scholarly journals ACT-1 Multicenter investigator-initiated registration-directed Phase 2 study of E7090 in subjects with advanced or recurrent solid tumors with fibroblast growth factor receptor (FGFR) gene alteration: FORTUNE trial

2021 ◽  
Vol 3 (Supplement_6) ◽  
pp. vi9-vi9
Author(s):  
Masamichi Takahashi ◽  
Yohei Chiba ◽  
Kazuki Sudo ◽  
Yuki Kojima ◽  
Hitomi Okuma ◽  
...  

Abstract Background: Genetic alterations of FGFRs are known to play an important role in the proliferation, survival, and migration of cancer cells as well as tumor angiogenesis and drug resistance. E7090 is an orally available selective tyrosine kinase inhibitor for FGFR1-3. A global Phase 2 study of E7090 in subjects with unresectable advanced or metastatic cholangiocarcinoma harboring FGFR2 gene fusion is ongoing (NCT04238715). We recently reported FGFR alterations that are highly sensitive to E7090 using a high-throughput functional evaluation method called MANO method (Nakamura et al. npj Precision Oncology, 2021), narrowing down the most promising FGFR alteration targets. Here, we designed a single-arm, open-label, investigator-initiated multicenter Phase 2 basket study to evaluate the efficacy and safety of E7090 in subjects with advanced or recurrent solid tumors harboring FGFR gene alterations, focusing on alterations identified by MANO method, as a sub-study under the nationwide large registry for rare cancers in Japan (MASTER KEY Project). Methods: The key eligibility criteria are: 1) Histologically confirmed metastatic or locally advanced solid tumor; 2) Ineffective to or intolerant to first line treatment, or for which standard treatment is no longer available; and 3) Confirmed FGFR gene alterations via next-generation sequencing assays that are reimbursed by insurance. Subjects will receive E7090 140 mg orally once daily until disease progression or development of unacceptable toxicity. The primary endpoint is objective response rate (ORR) by independent central review (RECIST v1.1), and the secondary endpoints include ORR by investigator assessment, progression-free survival, overall survival, disease control rate, safety, duration of response, and time to response. For primary brain tumors, RANO criteria is also applied in assessment of response. The study enrolls approximately 45 subjects. (Clinical Trial Registry: jRCT2031210043, ClinicalTrials.gov: NCT04962867)

2021 ◽  
Vol 3 (Supplement_4) ◽  
pp. iv8-iv8
Author(s):  
Masamichi Takahashi ◽  
Yohei Chiba ◽  
Kazuki Sudo ◽  
Yuki Kojima ◽  
Hitomi Okuma ◽  
...  

Abstract Background Genetic alterations of FGFRs are known to play an important role in the proliferation, survival, and migration of cancer cells as well as tumor angiogenesis and drug resistance. E7090 is an orally available selective tyrosine kinase inhibitor for FGFR1-3. A global Phase 2 study of E7090 in subjects with unresectable advanced or metastatic cholangiocarcinoma harboring FGFR2 gene fusion is ongoing (NCT04238715). We recently reported FGFR alterations that are highly sensitive to E7090 using a high-throughput functional evaluation method called MANO method (Nakamura et al. npj Precision Oncology, in press), narrowing down the most promising FGFR alteration targets. Here, we designed a single-arm, open-label, investigator-initiated multicenter Phase 2 basket study to evaluate the efficacy and safety of E7090 in subjects with advanced or recurrent solid tumors harboring FGFR gene alterations, focusing on alterations identified by MANO method, as a sub-study under the nationwide large registry for rare cancers in Japan (MASTER KEY Project). Methods The key eligibility criteria are: 1) Histologically confirmed metastatic or locally advanced solid tumor; 2) Ineffective to or intolerant to first line treatment, or for which standard treatment is no longer available; and 3) Confirmed FGFR gene alterations via next-generation sequencing assays that are reimbursed by insurance. Subjects will receive E7090 140 mg orally once daily until disease progression or development of unacceptable toxicity. The primary endpoint is objective response rate (ORR) by independent central review (RECIST v1.1), and the secondary endpoints include ORR by investigator assessment, progression-free survival, overall survival, disease control rate, safety, duration of response, and time to response. For primary brain tumors, RANO criteria is also applied in assessment of response. The study enrolls approximately 45 subjects. (Clinical Trial Registry: jRCT2031210043)


2021 ◽  
Vol 39 (3_suppl) ◽  
pp. TPS480-TPS480
Author(s):  
Shubham Pant ◽  
Josep Tabernero ◽  
Christophe Massard ◽  
Gopa Iyer ◽  
Olaf Witt ◽  
...  

TPS480 Background: The pan- FGFR tyrosine kinase inhibitor erdafitinib is approved by the US Food and Drug Administration for adults with locally advanced or metastatic urothelial carcinoma and susceptible FGFR3/2 genetic alterations who have progressed during or after ≥ 1 line of prior platinum-containing chemotherapy. FGFR gene alterations are potential oncogenic drivers that have been reported in many solid tumors in adult and pediatric pts. Because of limited response to standard of care options in pts failing systemic therapy, there is strong rationale to assess the safety and efficacy of erdafitinib in adolescent and adult pts with advanced solid tumors and FGFR alterations. Methods: This phase 2, open-label study (RAGNAR/42756493CAN2002; NCT04083976) will include pts aged ≥ 12 years with histologically confirmed unresectable, locally advanced, or metastatic solid tumors (except urothelial tumors) harboring predefined FGFR mutations or fusions. Eligibility screening includes molecular screening for FGFR alterations by central or local next-generation sequencing assays, and other clinical criteria. Pts will enroll into either a broad panel cohort (BPC) of target FGFR alterations or an exploratory cohort (EC) for FGFR alterations that do not meet criteria for BPC. Approximately 280 pts (BPC, n = 240; EC, n = 40) will be enrolled. The primary efficacy end point is overall response rate (ORR) as assessed by the independent review committee. Secondary end points include investigator-assessed ORR, duration of response, disease control rate, progression-free survival, overall survival, safety, pharmacokinetics, and health-related quality of life. Safety assessments include adverse events, vital signs, electrocardiograms, physical examinations, laboratory tests, performance status assessment, growth assessments in adolescents, and ophthalmologic examination. As of December 2019, pts are being enrolled at ~158 sites in 15 countries. Results of this study will provide efficacy and safety data for erdafitinib across multiple solid tumors with FGFR alterations and evaluate the potential benefit of targeting the underlying altered biology of FGFR irrespective of tumor histology in adult and adolescent pts. Clinical trial information: NCT04083976.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 5591-5591 ◽  
Author(s):  
Yasuhiro Funahashi ◽  
Richard T. Penson ◽  
Matthew A. Powell ◽  
David S. Miller ◽  
Jean Fan ◽  
...  

5591 Background: Lenvatinib is an oral receptor tyrosine kinase inhibitor targeting VEGFR1-3, FGFR1-4, RET, KIT, and PDGFRβ. A phase 2 study in patients with advanced endometrial cancer following 1 or 2 prior platinum-based treatments was performed and is presented in a separate abstract. Here we report potential predictive markers of clinical benefit. Methods: Pre- and post-treatment plasma samples collected from 122 of 133 treated subjects were analyzed for 50 circulating cytokine and angiogenic factors (CAFs) using ELISA and multiplex assay platforms; 107 archival tumor tissues obtained from 133 enrolled subjects were analyzed for gene mutation (mut) (81 samples) and gene expression profiling (GEP) (64 samples). For GEP analysis, the NanoString nCounter platform was used to measure the expression level of approximately 300 genes identified preclinically as relevant. Correlation with clinical outcome measures including maximal tumor shrinkage (MTS), objective response rate (ORR), PFS, and OS was performed. Results: Clinical correlation identified baseline levels of 7 CAFs related to angiogenesis (Ang-2, IL-8, HGF, VEGFA, PlGF, Tie-2, and TNFa) that associated with survival. Only baseline levels of Ang-2 associated with greater MTS (R=0.36 [Spearman], P<0.001), ORR (61% vs 18%), mPFS (9.5 vs 3.7 mos), and mOS (23 vs 8.9 mos) as determined using a defined cut-off value for baseline Ang-2. In gene mut analysis, no significant correlations were observed among the genes tested. Mut in PIK3CA showed a trend toward shorter OS (p = 0.085). In GEP analysis, expression levels of approximately 90 genes correlated with clinical outcome. Combination of GEP and CAF signatures identified signaling pathways, including Ang-2, that associated with OS. Clinical and preclinical GEP analysis identified overlapping gene signatures involving MAPK and PI3K signaling pathways that contributed to lenvatinib resistance. Conclusions: Baseline circulating Ang-2 levels may potentially predict outcomes in patients with advanced endometrial cancer and require further examination. This may provide a basis for stratification of patients in future clinical trials. Clinical trial information: NCT01111461.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 9526-9526 ◽  
Author(s):  
Alexander David Guminski ◽  
Annette May Ling Lim ◽  
Nikhil I. Khushalani ◽  
Chrysalyne D. Schmults ◽  
Leonel Fernando Hernandez-Aya ◽  
...  

9526 Background: Primary analysis (Oct 2017) of cemiplimab (REGN2810) in pts with mCSCC in a Phase 2 study demonstrated substantial antitumor activity, durable responses, and acceptable safety profile. We now report 12-month follow-up data from these pts (NCT02760498; data cutoff date: Sep 20, 2018). Methods: Pts with mCSCC received cemiplimab 3 mg/kg IV every 2 weeks (Q2W). Tumor measurements were performed Q8W. The primary objective was to evaluate objective response rate (ORR; complete response [CR] + partial response [PR]) according to independent central review (per RECIST 1.1 for scans; modified WHO criteria for photos). Results: 59 pts (median age: 71 years) were enrolled. Median duration of follow-up was 16.5 months (range: 1.1–26.6). ORR by central review was 49.2% (95% CI: 35.9–62.5; 10 CRs and 19 PRs [4 CRs and 25 PRs by investigator-assessment (INV)]). Median duration of response (DOR) has not been reached. The longest DOR at data cut-off was 21.6 months and was still ongoing. Observed DOR exceeded 12 months in 22/29 pts (75.9%) with response. Durable disease control rate (stable disease or response for ≥16 weeks) was 62.7% (95% CI: 49.1–75.0). Median observed time to response was 1.9 months (range: 1.7–9.1). Median progression-free survival was 18.4 months (95% CI: 7.3–not evaluable); median overall survival has not been reached. The most common treatment-emergent adverse events (all grades, Grade ≥3) were diarrhea (28.8%, 1.7%), fatigue (25.4%, 1.7%), and nausea (23.7%, 0%). By INV, grade ≥3 immune-related adverse events occurred in 13.6% of pts. Conclusions: This analysis demonstrates substantial antitumor activity and increasing DOR with cemiplimab 3 mg/kg Q2W in pts with mCSCC. There were no new safety signals. These data strongly support the recent FDA approval of cemiplimab-rwlc for pts with mCSCC or locally advanced CSCC who are not candidates for curative surgery or curative radiation. Clinical trial information: NCT02760498.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. TPS3157-TPS3157 ◽  
Author(s):  
David Michael Hyman ◽  
Lipika Goyal ◽  
Petros Grivas ◽  
Funda Meric-Bernstam ◽  
Josep Tabernero ◽  
...  

TPS3157 Background: Dysregulation of fibroblast growth factor receptor (FGFR) signaling by FGFR fusions is implicated in many cancers. Debio 1347 is a selective oral inhibitor of FGFR 1-3 tyrosine kinases. It exhibited high antitumor activity in in vitro and in vivo tumor models with FGFR1-3 gene fusions. Preliminary data from an ongoing phase 1 trial show efficacy and tolerability in patients (pts) harboring FGFR 1-3 fusion irrespectively of tumor type. We present the design for a multicenter, basket, 2-stage, adaptive single arm Phase 2 trial investigating Debio 1347 in pts with solid tumors harboring FGFR1-3 fusion/rearrangement. Methods: Adults with locally advanced/unresectable or metastatic tumors with documented FGFR1-3 gene fusion/rearrangement who require systemic therapy and have progression after ≥1 prior standard treatment or have no satisfactory alternative treatment option are eligible. Three cohorts are included: biliary tract cancer (cohort 1), urothelial cancer (cohort 2) and all other solid tumors (cohort 3). Primary brain tumors are excluded. Other key exclusion criteria include prior treatment with FGFR1-3 selective inhibitor; clinically significant corneal/retinal disorder; history of calcium/phosphate homeostasis disorder or systemic mineral imbalance with ectopic soft tissue calcification, and symptomatic/unstable brain metastases < 1 month before enrollment. Genomic screening of tumor tissue is done at local or central laboratory with post-hoc central confirmation by RNA sequencing. Eligible pts will receive Debio 1347, 80 mg PO once daily in 28-day cycles until occurrence of progression or unacceptable toxicity. Primary Endpoint is objective response rate (ORR) based on independent central review using RECIST v.1.1. The targeted sample size (N=125) will provide approximately 90% power to reject H0: ORR ≤ 15% at an overall 5% significance level based on an expected ORR of 30% in at least one of the cohorts. Secondary endpoints are: duration of response, disease control rate, progression-free survival, overall survival, safety, tolerability, and quality of life. An interim analysis for futility and homogeneity will be performed after 27 evaluable pts. PK sparse sampling is performed to assess exposure-response relationships with efficacy and safety. Biomarkers of response and resistance will be explored. Accrual is opened in US, EU, Asia and Australia. Clinical trial information: NCT03834220.


2020 ◽  
Vol 38 (4_suppl) ◽  
pp. TPS470-TPS470 ◽  
Author(s):  
Antoine Hollebecque ◽  
Toshihiko Doi ◽  
Omar Saavedra ◽  
Osamu Takahashi ◽  
Helen He ◽  
...  

TPS470 Background: FGFR genomic aberrations are known to drive oncogenesis in multiple tumor types via FGFR signaling pathway dysregulation. Futibatinib is an oral, highly selective, irreversible FGFR1–4 inhibitor that has shown potent antiproliferative activity against FGFR-deregulated tumors of diverse tissue origins in preclinical studies. In a phase 1 dose-escalation/expansion study, futibatinib showed promising antitumor activity and tolerability in previously treated pts with tumors harboring FGFR aberrations. This phase 2 study was designed to evaluate the efficacy and safety of futibatinib in pts with tumors harboring FGFR aberrations. The study will enroll pts in multiple cohorts based on diagnosis and FGFR aberration status; cohorts enrolling pts with adv solid tumors are reported here. Methods: In this global, open-label, phase 2 study, pts (≥18 years; Eastern Cooperative Oncology Group performance status of 0 or 1) will be enrolled in cohort A (~60 pts with metastatic/locally adv solid tumors, except primary brain tumors or intrahepatic cholangiocarcinoma, harboring FGFR1–4 rearrangements and with disease progression after standard treatment) or cohort B (~35 pts with metastatic/locally adv gastric tumors harboring FGFR2 amplifications and with ≥2 prior therapies). Key exclusion criteria are clinically significant alterations in calcium–phosphorus homeostasis, ectopic mineralization/calcification, and prior FGFR inhibitor treatment. Pts will receive 20 mg futibatinib once daily in a continuous 28-day cycle until disease progression, unacceptable toxicity, or other discontinuation criteria are met. The primary endpoint is objective response rate (ORR) per independent central review. Secondary endpoints include ORR per investigator, disease control rate, duration of response, progression-free survival, overall survival, and safety. The anticipated start date is in April 2020.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS3620-TPS3620
Author(s):  
Kanwal Pratap Singh Raghav ◽  
Takayuki Yoshino ◽  
Rosine Guimbaud ◽  
Ian Chau ◽  
Marc Van Den Eynde ◽  
...  

TPS3620 Background: Trastuzumab deruxtecan (T-DXd) is an antibody–drug conjugate consisting of an anti-HER2 antibody (trastuzumab) linked to a potent topoisomerase I inhibitor (DXd). T-DXd has been approved to treat HER2-positive metastatic breast cancer (United States, Japan, Europe) and advanced gastric cancer (United States, Japan). It is currently being evaluated in other solid tumor types including colorectal cancer. The phase 2 DESTINY-CRC01 study included patients with RAS wild-type mCRC, with median 4 (range, 2-11) prior lines of therapy. Preliminary results in patients with HER2-overexpressing (IHC 3+ or IHC 2+/ISH+) mCRC showed T-DXd treatment (6.4 mg/kg intravenously [IV] every 3 weeks [Q3W]) resulted in a confirmed objective response rate (ORR) of 45.3% (24/53; 95% CI, 31.6%-59.6%) and a median progression-free survival (PFS) of 6.9 months (95% CI, 4.1 months-not evaluable; Siena J Clin Oncol. 2020;38[15]:4000). Activity was also seen in patients treated with prior anti-HER2 therapy. Although 5.4-mg/kg and 6.4-mg/kg doses of T-DXd have shown clinical efficacy in multiple cancer indications, the lower dose has not yet been tested in patients with HER2-overexpressing mCRC. Preliminary data also suggest T-DXd may be active in RAS mutant mCRC, unlike other anti-HER2 therapies. The DESTINY-CRC02 study aims to determine efficacy and safety of T-DXd in patients with HER2-overexpressing, RAS wild-type or mutant mCRC at 5.4-mg/kg and 6.4-mg/kg doses. Methods: DESTINY-CRC02 (NCT04744831) is a multicenter, randomized, double-blind, 2-arm, parallel phase 2 study that will be conducted in 2 stages. Eligible patients (≥18 years; ≥20 years in Japan, Taiwan, and Korea) will have HER2-overexpressing (IHC 3+ or IHC 2+/ISH+) locally advanced, unresectable or metastatic CRC and have previously received chemotherapy, anti-EGFR therapy, anti-VEGF treatment, and/or anti–PD-1/PD-L1 therapy, as clinically indicated. Prior anti-HER2 therapy will be allowed. In stage 1, patients will be randomly assigned 1:1 to receive T-DXd IV Q3W at a dose of 5.4 mg/kg (n = 40; arm 1) or 6.4 mg/kg (n = 40; arm 2). Randomization will be stratified by ECOG PS (0 or 1), HER2 status (IHC 3+ or IHC 2+/ISH+), and RAS status (wild-type or mutant). After stage 1 enrollment is complete, eligible patients in stage 2 (n = 40) will receive T-DXd 5.4 mg/kg until disease progression or other treatment discontinuation criteria are met. The study is actively enrolling and aims to enroll 120 patients across 60 sites. The primary objective is to assess efficacy of T-DXd at the 5.4-mg/kg and 6.4-mg/kg doses, with a primary end point of confirmed ORR by blinded independent central review. Secondary end points include investigator-assessed ORR, PFS, duration of response, disease control rate, clinical benefit rate, overall survival, pharmacokinetics, and safety. Clinical trial information: NCT04744831.


Sign in / Sign up

Export Citation Format

Share Document