroblast growth factor receptor inhibitor AZD4547 in Japanese patients with advanced solid tumours: a phase I study
.
Invest New Drugs
2017
;
35
:
451
62
.
36.
Saleh
M
,
Gutierrez
M
,
Subbiah
V
,
Smith
DC
,
Féliz
L
,
Zhen
H
, et al
Preliminary results from a phase 1/2 study of INCB054828, a highly selective fibroblast growth factor receptor (FGFR) inhibitor, in patients (pts) with advanced malignancies. [abstract]
.
In
:
Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26–30
;
Philadelphia, PA. Philadelphia (PA)
:
AACR
; 
2018
.
Abstract nr A098
.
37.
Schuler
M
,
Cho
BC
,
Sayehli
CM
,
Navarro
A
,
Soo
RA
,
Richly
H
, et al
Rogaratinib in patients with advanced cancers selected by FGFR mRNA expression: a phase 1 dose-escalation and dose-expansion study
.
Lancet Oncol
2019
;
20
:
1454
66
.
38.
Voss
MH
,
Hierro
C
,
Heist
RS
,
Cleary
JM
,
Meric-Bernstam
F
,
Tabernero
J
, et al
A phase I, open-label, multicenter, dose-escalation study of the oral selective FGFR inhibitor Debio 1347 in patients with advanced solid tumors harboring FGFR gene alterations
.
Clin Cancer Res
2019
;
25
:
2699
707
.
39.
Loriot
Y
,
Necchi
A
,
Park
SH
,
Garcia-Donas
J
,
Huddart
R
,
Burgess
E
, et al
Erdafitinib in locally advanced or metastatic urothelial carcinoma
.
N Engl J Med
2019
;
381
:
338
48
.
40.
Pal
SK
,
Rosenberg
JE
,
Hoffman-Censits
JH
,
Berger
R
,
Quinn
DI
,
Galsky
MD
, et al
Efficacy of BGJ398, a fibroblast growth factor receptor 1-3 inhibitor, in patients with previously treated advanced urothelial carcinoma with FGFR3 alterations
.
Cancer Discov
2018
;
8
:
812
21
.
41.
Necchi
A
,
Pouessel
D
,
Leibowitz-Amit
R
,
Flechon
A
,
Gupta
S
,
Barthelemy
P
, et al
Interim results of Fight-201, a phase 2, open-label, multicenter study of INCB054828 dosed intermittently in patients with metastatic or surgically unresectable urothelial carcinoma (UC) harboring fibroblast growth factor (FGF)/FGF receptor (FGFR) genetic alterations (GA)
.
Ann Oncol
2018
;
29
:
viii303
31
.
42.
Mazzaferro
V
,
El-Rayes
BF
,
Droz Dit Busset
M
,
Cotsoglou
C
,
Harris
WP
,
Damjanov
N
, et al
Derazantinib (ARQ 087) in advanced or inoperable FGFR2 gene fusion-positive intrahepatic cholangiocarcinoma
.
Br J Cancer
2018
;
120
:
165
71
.
43.
Javle
M
,
Lowery
M
,
Shroff
RT
,
Weiss
KH
,
Springfeld
C
,
Borad
MJ
, et al
Phase II study of BGJ398 in patients with FGFR-altered advanced cholangiocarcinoma
.
J Clin Oncol
2018
;
36
:
276
82
.
44.
Javle
M
,
Kelley
RK
,
Roychowdhury
S
,
Weiss
KH
,
Abou-Alfa
GK
,
Macarulla
T
, et al
Updated results from a phase II study of infigratinib (BGJ398), an FGFR-selective tyrosine kinase inhibitor (TKI), in patients with previously-treated advanced cholangiocarcinoma containing FGFR2 fusions
.
Ann Oncol
2018
;
29
:
LBA 28
.
Available from:
https://oncologypro.esmo.org/Meeting-Resources/ESMO-2018-Congress/Updated-results-from-a-phase-II-study-of-infigratinib-BGJ398-a-selective-pan-FGFR-kinase-inhibitor-in-patients-with-previously-treated-advanced-cholangiocarcinoma-containing-FGFR2-fusions.
45.
Hollebecque
A
,
Borad
M
,
Sahai
V
,
Catenacci
DVT
,
Murphy
A
,
Vaccaro
G
, et al
Interim results of Fight-202, a phase 2, open-label, multicenter study of INCB054828 in patients with previously treated advanced/metastatic or surgically unresectable cholangiocarcinoma (CCA) with/without fibroblast growth factor (FGF)/FGF receptor (FGFR) genetic alterations
.
Ann Oncol
2018
;
29
:
Poster 756P. Available from:
https://oncologypro.esmo.org/Meeting-Resources/ESMO-2018-Congress/Interim-results-of-fight-202-a-phase-2-open-label-multicenter-study-of-INCB054828-in-patients-pts-with-previously-treated-advanced-metastatic-or-surgically-unresectable-cholangiocarcinoma-CCA-with-without-fibroblast-growth-factor-FGF-FGF-receptor.
46.
Tran
B
,
Meric-Bernstam
F
,
Arkenau
HT
,
Bahleda
R
,
Kelley
RK
,
Hierro
C
, et al
Efficacy of TAS-120, an irreversible fibroblast growth factor receptor (FGFR) inhibitor, in patients with cholangiocarcinoma and FGFR pathway alterations who were previously treated with chemotherapy and other FGFR inhibitors
.
Ann Oncol
2018
;
29
:
ix46
66
.
47.
Meric-Bernstam
F
,
Arkenau
HT
,
Tran
B
,
Bahleda
R
,
Kelley
RK
,
Hierro
C
, et al
Efficacy of TAS-120, an irreversible fibroblast growth factor receptor (FGFR) inhibitor, in cholangiocarcinoma patients with FGFR pathway alterations who were previously treated with chemotherapy and other FGFR inhibitors
.
Ann Oncol
2018
;
29
:
mdy149
.
48.
Park
JO
,
Feng
YH
,
Chen
YY
,
Su
WC
,
Oh
DY
,
Shen
L
, et al
Updated results of a phase IIa study to evaluate the clinical efficacy and safety of erdafitinib in Asian advanced cholangiocarcinoma (CCA) patients with FGFR alterations
.
J Clin Oncol
37
:
15s
, 
2019
(
suppl; abstr 4117
).
49.
Seckl
M
,
Badman
PD
,
Liu
X
,
MacPherson
IR
,
Zubairi
IH
,
Baird
RD
, et al
RADICAL trial: a phase Ib/IIa study to assess the safety and efficacy of AZD4547 in combination with either anastrozole or letrozole in ER positive breast cancer patients progressing on these aromatase inhibitors (AIs)
.
J Clin Oncol
2017
;
35
:
1059
.
50.
Joerger
M
,
Takahashi
S
,
Sayehli
CM
,
Slosarczyk
S
,
Navarro Mendivil
A
,
Cassier
PA
, et al
Phase I experience with rogaratinib in patients with head and neck cancer selected based on FGFR mRNA overexpression
.
Ann Oncol
2018
;
29
:
viii372
99
.
51.
Joerger
M
,
Cho
BC
,
Mach
N
,
Caballero
C
,
Soo
RA
,
Wirth
M
, et al
Early clinical experience with the pan-FGFR inhibitor rogaratinib in patients with non-small cell lung cancer selected based on FGFR mRNA expression levels
.
J Clin Oncol
37
:
15s
, 
2019
(
suppl; abstr e20661
).
52.
Schulze
K
,
Imbeaud
S
,
Letouzé
E
,
Alexandrov
LB
,
Calderaro
J
,
Rebouissou
S
, et al
Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets
.
Nat Genet
2015
;
47
:
505
11
.
53.
Harmer
NJ
,
Pellegrini
L
,
Chirgadze
D
,
Fernandez-Recio
J
,
Blundell
TL
. 
The crystal structure of fibroblast growth factor (FGF) 19 reveals novel features of the FGF family and offers a structural basis for its unusual receptor affinity
.
Biochemistry
2004
;
43
:
629
40
.
54.
Kim
R
,
Sarker
D
,
Macarulla
T
,
Yau
T
,
Choo
SP
,
Meyer
T
, et al
Phase 1 safety and clinical activity of BLU-554 in advanced hepatocellular carcinoma (HCC)
.
Ann Oncol
2017
;
28
:
v122
41
.
55.
Yanochko
GM
,
Vitsky
A
,
Heyen
JR
,
Hirakawa
B
,
Lam
JL
,
May
J
, et al
Pan-FGFR inhibition leads to blockade of FGF23 signaling, soft tissue mineralization, and cardiovascular dysfunction
.
Toxicol Sci
2013
;
135
:
451
64
.
56.
Bétrian
S
,
Gomez-Roca
C
,
Vigarios
E
,
Delord
JP
,
Sibaud
V
. 
Severe onycholysis and eyelash trichomegaly following use of new selective pan-FGFR inhibitors
.
JAMA Dermatology
2017
;
153
:
723
5
.
57.
Harbinski
F
,
Craig
VJ
,
Sanghavi
S
,
Jeffery
D
,
Liu
L
,
Sheppard
KA
, et al
Rescue screens with secreted proteins reveal compensatory potential of receptor tyrosine kinases in driving cancer growth
.
Cancer Discov
2012
;
2
:
948
59
.
58.
Bockorny
B
,
Rusan
M
,
Chen
W
,
Liao
RG
,
Li
Y
,
Piccioni
F
, et al
RAS–MAPK reactivation facilitates acquired resistance in FGFR1-amplified lung cancer and underlies a rationale for upfront FGFR–MEK blockade
.
Mol Cancer Ther
2018
;
17
:
1526
39
.
59.
Herrera-Abreu
MT
,
Pearson
A
,
Campbell
J
,
Shnyder
SD
,
Knowles
MA
,
Ashworth
A
, et al
Parallel RNA interference screens identify EGFR activation as an escape mechanism in FGFR3-mutant cancer
.
Cancer Discov
2013
;
3
:
1058
71
.
60.
Wang
L
,
Šuštić
T
,
Leite de Oliveira
R
,
Lieftink
C
,
Halonen
P
,
van de Ven
M
, et al
A functional genetic screen identifies the phosphoinositide 3-kinase pathway as a determinant of resistance to fibroblast growth factor receptor inhibitors in FGFR mutant urothelial cell carcinoma
.
Eur Urol
2017
;
71
:
858
62
.
61.
Goyal
L
,
Saha
SK
,
Liu
LY
,
Siravegna
G
,
Leshchiner
I
,
Ahronian
LG
, et al
Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive cholangiocarcinoma
.
Cancer Discov
2017
;
7
:
252
63
.
62.
Heitzer
E
,
Haque
IS
,
Roberts
CES
,
Speicher
MR
. 
Current and future perspectives of liquid biopsies in genomics-driven oncology
.
Nat Rev Genet
2019
;
20
:
71
88
.
63.
Cowell
JK
,
Qin
H
,
Hu
T
,
Wu
Q
,
Bhole
A
,
Ren
M
. 
Mutation in the FGFR1 tyrosine kinase domain or inactivation of PTEN is associated with acquired resistance to FGFR inhibitors in FGFR1-driven leukemia/lymphomas
.
Int J Cancer
2017
;
141
:
1822
9
.
64.
Wu
D
,
Guo
M
,
Min
X
,
Dai
S
,
Li
M
,
Tan
S
, et al
LY2874455 potently inhibits FGFR gatekeeper mutants and overcomes mutation-based resistance
.
Chem Commun
2018
;
54
:
12089
92
.
65.
Chell
V
,
Balmanno
K
,
Little
AS
,
Wilson
M
,
Andrews
S
,
Blockley
L
, et al
Tumour cell responses to new fibroblast growth factor receptor tyrosine kinase inhibitors and identification of a gatekeeper mutation in FGFR3 as a mechanism of acquired resistance
.
Oncogene
2013
;
32
:
3059
70
.
66.
Goyal
L
,
Shi
L
,
Liu
LY
,
Fece de la Cruz
F
,
Lennerz
JK
,
Raghavan
S
, et al
TAS-120 overcomes resistance to ATP-competitive FGFR inhibitors in patients with FGFR2 fusion-positive intrahepatic cholangiocarcinoma
.
Cancer Discov
2019
;
9
:
1064
79
.
67.
Kalyukina
M
,
Yosaatmadja
Y
,
Middleditch
MJ
,
Patterson
AV
,
Smaill
JB
,
Squire
CJ
. 
TAS-120 cancer target binding: defining reactivity and revealing the first fibroblast growth factor receptor 1 (FGFR1) irreversible structure
.
ChemMedChem
2019
;
14
:
494
500
.
68.
Varghese
AM
,
Patel
JAA
,
Janjigian
YY
,
Meng
F
,
Selcuklu
SD
,
Zimel
C
, et al
Non-invasive detection of acquired resistance to FGFR inhibition in patients with cholangiocarcinoma harboring FGFR2 alterations
.
J Clin Oncol
27
:
15s
, 
2019
(
suppl; abstr 4096
).
69.
Moss
TJ
,
Rodon
J
,
Oakley
HD
,
Kahle
M
,
Karp
DD
,
Pant
S
, et al
Baseline cfDNA characteristics and evolution of cfDNA profile during treatment with selective FGFR inhibitor TAS-120
.
J Clin Oncol
37
:
15s
, 
2019
(
suppl; abstr 3056
).
70.
Hyman
DM
,
Goyal
L
,
Grivas
P
,
Meric-Bernstam
F
,
Tabernero
J
,
Hu
Y
, et al
FUZE clinical trial: a phase 2 study of Debio 1347 in FGFR fusion-positive advanced solid tumors irrespectively of tumor histology
.
J Clin Oncol
37
:
15s
, 
2019
(
suppl; abstr TPS3157
).
71.
Meric-Bernstam
F
,
Goyal
L
,
Tran
B
,
Bahleda
R
,
Kelley
RB
,
Hierro
C
, et al
TAS-120 in patients with advanced solid tumors bearing FGF/FGFR aberrations: a phase I study. [abstract]
.
In
:
Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29–Apr 3
;
Atlanta, GA. Philadelphia (PA)
:
AACR
;
Cancer Res
2019
.
Abstract nr CT238
.