Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Predictive Biomarkers and Personalized Medicine

Targeting FGFR with Dovitinib (TKI258): Preclinical and Clinical Data in Breast Cancer

Fabrice André, Thomas Bachelot, Mario Campone, Florence Dalenc, Jose M. Perez-Garcia, Sara A. Hurvitz, Nicholas Turner, Hope Rugo, John W. Smith, Stephanie Deudon, Michael Shi, Yong Zhang, Andrea Kay, Diana Graus Porta, Alejandro Yovine and José Baselga
Fabrice André
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thomas Bachelot
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mario Campone
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Florence Dalenc
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jose M. Perez-Garcia
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sara A. Hurvitz
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nicholas Turner
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hope Rugo
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
John W. Smith
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stephanie Deudon
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael Shi
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yong Zhang
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Andrea Kay
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Diana Graus Porta
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alejandro Yovine
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
José Baselga
1Department of Medical Oncology, INSERM Unit U981, Paris Sud University, Institut Gustave-Roussy, Villejuif; 2Centre Léon-Bérard, Lyon; 3Centre René Gauducheau, Saint-Herblain; 4Institut Claudius Regaud, Toulouse, France; 5Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; 6David Geffen School of Medicine, University of California, Los Angeles; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; 8US Oncology, NW Cancer Specialists, Portland, Oregon; 9Novartis Pharmaceuticals Corporation, East Hanover, New Jersey; 10Royal Marsden Hospital, London, United Kingdom; 11Novartis Pharma AG; and 12Novartis Institutes for BioMedical Research, Basel, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-13-0190 Published July 2013
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Purpose: Fibroblast growth factor receptor 1 (FGFR1) and FGFR2 amplifications are observed in approximately 10% of breast cancers and are related to poor outcomes. We evaluated whether dovitinib (TKI258), an inhibitor of FGFR1, FGFR2, and FGFR3, presented antitumor activity in FGFR-amplified breast cancers.

Experimental Design: Preclinical activity of dovitinib was evaluated in both breast cancer cell lines and an FGFR1-amplified xenograft model (HBCx2). Dovitinib was then evaluated in a phase II trial that included 4 groups of patients with human EGF receptor 2–negative metastatic breast cancer on the basis of FGFR1 amplification and hormone receptor (HR) status. FGFR1 amplification was assessed by silver in situ hybridization. Preplanned retrospective analyses assessed predictive value of FGFR1, FGFR2, and FGF3 amplifications by quantitative PCR (qPCR).

Results: Dovitinib monotherapy inhibits proliferation in FGFR1- and FGFR2-amplified, but not FGFR-normal, breast cancer cell lines. Dovitinib also inhibits tumor growth in FGFR1-amplified breast cancer xenografts. Eighty-one patients were enrolled in the trial. Unconfirmed response or stable disease for more than 6 months was observed in 5 (25%) and 1 (3%) patient(s) with FGFR1-amplified/HR-positive and FGFR1-nonamplified/HR-positive breast cancer. When qPCR-identified amplifications in FGFR1, FGFR2, or FGF3 were grouped to define an FGF pathway–amplified breast cancer in HR-positive patients, the mean reduction in target lesions was 21.1% compared with a 12.0% increase in patients who did not present with FGF pathway–amplified breast cancer.

Conclusion: Dovitinib showed antitumor activity in FGFR-amplified breast cancer cell lines and may have activity in breast cancers with FGF pathway amplification. Clin Cancer Res; 19(13); 3693–702. ©2013 AACR.

Translational Relevance

FGFR1 amplification in breast cancer is associated with resistance to endocrine therapy and poor prognosis. Dovitinib has shown antitumor activity in advanced breast cancer with FGF pathway alterations, suggesting that FGFR could be a therapeutic target in these patients and warrants further investigation.

Introduction

Breast cancer is segmented into molecular subgroups defined by genomic alterations involved in tumor progression, which could identify patient populations best treated with targeted agents. One such population may be patients with gene alterations in the fibroblast growth factor (FGF) pathway, which consists of 18 different FGF ligands that act on 4 transmembrane tyrosine kinase FGF receptors (FGFR 1–4; ref. 1).

Multiple genetic alterations in FGFRs have been identified in breast cancer. For example, amplification of FGFR1 (8p11-12) is present in 8% to 15% of all breast cancer (2–4) and 16% to 27% of luminal type B breast cancer (5). These amplifications correlate with FGFR1 overexpression and are associated with resistance to endocrine therapy and poor prognosis (3, 5). Preclinical studies suggest that targeting FGFR1 could lead to antitumor effects by decreasing cell viability and restoring endocrine therapy sensitivity. Several other genomic alterations of the FGF pathway have also been observed in breast cancer (6, 7), including FGFR2 and FGF3/4/19 amplification.

Dovitinib (TKI258) is an oral tyrosine kinase inhibitor (TKI) with in vitro IC50 values against FGFR1–3, VEGFR1–3, and platelet-derived growth factor receptor (PDGFR) of approximately 10 nmol/L (8, 9). Phase I studies have suggested that dovitinib has a tolerable safety profile and effectively targets FGFR at therapeutic doses. In order to determine whether FGFR inhibition could lead to antitumor effects in patients with breast tumors harboring FGFR amplifications, we first evaluated the preclinical activity of dovitinib in breast cancer preclinical models, and then conducted a phase II trial of dovitinib in patients with metastatic breast cancer, with and without FGFR1 amplification. Importantly, this is the first report of a study selecting patients on the basis of FGFR1-amplification status.

Materials and Methods

Preclinical studies

Effects of dovitinib on cell proliferation using methylene blue staining were assessed after a 72-hour exposure in 13 breast cancer cell lines, including 2 with FGFR1 (MDA-MB-134) or FGFR2 (SUM52) amplification. Effects of dovitinib on FGFR signaling were assessed by detection of phosphorylated FGFR substrate 2 (pFRS2) and phosphorylated extracellular signal–regulated kinase/mitogen-activated protein kinase (pERK/MAPK) via Western blot using rabbit polyclonal anti-pFRS2 (Tyr196), anti-pMAPK (Thr202/Tyr204), or mouse monoclonal anti–β-tubulin (Cell Signaling Technology). Athymic nude mice were implanted with a human FGFR1- or FGFR2-amplified breast cancer xenograft, HBCx-2 or HBCx-3, respectively. Ten mice per group were treated for up to 42 days (HBCx-2) or 35 days (HBCx-3) with vehicle or dovitinib [30 or 50 mg/kg daily (HBCx-2) or 40 mg/kg daily (HBCx-3) by oral gavage] and tumors were measured twice weekly. The percentage tumor volume change of the treatment over control group (T/C) was calculated by dividing the change in mean tumor volume of the drug-treated group by the change in the mean tumor volume of the control-treated group and multiplying by 100.

Study design and treatment

This nonrandomized phase II trial evaluated dovitinib in female patients with HER2-negative metastatic breast cancer. Patients were divided into 4 cohorts on the basis of hormone receptor (HR; ER or PR) status (positive [+] and negative [−]) and FGFR1 amplification status. Patients having an average of 6 or more copies of FGFR1 assessed by fluorescent–, chromogenic–, or silver in situ hybridization (FISH, CISH, or SISH) were considered as FGFR1 amplified (FGFR1+). SISH was carried out centrally in formalin-fixed and paraffin-embedded (FFPE) tumor tissue sections for all patients using the ultraView SISH DNP Detection Kit (Ventana Medical Systems, Inc.), either retrospectively for confirmation of local assessments, if available, or prospectively for those centers where local assessment was not done. The study results are presented using central SISH. The study followed a Simon 2-stage design (10) for each cohort to test the null hypothesis that the response rate (confirmed complete response + partial response) was 5% or less versus the alternative hypothesis that the response rate was greater than 5% using a 1-sided test with 10% level of significance and 78% power at the alternative response rate of 15%. Twenty patients were planned to be enrolled in each cohort for each stage. All patients provided informed consent. The study was conducted in accordance with the ethical principles of the Declaration of Helsinki, with the protocol and all amendments reviewed by the independent ethics committee or institutional review board for each center. The trial was registered on www.clinicaltrials.gov (NCT00958971).

Dovitinib was administered orally at 500 mg/day (5 days on/2 days off) in 28-day cycles (11). The primary objective was to determine the overall response rate in patients with measurable disease at baseline according to Response Evaluation Criteria In Solid Tumors (RECIST) v1.0 (12). Complete response and partial response (PR) had to be confirmed in a second assessment after 4 to 6 weeks. Similarly, stable disease (SD) had to last for at least 6 weeks. Secondary objectives and exploratory analyses included progression-free survival (PFS), safety, and analysis of predictive value for FGFR1, FGF3, and FGFR2 as determined by quantitative PCR (qPCR). Tumor measurability at baseline, responses, and PFS were assessed locally and centrally. A second independent central review (adjudication) was conducted for discordant cases between the local and first central reviews. The adjudicating radiologist was blinded to treatment arm and previous assessment. Adjudicated results are reported here.

Eligibility criteria

Women 18 years or older with histologically confirmed HER2-negative metastatic breast cancer were eligible for the trial. The primary tumor, metastatic axillary lymph nodes, or biopsy of metastatic tumor must have been tested by FISH, CISH, or SISH for FGFR1 amplification (defined as ≥6 copies) by a designated local investigator or a reference laboratory before study entry with archival tumor tissue available for central confirmation. Patients with HR+ disease had to have received at least 1 line of endocrine therapy, and less than 3 lines of chemotherapy in the metastatic setting, whereas patients with HR− disease had to have received between 1 and 3 lines of chemotherapy in the metastatic setting. Additional inclusion criteria included measurable disease, World Health Organization (WHO) performance status of 0 or 1, left ventricular ejection fraction (LVEF) 45% or higher, and adequate bone marrow, hepatic, and renal function. Patients with liver metastases were eligible if they had grade 2 or lower alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Patients on chronic treatment with corticosteroids or other immunosuppressive agent or those with brain metastases or significantly decreased cardiac function were excluded from study participation.

Biomarker analyses

Blood samples were collected predose on days 1, 5, and 26 of cycle 1, day 26 of cycle 2, and day 1 of odd-numbered cycles beginning at cycle 5. A 6-hour postdose sample was also collected on cycle 1, day 5. Plasma FGF23 was measured as a surrogate marker of FGFR1 inhibition using a commercial ELISA (Kinos Laboratories, Inc.). Tumor copy number variations of FGFR1, FGFR2, and FGF3 were quantified by qPCR to test the predefined hypothesis that FGFR1, FGFR2, or FGF3 amplifications would identify patients who are highly sensitive to dovitinib. Briefly, DNA was extracted from FFPE sections using the QIAamp DNA FFPE tissue kit (Qiagen). TaqMan Copy Number Assays (Life Technologies Corporation) for FGFR1, FGFR2, and FGF3 were normalized with the copy number obtained from a simultaneous analysis using the TaqMan Copy Number Reference Assay [ribonuclease P (RNAase P)] in a duplex real-time PCR. The cutoff for FGFR1 and FGF3 gene amplification was 6 copies or more, consistent with the predefined FGFR1 gene amplification threshold (by hybridization) in the trial protocol. A threshold of 4 FGFR2 copies or more by qPCR was predefined on the basis of a previous report of Turner and colleagues (7).

Results

Dovitinib had antitumor activity in FGFR-amplified breast cancer models

Dovitinib decreased the concentrations of pFRS2 and pERK/MAPK in a dose -dependent manner in MDA-MB-134 (FGFR1 amplified) and SUM52 (FGFR2 amplified) cell lines (Fig. 1A). Because these data indicated that dovitinib effectively inhibited FGFR signaling in vitro, we further explored whether dovitinib preferentially inhibited proliferation of FGFR1- and FGFR2-amplified breast cancer cell lines. The IC50 for cell growth inhibition was 190 and 180 nmol/L in MDA-MB-134 and SUM52, respectively (Fig. 1B and Supplementary Table S1). Conversely, IC50 values were more than 2,000 nmol/L in the 11 breast cancer cell lines that had neither FGFR1 nor FGFR2 amplification. We then tested the antitumor activity of dovitinib in a FGFR1-amplified in vivo model (HBCx-2 breast cancer primary xenograft, with 8 FGFR1 gene copies; ref. 13). As shown in Fig. 1C, dovitinib prevented tumor growth at the 30 mg/kg dose and caused tumor regression at the 50 mg/kg dose. On day 28, T/C calculations showed that the mean tumor volume was 24.6% and 7.2% of the vehicle control for the 30 and 50 mg/kg groups, respectively (P < 0.001 by Mann–Whitney nonparametric test). In addition, we evaluated dovitinib in an FGFR2-amplified in vivo model (HBCx-3; 10 FGFR2 copies). Similar to the results described earlier, dovitinib caused tumor regression in HBCx-3 xenografts when administered at a dose of 40 mg/kg daily until day 35 (Fig. 1D). On day 28, T/C calculations showed a mean tumor volume for the dovitinib-treated group that was 23.4% of the vehicle control group (P < 0.001). Overall, these data suggested that dovitinib had antitumor activity in FGFR-amplified breast cancer models.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Dovitinib inhibits FGFR downstream signaling and cell proliferation in FGFR1- or FGFR2-amplified breast cancer cell lines and growth of FGFR1-amplified HBCx-2 xenografts. A, effects of dovitinib on FGFR signaling were assessed by Western blot detection of pFRS2 and pERK/MAPK in FGFR1-amplified MDA-MB-124 and FGFR2-amplified SUM52 cells. B, dovitinib inhibited cell proliferation in FGFR-amplified, but not FGFR-nonamplified, breast cancer cell lines. C and D, dovitinib showed tumor growth inhibition in (C) FGFR1-amplified HBCx-2 and (D) FGFR2-amplified HBCx-3 mouse xenograft models. Error bars indicate standard error of the mean. The T/C values and P values for the Mann–Whitney nonparametric tests compared with vehicle control are shown below each graph.

Patient characteristics

On the basis of the data observed in cell lines, together with consistent results from other groups (5, 14), we conducted a phase II trial to test the hypothesis that dovitinib has antitumor activity in patients with FGFR1-amplified breast cancer. In addition to enrolling patients with FGFR1-amplified tumors, patients with nonamplified tumors were enrolled to determine if dovitinib's ability to inhibit other targets (i.e., VEGFR and PDGFR) would mediate FGFR-independent antitumor activity. A retrospective analysis was also preplanned to evaluate the activity of dovitinib in patients with FGFR2- and FGF3-amplified breast cancers. Of the 243 HER2− patients who underwent molecular prescreening (i.e., tumor sample analyzed for FGFR1 assessment), 116 did not continue to the protocol-specific screening phase due to nonassessable FGFR1 status (insufficient tumor or insufficient SISH signal, n = 24) or the assigned treatment arm being closed to additional enrollment (n = 92; Fig. 2). For example, enrollment to the FGFR1− arms was completed early and closed to accrual, with screening continued in order to complete accrual of the FGFR1+ arms. Of the 127 patients who entered the protocol-specific screening phase, 46 were excluded from study entry due to not meeting inclusion criteria, most commonly due to hematologic and biochemical laboratory values outside the requested range. The remaining 81 patients were enrolled and received treatment, with a median time of 18.3 months from first treatment to analysis cutoff. These 81 patients were divided into 4 cohorts on the basis of FGFR1 amplification and HR status: FGFR1+/HR+ (n = 23), FGFR1+/HR− (n = 2), FGFR1−/HR+ (n = 34), and FGFR1−/HR− (n = 22; Table 1). Notably, there were very few cases of FGFR1-amplified HR− disease (n = 2), confirming there is a very low incidence of FGFR1 amplification in patients with triple-negative breast cancer, and this arm was stopped before completion of enrollment.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Study design. Patients were prescreened to determine FGFR1 status, then stratified into 4 cohorts on the basis of HR status and FGFR1 amplification. The study was designed as a Simon 2-stage with 20 patients planned for enrollment in each cohort for each stage. No arm proceeded to stage 2.

View this table:
  • View inline
  • View popup
Table 1.

Patient characteristics

Patient characteristics for the fully enrolled cohorts (FGFR1+/HR+, FGFR1−/HR+, and FGFR1−/HR−) are reported in Table 1. Most patients had late-stage breast cancer and were heavily pretreated. For example, 70% and 78% of the FGFR1+/HR+ patients presented with 3 organs or more involved and liver metastases, respectively. In addition, 95% of all patients were previously treated with chemotherapy in the metastatic setting and all but 10 HR+ patients previously received therapeutic endocrine therapy. Two patients who did not receive any endocrine therapy (adjuvant, neoadjuvant, or therapeutic) were already refractory to previous chemotherapy at study entry and were not considered as candidates for endocrine therapy as per the investigator's judgment.

Dovitinib effectively increases FGF23 plasma levels, indicative of FGFR1 inhibition

FGF23 has been identified as a target gene of FGF signaling in vitro (15). Furthermore, increases in FGF23 have previously been reported as a surrogate for FGFR1 inhibition in clinical trials of dovitinib in renal cell carcinoma and melanoma (11, 16, 17). In these trials, 50% to 100% increases in plasma FGF23 from baseline were detected at cycle 1, day 15 and were correlated with reduced pERK (17). In the data presented here, plasma FGF23 levels peaked ≈100% above baseline at cycle 1, day 8 and were sustained over time, suggesting that dovitinib effectively inhibited FGFR1 signaling (Supplementary Fig. S1). The level of increase was similar among the 3 groups of patients (FGFR1+/HR+, FGFR1−/HR+, and FGFR1−/HR−), suggesting that there were no major differences in pharmacodynamic activity between the 3 groups.

Dovitinib exhibits greater antitumor effects in FGFR1-amplified as compared with nonamplified breast cancers

No complete responses or confirmed PRs were observed (overall response rate, 0%) as per the adjudicated central review (Table 2). Three patients with FGFR1+/HR+ breast cancer achieved an initial objective response, although these responses were not confirmed at a subsequent assessment. One patient showed a complete disappearance of liver lesions with persistence of nontarget bone and lymph node lesions in the first postbaseline evaluation. This would have qualified as a PR, but was not confirmed in the second evaluation (day 109) because the investigator assessed disease progression in the bone and discontinued therapy, while the liver disease remained in complete response. The second patient showed a reduction of 30.8% in target lesions in the liver, retroperitoneum, and ovaries in the second postbaseline evaluation, but was not confirmed in the 3 subsequent evaluations that showed 25.8%, 27.1%, and 28.5% reductions from baseline. The patient experienced disease progression (observed on day 274; new peritoneal lesion). The third patient had a reduction of 40.3% in target lesions. However, this was not confirmed due to clinical disease progression at day 87 (increased markers cancer antigen 15.3 and carcinoembryonic antigen, and decline in performance status). The patient refused further assessment.

View this table:
  • View inline
  • View popup
Table 2.

Best overall response summary on the basis of adjudicated data according to RECIST criteria

Two additional FGFR1+/HR+ evaluable patients had SD for 24 weeks or longer. Therefore, 5 of 20 FGFR1+/HR+ patients (25%) achieved either an unconfirmed PR or SD 24 weeks or longer. On the contrary, only 1 patient (3%) in the FGFR1−/HR+ cohort and 2 patients (13%) in the FGFR1−/HR− cohort achieved long-term (≥24 weeks) SD and none had tumor shrinkage greater than 30%. None of the study arms proceeded to the second stage because the predefined criteria (at least 2 confirmed objective responses) were not met.

We further explored whether higher levels of FGFR1 amplification detected by qPCR are predictive for dovitinib sensitivity in patients with HR+ disease. Tissue for qPCR assessment was available for 42 of 51 HR+ patients with measurable disease. Thirty-eight of these patients had assessable disease, and in 35 of them the reported change in tumor size was in accordance with the overall tumor response; these are included in this analysis. Thirteen and 22 of these patients were FGFR1 amplified and nonamplified by SISH, respectively. Using qPCR, with a cutoff of 6 copies for FGFR1, a total of 7 patients were identified with FGFR1 amplification. Interestingly, qPCR data suggested that 6 cases presented FGFR1 amplification by SISH but not by qPCR. There was full concordance for the remaining 22 patients with no amplification of FGFR1 by SISH (all of them nonamplified by qPCR as well). In the 7 patients amplified by qPCR (and SISH), a 20.2% reduction in the mean tumor size was observed (range, 100% reduction to 28.4% increase, with 6 of the 7 patients showing tumor shrinkage and only 1 showing tumor increase) compared with a 14.2% increase in mean tumor size for the 6 patients with FGFR1 amplification detected by SISH but not qPCR (range, 11.6% reduction to 54.0% increase). As shown in Fig. 3A, dovitinib was more effective in FGFR1-highly amplified (≥6 gene copies) breast cancer as compared with tumors with lower levels of FGFR1 amplification in this population of HR+ patients. In the FGFR1-amplified breast cancer group (n = 7), dovitinib induced a mean 20.2% reduction in tumor size (range, 28.4% increase to 100% reduction). Conversely, tumors with less than 6 FGFR1 copies (n = 28) had a mean 8.3% increase (range, 54.2% increase to 28.2% reduction). Overall, these data suggested that dovitinib shows more potent antitumor activity in patients with high levels of FGFR1 amplification compared with those tumors without FGFR1 amplification by qPCR.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Dovitinib induces target lesion reductions in FGF-pathway–amplified tumors. A, box plot showing the best tumor change of the target lesions on the basis of FGFR1 copy number as assessed by qPCR in HR+ patients with measureable disease at baseline and evaluable for this plot (n = 35). The line and + symbol within each box represents the median and mean value for that group. Whiskers above and below each box indicate the maximum and minimum values in that group, respectively. B, waterfall plot of HR+ FGF-pathway–amplified (black bars; FGFR1, FGFR2, or FGF3 amplification by qPCR) or FGF-pathway–nonamplified (gray bars) patients evaluable for this plot (n = 38). The asterisk (*) denotes patients whose overall lesion response is progressive disease, but the target lesion response is PR or SD. The dagger (†) denotes the FGFR1−/FGF3+ patient with 3.4 copies of FGFR1 by qPCR, the double dagger (‡) denotes the FGFR1−/FGFR2+ patients, and the section mark (§) denotes the FGFR1+/FGFR3+ patients.

FGF3 and FGFR2 amplification to complement FGFR1 amplification in selecting individuals most likely to respond to dovitinib

Because FGF3 and FGFR2 amplifications have also been reported in breast cancers (6, 18, 19), we further evaluated by qPCR whether such amplifications could define additional subsets of sensitive patients. Four patients had FGF3 amplification as measured by qPCR, with tumor reductions of 100%, 30.8%, 23.0%, and 7.5%, respectively. Interestingly, 3 of these 4 patients also had a high level of FGFR1 amplification (≥6 copies by qPCR) and the fourth presented with FGFR1-gene gain (3.4 copies of FGFR1 by qPCR and SISH negative).

FGFR2 amplification was also assessed by qPCR and detected (≥4 copies) in 2 of the HR+ patients (both FGFR1 nonamplified by SISH and qPCR). These 2 patients with FGFR2+/FGFR1−/HR+ breast cancer had tumor reductions of 28.2% and 18.5%. Two additional patients with HR− disease had FGFR2 amplification by qPCR, but discontinued due to adverse events (AEs) before a postbaseline assessment.

We further analyzed the impact of amplification in FGFR1 (≥6 copies), FGFR2 (≥4 copies), or FGF3 (≥6 copies) by qPCR in the above-described group of patients with assessable and HR+ disease (n = 38). Ten of these 38 HR+ patients were defined as having FGF-pathway–amplified breast cancer (FGFR1 and/or FGFR2 and/or FGF3 amplification). Interestingly, the mean reduction in target lesions was 21.1% from baseline (range, 28.4% increase to 100% reduction) in these 10 patients. Conversely, target lesions in the remaining 28 patients who did not present with FGF-pathway–amplified breast cancer had a mean 12.0% increase from baseline (range, 54.2% increase to 15.4% reduction). Best tumor response according to the presence of gene amplification on FGF-pathway is reported in Fig. 3B. Interestingly, this analysis showed that FGF pathway amplification–negative patients were unlikely to respond to dovitinib, as 16 out of 28 patients (57.1%) without FGF-pathway amplification presented either a new lesion or tumor size increase as best response, compared with only 1 out of 10 (10.0%) FGF-pathway–amplified patients. Similar results were obtained when amplification of FGFR1 or FGFR2 was used to define FGF-pathway amplification as only 1 patient presented with FGF3 amplification but not FGFR1 or FGFR2 amplification.

Safety

All patients eventually discontinued the study, with disease progression as the most common reason reported for discontinuation (n = 47, 58.0%). AEs regardless of relationship to dovitinib were reported as the primary reason for discontinuation in 22 patients (27.2%). AEs leading to discontinuation were most commonly grade 3, the most common being asthenia/fatigue (n = 7), gastrointestinal disorders (n = 5), and investigations of laboratory abnormalities (n = 6, mainly liver function test abnormalities).

All patients (100.0%) experienced at least 1 AE regardless of relationship to study drug, most commonly vomiting (77.8%), diarrhea (76.5%), asthenia (67.9%), and nausea (67.9%; Table 3). Sixty-three patients (77.8%) experienced a grade 3/4 AE. The most common grade 3/4 AEs regardless of study-drug relationship were asthenia (23.5%), ALT increase (11.1%), diarrhea (8.6%), and vomiting, fatigue, and AST increase (7.4% each). No notable differences across the groups were observed (data not shown).

View this table:
  • View inline
  • View popup
Table 3.

Adverse events (≥15% any grade) regardless of study drug relationship (N = 81)

Patients were also monitored for clinical laboratory abnormalities. Overall, new or worsened grade 3 and 4 alkaline phosphatase was observed in 21.1% and 1.3% of patients, respectively, and was more common in the FGFR1+/HR+ group (43.5%). New or worsened grade 3 or 4 AST increase was observed in 16.7% of the patients (all grade 3), and new or worsened grade 3 and 4 ALT increase was also observed in 15.4% and 1.3% of patients, respectively. New or worsening grade 3 and 4 total bilirubin was observed in only 2.6% and 1.3% of patients, respectively.

The most frequently observed grade 3 or 4 hematologic clinical laboratory abnormality was lymphopenia (16.9%, all grade 3). The majority of new or worsened abnormalities in absolute neutrophils, hemoglobin, white blood cell, and platelet counts were grades 1 and 2, with few patients exhibiting a shift to grade 4. No major difference in hematologic clinical laboratory abnormalities was observed between the treatment groups.

Eight patients died on the study or during the 28-day follow-up period. In 3 cases, the fatal event was assessed by the investigator as being related to the study drug, and in the remaining 5 cases, the death was deemed as secondary to progression of underlying disease. A brief description of the 3 deaths not related to progressive disease follows. Patient #1 was a 62-year-old female with history of hypertension, diabetes mellitus, and heavily pretreated (anastrozole, tamoxifen, fulvestrant, paclitaxel, bevacizumab, and capecitabine) metastatic breast cancer (widespread metastatic liver and bone disease). After 4 weeks of therapy, the patient experienced grade 3 AST and grade 4 ALT increases, grade 4 alkaline phosphatase increase, and grade 4 total bilirubin increase. Dovitinib treatment was discontinued due to these events, and the patient died 25 days after the last dose of therapy due to acute liver toxicity (cholestatic liver damage). Patient #2, a 44-year-old female who was heavily pretreated (doxorubicin, cyclophosphamide, docetaxel, cisplatin, vinorelbine, tamoxifen, and anastrozole), had breast cancer with multiple mediastinal, pulmonary, and bone metastases at baseline. She received 8 weeks of therapy until discontinuation due to disease progression (new pleural mass and subpleural nodes). The patient started paclitaxel 12 days after study discontinuation and died of pleural hemorrhage and acute respiratory failure 15 days later (27 days after the last dose of dovitinib). Patient #3, a 62-year-old female with metastatic breast cancer (single measurable liver lesion at baseline), had previously been treated with doxorubicin, cyclophosphamide, docetaxel, bevacizumab, and capecitabine. The patient received 25 doses of dovitinib and died at day 51, 4 days after the last dose. The death was secondary to listeria infection and sepsis following treatment-emergent grade 3 neutropenia.

Discussion

We examined the activity of dovitinib in preclinical models and patients with breast cancer. Our findings suggest that dovitinib could have modest antitumor activity in FGF-pathway–amplified tumors, but not in FGF-pathway–nonamplified tumors. These data suggest that future trials testing FGFR inhibitors should focus on FGF-pathway–amplified breast cancer.

This study was conducted in a population who was unlikely to respond. These patients had advanced disease (70% had ≥3 organs involved and 78% had liver metastases) and were heavily pretreated, with most having received more than 1 line of chemotherapy in the metastatic setting in addition to endocrine therapy. In addition, the expected response rates in patients with breast cancer treated with targeted agents alone is generally low. For example, single-agent treatment of advanced breast cancer patients with lapatinib was associated with a low response rate but was improved when used in combination with trastuzumab (20). Therefore, the observed results with dovitinib in patients with FGFR1-amplified breast cancer may be consistent with results obtained in the same setting with successful targeted therapies in breast cancers. Additional data obtained with qPCR suggested that, within FGFR1-amplified cancers detected by SISH, those who presented higher levels of FGFR1-amplification (qPCR, ≥6 copies) could be even more sensitive to dovitinib. In these latter 7 patients, a 20.2% reduction in the mean tumor size was observed, as opposed to 14.2% increase in the mean tumor size for the 6 patients with FGFR1 amplification detected by SISH, but not qPCR. Finally, FGFR1 was not the only genomic alteration identified within the FGF pathway. In our study, FGFR2 amplifications were also associated with a higher number of responders. Indeed, tumor reductions of 28.2% and 18.5% were observed in 2 evaluable patients with FGFR2 amplification, suggesting that dovitinib could have antitumor activity in this small subset of patients. FGF3 amplification was also associated with tumor shrinkage, with 2 of the 4 FGF3-amplified patients achieving an unconfirmed partial response. Whether FGF3 is a biomarker by itself or a surrogate of high-level FGFR1 amplification is unclear because amplification of the 11q12 amplicon is more likely to occur when FGFR1 is amplified (6). In the present study, 3 of 4 patients with FGF3 amplification also had a high level of FGFR1 amplification and the fourth patient had an FGFR1-gene gain by qPCR (3.4 copies). In addition, the precise role of FGF3 in the amplicon and oncogenesis is unclear. Indeed, several other candidate oncogenic drivers are present on the amplicon including CCND1 and PAK1. Overall, the present study could not address whether FGF3 amplification, by itself, contributed to the definition of dovitinib-sensitive patients. Another limitation was that the study was not sufficiently powered for statistical analyses on FGF-pathway–amplified versus nonamplified patients.

Further studies of dovitinib will be in combination with endocrine therapy, as targeted therapies present optimal efficacy when combined with other agents. In a similar fashion as the synergy that was observed with everolimus and aromatase inhibitors (21, 22), FGFR1 inhibition has been shown to reverse endocrine resistance in preclinical models (5). As the present study did not address the role of sensitization to endocrine therapy, future combination trials should include both FGF-pathway–amplified and–nonamplified patients. Finally, additional molecular analyses will determine whether FGFR amplifications could be associated with additional mutations on oncogenes. This biomarker work could provide some rationale for combining dovitinib with other targeted agents with the aim of delaying resistance.

The safety profile of dovitinib includes gastrointestinal toxicity (nausea, vomiting), liver toxicity, and asthenia. This profile is comparable with TKIs (e.g., pazopanib, sunitinib, and lapatinib) but higher than monoclonal antibodies (23–25). Increasing awareness of these effects among physicians should accelerate detection and reduce severity of these events.

Overall, the present study provides the first detailed report examining FGFR-pathway status and response in a phase II trial testing an FGFR inhibitor. The results suggest that targeting FGFR could lead to modest antitumor activity in patients with FGF-pathway–deregulated breast cancer. On the basis of these results – together with biomarker exploration on FGFR2, FGF3, and preclinical data – dovitinib is being studied in combination with fulvestrant in a phase II randomized trial in patients with breast cancer who have FGF-pathway amplifications (FGFR1, FGFR2, or FGF3) as determined by qPCR (www.clinicaltrials.gov registry number NCT01528345). Further studies will show whether the FGF pathway should be targeted in other cancers with a deregulated pathway. For example, a phase II trial of dovitinib in metastatic endometrial cancer (www.clinicaltrials.gov registry number NCT01379534) is screening and grouping patients on the basis of FGFR2-mutation status (26). In addition, these data open new avenues in the field of FGFR targeting in other tumor types including lung and gastric cancers.

Disclosure of Potential Conflicts of Interest

S. Deudon, M. Shi, Y. Zhang, A. Kay, A. Yovine, and D. Graus Porta are employees, and F. André, T. Bachelot, N. Turner, J. Baselga, and M. Campone are consultants of Novartis Pharmaceuticals Corporation. M. Campone and H. Rugo have received commercial research grants from Novartis Pharmaceuticals Corporation. No potential conflicts of interest were disclosed by the other authors.

Authors' Contributions

Conception and design: F. André, N.C. Turner, S. Deudon, M.M. Shi, Y. Zhang, A. Kay, J. Baselga

Development of methodology: S. Deudon, M.M. Shi, Y. Zhang

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): F. André, T. Bachelot, M. Campone, F. Dalenc, S.A. Hurvitz, N.C. Turner, H. Rugo, J.W. Smith, S. Deudon, M.M. Shi, D. Graus Porta, J. Baselga

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): F. André, M. Campone, F. Dalenc, H. Rugo, M.M. Shi, Y. Zhang, A. Kay, A. Yovine, J. Baselga

Writing, review, and/or revision of the manuscript: F. André, T. Bachelot, M. Campone, J. Perez-Garcia, S.A. Hurvitz, N.C. Turner, H. Rugo, J.W. Smith, S. Deudon, M.M. Shi, Y. Zhang, A. Kay, D. Graus Porta, A. Yovine, J. Baselga

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Deudon, Y. Zhang, A. Yovine

Study supervision: S.A. Hurvitz, S. Deudon, A. Kay, A. Yovine

Grant Support

F. André is financially supported by the Operation Parrains Chercheurs. Financial support for medical editorial assistance of this article was provided by Novartis Pharmaceuticals Corporation.

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.

Acknowledgments

The authors thank Peter J. Simon, PhD, for medical editorial assistance with this article. The authors also thank Matthew Squires, for guidance and thorough review of the article, and all the investigators.

Footnotes

  • Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

  • Received January 21, 2013.
  • Revision received April 19, 2013.
  • Accepted April 28, 2013.
  • ©2013 American Association for Cancer Research.

References

  1. 1.↵
    1. Turner N,
    2. Grose R
    . Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 2010;10:116–29.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Andre F,
    2. Job B,
    3. Dessen P,
    4. Tordai A,
    5. Michiels S,
    6. Liedtke C,
    7. et al.
    Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array. Clin Cancer Res 2009;15:441–51.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    1. Elbauomy Elsheikh S,
    2. Green AR,
    3. Lambros MB,
    4. Turner NC,
    5. Grainge MJ,
    6. Powe D,
    7. et al.
    FGFR1 amplification in breast carcinomas: a chromogenic in situ hybridisation analysis. Breast Cancer Res 2007;9:R23.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Reis-Filho JS,
    2. Simpson PT,
    3. Turner NC,
    4. Lambros MB,
    5. Jones C,
    6. Mackay A,
    7. et al.
    FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas. Clin Cancer Res 2006;12:6652–62.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Turner N,
    2. Pearson A,
    3. Sharpe R,
    4. Lambros M,
    5. Geyer F,
    6. Lopez-Garcia MA,
    7. et al.
    FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res 2010;70:2085–94.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Albiges L,
    2. Quidville V,
    3. Valent A,
    4. Mathieu MC,
    5. Drusch F,
    6. Job B,
    7. et al.
    FGFR1 and FGF coamplification in breast cancer. 32nd Annual San Antonio Breast Cancer Symposium 2009 December 9–13. Cancer Res 2009;69:762S–3S.
    OpenUrl
  7. 7.↵
    1. Turner N,
    2. Lambros MB,
    3. Horlings HM,
    4. Pearson A,
    5. Sharpe R,
    6. Natrajan R,
    7. et al.
    Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets. Oncogene 2010;29:2013–23.
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. Chase A,
    2. Grand FH,
    3. Cross NCP
    . Activity of TKI258 against primary cells and cell lines with FGFR1 fusion genes associated with the 8p11 myeloproliferative syndrome. Blood 2007;110:3729–34.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Lee SH,
    2. Lopes de Menezes D,
    3. Vora J,
    4. Harris A,
    5. Ye H,
    6. Nordahl L,
    7. et al.
    In vivo target modulation and biological activity of CHIR-258, a multitargeted growth factor receptor kinase inhibitor, in colon cancer models. Clin Cancer Res 2005;11:3633–41.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    1. Simon R
    . Optimal two-stage designs for phase II clinical trials. Control Clin Trials 1989;10:1–10.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Angevin E,
    2. Lopez JA,
    3. Pande A,
    4. Moldovan C,
    5. Shi M,
    6. Soria JC,
    7. et al.
    TKI258 (dovitinib lactate) in metastatic renal cell carcinoma (mRCC) patients refractory to approved targeted therapies: a phase I/II dose finding and biomarker study. J Clin Oncol 2009;27(15 suppl):abstr 3563.
  12. 12.↵
    1. Therasse P,
    2. Arbuck SG,
    3. Eisenhauer EA,
    4. Wanders J,
    5. Kaplan RS,
    6. Rubinstein L,
    7. et al.
    New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000;92:205–16.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Marangoni E,
    2. Vincent-Salomon A,
    3. Auger N,
    4. Degeorges A,
    5. Assayag F,
    6. de Cremoux P,
    7. et al.
    A new model of patient tumor-derived breast cancer xenografts for preclinical assays. Clin Cancer Res 2007;13:3989–98.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    1. Shiang CY,
    2. Qi Y,
    3. Wang B,
    4. Lazar V,
    5. Wang J,
    6. Fraser Symmans W,
    7. et al.
    Amplification of fibroblast growth factor receptor-1 in breast cancer and the effects of brivanib alaninate. Breast Cancer Res Treat 2010;123:747–55.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Wohrle S,
    2. Bonny O,
    3. Beluch N,
    4. Gaulis S,
    5. Stamm C,
    6. Scheibler M,
    7. et al.
    FGF receptors control vitamin D and phosphate homeostasis by mediating renal FGF-23 signaling and regulating FGF-23 expression in bone. J Bone Miner Res 2011;26:2486–97.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Angevin E,
    2. Grünwald V,
    3. Ravaud A,
    4. Castellano DE,
    5. Lin C,
    6. Gschwend JE,
    7. et al.
    A phase II study of dovitinib (TKI258), an FGFR- and VEGFR-inhibitor, in patients with advanced or metastatic renal cell cancer (mRCC). J Clin Oncol 2011;29 suppl:abstr 4551.
  17. 17.↵
    1. Kim KB,
    2. Chesney J,
    3. Robinson D,
    4. Gardner H,
    5. Shi MM,
    6. Kirkwood JM
    . Phase I/II and pharmacodynamic study of dovitinib (TKI258)-an inhibitor of fibroblast growth factor receptors and VEGF receptors-in patients with advanced melanoma. Clin Cancer Res 2011;17:7451–61.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Easton DF,
    2. Pooley KA,
    3. Dunning AM,
    4. Pharoah PD,
    5. Thompson D,
    6. Ballinger DG,
    7. et al.
    Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 2007;447:1087–93.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Garcia-Closas M,
    2. Hall P,
    3. Nevanlinna H,
    4. Pooley K,
    5. Morrison J,
    6. Richesson DA,
    7. et al.
    Heterogeneity of breast cancer associations with five susceptibility loci by clinical and pathological characteristics. PLoS Genet 2008;4:e1000054.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Blackwell KL,
    2. Burstein HJ,
    3. Storniolo AM,
    4. Rugo H,
    5. Sledge G,
    6. Koehler M,
    7. et al.
    Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol 2010;28:1124–30.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    1. Boulay A,
    2. Rudloff J,
    3. Ye J,
    4. Zumstein-Mecker S,
    5. O'Reilly T,
    6. Evans DB,
    7. et al.
    Dual inhibition of mTOR and estrogen receptor signaling in vitro induces cell death in models of breast cancer. Clin Cancer Res 2005;11:5319–28.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    1. Baselga J,
    2. Campone M,
    3. Piccart M,
    4. Burris HA,
    5. Rugo HS,
    6. Sahmoud T,
    7. et al.
    Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med 2012;366:520–9.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Eisen T,
    2. Sternberg CN,
    3. Robert C,
    4. Mulders P,
    5. Pyle L,
    6. Zbinden S,
    7. et al.
    Targeted therapies for renal cell carcinoma: review of adverse event management strategies. J Natl Cancer Inst 2012;104:93–113.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    1. Nieto M,
    2. Borregaard J,
    3. Ersboll J,
    4. ten Bosch GJ,
    5. van Zwieten-Boot B,
    6. Abadie E,
    7. et al.
    The European Medicines Agency review of pazopanib for the treatment of advanced renal cell carcinoma: summary of the scientific assessment of the Committee for Medicinal Products for Human Use. Clin Cancer Res 2011;17:6608–14.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    1. Moy B,
    2. Rappold E,
    3. Williams L,
    4. Kelly T,
    5. Nicolodi L,
    6. Maltzman JD,
    7. et al.
    Hepatobiliary abnormalities in patients with metastatic cancer treated with lapatinib. ASCO Meeting Abstracts 2009;27(15 suppl):1043.
    OpenUrlAbstract
  26. 26.↵
    1. Konecny G,
    2. Finkler N,
    3. Lee P,
    4. Yovine A,
    5. Liu A,
    6. Sen P,
    7. et al.
    A multicenter, nonrandomized, open-label, single-arm phase 2 trial of dovitinib (TKI258) as second-line therapy in patients with fibroblast growth factor receptor 2-mutated or -wild-type advanced and/or metastatic endometrial cancer. Gynecol Oncol 2012;125:S80.
    OpenUrl
View Abstract
PreviousNext
Back to top
Clinical Cancer Research: 19 (13)
July 2013
Volume 19, Issue 13
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Targeting FGFR with Dovitinib (TKI258): Preclinical and Clinical Data in Breast Cancer
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Targeting FGFR with Dovitinib (TKI258): Preclinical and Clinical Data in Breast Cancer
Fabrice André, Thomas Bachelot, Mario Campone, Florence Dalenc, Jose M. Perez-Garcia, Sara A. Hurvitz, Nicholas Turner, Hope Rugo, John W. Smith, Stephanie Deudon, Michael Shi, Yong Zhang, Andrea Kay, Diana Graus Porta, Alejandro Yovine and José Baselga
Clin Cancer Res July 1 2013 (19) (13) 3693-3702; DOI: 10.1158/1078-0432.CCR-13-0190

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Targeting FGFR with Dovitinib (TKI258): Preclinical and Clinical Data in Breast Cancer
Fabrice André, Thomas Bachelot, Mario Campone, Florence Dalenc, Jose M. Perez-Garcia, Sara A. Hurvitz, Nicholas Turner, Hope Rugo, John W. Smith, Stephanie Deudon, Michael Shi, Yong Zhang, Andrea Kay, Diana Graus Porta, Alejandro Yovine and José Baselga
Clin Cancer Res July 1 2013 (19) (13) 3693-3702; DOI: 10.1158/1078-0432.CCR-13-0190
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Authors' Contributions
    • Grant Support
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Aromatase in Lung Adenocarcinomas with EGFR Mutations
  • Relative Lymphocyte Count as a Biomarker for Survival with Catumaxomab
  • Four-Gene Blood Panel Predicts Survival after Tremelimumab
Show more Predictive Biomarkers and Personalized Medicine
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement