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
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • 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
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • 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

Translational Cancer Mechanisms and Therapy

Adaptive Resistance to Dual BRAF/MEK Inhibition in BRAF-Driven Tumors through Autocrine FGFR Pathway Activation

Victoria E. Wang, Jenny Y. Xue, Dennie T. Frederick, Yi Cao, Eva Lin, Catherine Wilson, Anatoly Urisman, David P. Carbone, Keith T. Flaherty, Rene Bernards, Piro Lito, Jeff Settleman and Frank McCormick
Victoria E. Wang
1Department of Medicine, University of California, San Francisco, San Francisco, California.
2Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jenny Y. Xue
3Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
4Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York.
5Weill Cornell Medical College, Cornell University, New York, New York.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Jenny Y. Xue
Dennie T. Frederick
6Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yi Cao
7Discovery Oncology, Genentech, South San Francisco, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Eva Lin
7Discovery Oncology, Genentech, South San Francisco, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Catherine Wilson
7Discovery Oncology, Genentech, South San Francisco, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Anatoly Urisman
2Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
9The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Anatoly Urisman
David P. Carbone
10Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Keith T. Flaherty
6Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rene Bernards
10Department of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Piro Lito
3Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
4Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York.
5Weill Cornell Medical College, Cornell University, New York, New York.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jeff Settleman
7Discovery Oncology, Genentech, South San Francisco, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: Frank.mccormick@ucsf.edu jeffrey.settleman@pfizer.com
Frank McCormick
2Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: Frank.mccormick@ucsf.edu jeffrey.settleman@pfizer.com
DOI: 10.1158/1078-0432.CCR-18-2779 Published December 2019
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Article Figures & Data

Figures

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

    Pharmacologic synthetic lethal screen to identify resistance mechanism to dual BRAF and MEK inhibition. A, Drug sensitivity profile of A375-resistant lines generated by escalating drug dosage to BRAF (vemurafenib), MEK (cobimetinib), and ERK (Vx-11e) inhibitors. Cell proliferation was quantitated by CellTiter-Glo after 72 hours of drug treatment. Mean and SEM shown for three replicates. B, Western blots of pERK and multiple downstream MAPK effector pathways from A375 parental and reversibly resistant cells. Cells were grown off of drug for 24 hours, then with drug added for 4 hours, and lysed. GAPDH served as control. C, Schematic of a pharmacologic synthetic lethal screen to identify mediators of resistance to the BRAF and MEK combination. D, Screening hits are visualized by plotting the function y = the IC50 ratios of either A375 parental treated with drug X/dual resistant cells treated with drug X plus the BRAF and MEK combination (left) or A375 dual resistant cells treated with X/dual resistant cells treated with drug X plus the BRAF and MEK combination (right); x, compounds. The representative candidate compounds are indicated.

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

    FGFR inhibition synergizes with dual BRAF and MEK inhibition to enhance tumor killing through suppression of pERK. A, Top, viability of A375 cells with various FGFR inhibitors in the presence of BRAF and MEK inhibitors, as quantitated by CellTiter-Glo after 72 hours of drug treatment. Bottom, viability of Mel888 parental and dual resistant cells to BRAF and MEK inhibitors as well as to ponatinib. Viability at zero concentration for a given drug is normalized to be one-hundred percent. Mean and SEM are shown for three replicates. B, Colony formation assay with various FGFR inhibitors. BLU-554 is a FGFR4-selective inhibitor. Vemurafenib (Vem, 1.5 μmol/L) and cobimetinib (Cobi, 0.5 μmol/L) treated A375 VCR cells served as baseline control. Two different concentrations of each inhibitor [ponatinib (Pona), NVP-BGJ398, and BLU-554], 0.1 and 1 μmol/L, were tested in conjunction with vemurafenib and cobimetinib. A375 VCR cells were grown for 21 days and then stained with crystal violet. Mel888 DR cells were treated with dabrafenib (0.5 μmol/L) and trametinib (0.02 μmol/L), plus either ponatinib or NVP-BGJ398 at 0.1 and 1 μmol/L for 9 days. WM9 DR were treated with dabrafenib (1.0 μmol/L) and trametinib (0.01 μmol/L), plus either ponatinib or NVP-BGJ398 at 0.1 and 1 μmol/L for 10 days. Media were changed with fresh drug weekly. C, Western blots of A375 parental and reversibly resistant A375-VCR cells treated with ponatinib in combination with dual BRAF/MEK inhibition. Cells were grown off of drug for 24 hours, then with drug added for 4 hours, and lysed. GAPDH served as control.

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

    Conditioned media from drug treated cells confers drug resistance to BRAF and MEK inhibitors. A, Conditioned media was collected from A375 parental cells (initial plating 1 × 106million cells) treated with vehicle or vemurafenib (0.1 μmol/L) and cobimetinib (0.01 μmol/L) combination for 72 hours respectively. Fresh drug-free media was exchanged and the supernatant collected at 48 hours and 7 days later. Each conditioned media was mixed with fresh media in 2:1 ratio and used to culture A375 cells naïve to the BRAF and MEK combination. The cells were treated for 72 hours with titration of the drug combination. Cell viability was quantitated by CellTiter-Glo after 72 hours of drug treatment. Mean and SEM are shown for three replicates. B, Transcriptional profiling of FGF pathway using TaqMan real-time PCR assay. Top, A375 parental cells were treated with vemurafenib (0.1 μmol/L) and cobimetinib (0.01 μmol/L) for 72 hours. A375 VCR dual resistant cells were treated with vemurafenib (1.5 μmol/L) and cobimetinib (0.5 μmol/L) for 72 hours and compared to the parental line without drug treatment. Mean and SEM are shown for three replicates. Bottom, Mel888 parental and dual resistant cells were both treated with dabrafenib (0.5 μmol/L) and trametinib (0.02 μmol/L) for 72 hours prior to RNA extraction. In all cases, gene expression is normalized to that of the parental cells without drug treatment. C, Cellular survival of A375 parental cells treated with exogenous hFGFs and combined BRAF/MEK inhibitors for 72 hours was quantitated by CellTiter-Glo. Mean and SEM are shown for three replicates. Western blot analysis of pFGFR1 with GAPDH as control. D, ELISA was used to detect hFGF1 from supernatants of vehicle versus drug-treated A375 parental cells. E, Cellular survival of A375 parental cells treated either with a rabbit polyclonal FGF1-specific antibody (pink) or isotype control (black) and BRAF/MEK combination for 72 hours was quantitated by CellTiter-Glo. Mean and SEM are shown for three replicates.

  • Figure 4.
    • Download figure
    • Open in new tab
    • Download powerpoint
    Figure 4.

    BRAFV600E-driven tumor cells transcriptionally upregulate FGF1 in response to dual BRAF and MEK inhibition. A, Results of a customized TaqMan human FGF pathway array (Thermo Fisher Scientific). Differential expression change comparing cells treated with vemurafenib and cobimetinib for 72 hours versus the same cell line treated with vehicle calculated using the ΔΔCt method. Data represented average of three TaqMan experiments and the log2 fold change was plotted. B, Cellular survival of various BRAFV600E-driven tumor cell lines treated with exogenous hFGF1 and BRAF/MEK inhibitor combination for 72 hours was quantitated by CellTiter-Glo. Mean and SEM are shown for three replicates. C, ELISA to assay for hFGF1 in supernatant collected from several BRAFV600E-driven tumor cell lines treated with either vehicle or BRAF/MEK combination for 72 hours. D, Phospho-RTK arrays from A375 parental, VemR, CobiR, and VCR cells.

  • Figure 5.
    • Download figure
    • Open in new tab
    • Download powerpoint
    Figure 5.

    FGFR inhibition in combination of dual BRAF and MEK inhibition induces tumor regression in xenograft models. A, Mice bearing the dual resistant A375 VCR cells were treated via oral gavage with the indicated drugs for 28 days (vemurafenib at 25 mg/kg, cobimetinib at 4 mg/kg, and ponatinib at 20 mg/kg). ***, comparing the double versus the triple combination. *, comparing either vehicle or ponatinib alone versus the triple combination. Error bars represent mean and SEM. P value was calculated using the unpaired t test. B, Mice treated with the dual vemurafenib and cobimetinib combination from A were rerandomized either to continue receiving the combination at the same dose or incorporating ponatinib at 20 mg/kg for an additional 11 days. Error bars represent mean and SEM. P value calculated using the unpaired t test. C, Cell viability and colony formation assay using the triple combination of BRAF, MEK, and FGFR inhibition on the A375 cells. Vemurafenib concentration used was 0.1 μmol/L, cobimetinib 0.01 μmol/L, and ponatinib and NVP-BGJ398 0.1 μmol/L and 0.5 μmol/L, respectively. A total of 5 × 105cells were seeded initially. Media were changed with fresh drug every 3–4 days and total cells remaining were counted at each time point using the Countess II automated cell counter with Trypan blue exclusion. Mean and SEM are shown for three replicates. D, Mice bearing tumors from the A375 parental cells were grouped and treated with vehicle, vemurafenib (25 mg/kg) and cobimetinib (4 mg/kg), or the triple combination (addition of ponatinib at 20 mg/kg) for 35 days. Error bars represent mean and SEM. P value was calculated using the unpaired t test. E, IHC cross section of dual resistant A375 xenograft tumors stained with hematoxylin and eosin, pERK, and Ki-67. The mice were treated with vehicle, ponatinib, vemurafenib and cobimetinib, or the triple combination as indicated in C 4 hours prior to tumor harvesting. F, Serum was collected from A375 parental cells tumor bearing mice at baseline and after treatment with vemurafenib and cobimetinib daily for 96 hours. Serum was diluted 1:2 before running ELISA to quantitate the level of hFGF1. G, Mice bearing tumors from the Mel888 DR cells were treated with either vemurafenib (25 mg/kg) and cobimetinib (4 mg/kg), or the triple combination (addition of ponatinib at 20 mg/kg) for 32 days. Error bars represent mean and SEM. P value was calculated using the unpaired t test and adjusted for multiple comparisons.

  • Figure 6.
    • Download figure
    • Open in new tab
    • Download powerpoint
    Figure 6.

    FGF1 mRNA is increased in BRAFV600Epatient samples at progression and portends a worse prognosis. A, Plot represents log2 fold changes of progression/pretreatment nonsmall cell lung cancer samples based on normalized RNA sequencing results for FGF1. B, Patient-derived xenograft model from a patient with BRAFV600ENSCLC treated with dabrafenib and trametinib combination. Mice were grouped and treated with vehicle, vemurafenib (25 mg/kg) and cobimetinib (4 mg/kg), or the triple combination (addition of ponatinib at 20 mg/kg) for 17 days. Error bars represent mean and SEM. P value was calculated using the unpaired t test. C, Plot represents log2 fold changes of posttreatment/pre-treatment melanoma samples based on normalized RNA sequencing results for FGF1. Left, patients who were treated with a BRAF inhibitor. Right, patients receiving dual BRAF and MEK inhibitors. D, Kaplan–Meier analysis of overall survival for patients with melanoma (N = 20), stratified by intratumoral FGF1 levels. P value was calculated using the log-rank (Mantel–Cox) test.

Additional Files

  • Figures
  • Supplementary Data

    • Supplementary Figure Legends - Supplementary Figure Legends
    • Supplementary Data S1-S3 - Figure S1 shows the A375 VCR cells being resistant and addicted to dabrafenib and trametinib (A,B), that knockdown of STAT3 had no effect on drug resistance (C,D), Caspase-Glo assays of drug-treated cells (F), and time course of pFGFR1 and pERK of drug-treated A375 parental and VCR cells (A) and Mel888 parental and VCR cells (B). Figure S2 shows suppression of pERK with BGJ398, PD173074 but not with BLU-554 compound (A), that drug-treated conditioned media can confer resistant to BRAF/MEK inhibitors and this phenomenon is extinguishable over time after withdrawal of the drugs (B), and the level of FGF1 secreted in response to single agent BRAFi or MEKi treatment (c). Figure S3 shows combined BRAF/MEK inhibition results in attenuation of xenograft tumor growth through suppression of pERK (A, B) and the triple combinations used do not result in increased toxicity as manifested by change in weight (C).
    • Supplementary Data Table 1 - Table S1 shows mutations in A375 VemR cells compared to the parental using exome sequencing.
    • Supplementary Data Table 2 - Table S2 shows mutations in A375 CobiR cells compared to the parental using exome sequencing
    • Supplementary Data Table 3 - Table S3 shows unique mutations in A375 VCR cells compared to parental using exome sequencing
    • Supplementary Data Table 4 - Table S4 shows the moleculared used in the pharmacological screen and the corresponding IC50 values
    • Supplementary Data Table 5 - Table S5 shows the raw counts from RNA sequencing of paired non-small cell lung cancer samples with BRAF mutations pre- and post-treatment using BRAF/MEK inhibitor combinations
PreviousNext
Back to top
Clinical Cancer Research: 25 (23)
December 2019
Volume 25, Issue 23
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Editorial Board (PDF)

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.
Adaptive Resistance to Dual BRAF/MEK Inhibition in BRAF-Driven Tumors through Autocrine FGFR Pathway Activation
(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
Adaptive Resistance to Dual BRAF/MEK Inhibition in BRAF-Driven Tumors through Autocrine FGFR Pathway Activation
Victoria E. Wang, Jenny Y. Xue, Dennie T. Frederick, Yi Cao, Eva Lin, Catherine Wilson, Anatoly Urisman, David P. Carbone, Keith T. Flaherty, Rene Bernards, Piro Lito, Jeff Settleman and Frank McCormick
Clin Cancer Res December 1 2019 (25) (23) 7202-7217; DOI: 10.1158/1078-0432.CCR-18-2779

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Adaptive Resistance to Dual BRAF/MEK Inhibition in BRAF-Driven Tumors through Autocrine FGFR Pathway Activation
Victoria E. Wang, Jenny Y. Xue, Dennie T. Frederick, Yi Cao, Eva Lin, Catherine Wilson, Anatoly Urisman, David P. Carbone, Keith T. Flaherty, Rene Bernards, Piro Lito, Jeff Settleman and Frank McCormick
Clin Cancer Res December 1 2019 (25) (23) 7202-7217; DOI: 10.1158/1078-0432.CCR-18-2779
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
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Molecular Profiling of Cancer and Stroma Populations of PDAC
  • BCL-2 and IKKB Dependencies in Enzalutamide-Resistant CRPC
  • NAMPTi/HDACi DDR-Dependent Anti-AML Activity
Show more Translational Cancer Mechanisms and Therapy
  • 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