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
    • 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
  • 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

Translational Cancer Mechanisms and Therapy

Macrophage-Derived CXCL9 and CXCL10 Are Required for Antitumor Immune Responses Following Immune Checkpoint Blockade

Imran G. House, Peter Savas, Junyun Lai, Amanda X.Y. Chen, Amanda J. Oliver, Zhi L. Teo, Kirsten L. Todd, Melissa A. Henderson, Lauren Giuffrida, Emma V. Petley, Kevin Sek, Sherly Mardiana, Tuba N. Gide, Camelia Quek, Richard A. Scolyer, Georgina V. Long, James S. Wilmott, Sherene Loi, Phillip K. Darcy and Paul A. Beavis
Imran G. House
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Peter Savas
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
3Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Junyun Lai
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Amanda X.Y. Chen
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Amanda J. Oliver
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zhi L. Teo
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
3Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kirsten L. Todd
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Melissa A. Henderson
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lauren Giuffrida
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Emma V. Petley
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kevin Sek
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sherly Mardiana
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tuba N. Gide
4The University of Sydney, Melanoma Institute Australia, Sydney, New South Wales, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Camelia Quek
4The University of Sydney, Melanoma Institute Australia, Sydney, New South Wales, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Richard A. Scolyer
4The University of Sydney, Melanoma Institute Australia, Sydney, New South Wales, Australia.
5Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Georgina V. Long
4The University of Sydney, Melanoma Institute Australia, Sydney, New South Wales, Australia.
6Royal North Shore Hospital, Sydney, New South Wales, Australia.
7Mater Hospital, North Sydney, New South Wales, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Georgina V. Long
James S. Wilmott
4The University of Sydney, Melanoma Institute Australia, Sydney, New South Wales, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for James S. Wilmott
Sherene Loi
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
3Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Sherene Loi
Phillip K. Darcy
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
8Department of Pathology, University of Melbourne, Parkville, Victoria, Australia.
9Department of Immunology, Monash University, Clayton, Victoria, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: paul.beavis@petermac.org phil.darcy@petermac.org
Paul A. Beavis
1Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Australia.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: paul.beavis@petermac.org phil.darcy@petermac.org
DOI: 10.1158/1078-0432.CCR-19-1868 Published January 2020
  • 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.

    CXCL9 and CXCL10 are upregulated upon dual PD-1 and CTLA-4 blockade and correlate with survival in patient cohorts treated with immune checkpoint blockade. A, C57BL/6 mice were injected subcutaneously with 5 × 105 AT-3ova cells and allowed to establish for 14 days, following which they were treated on days 14, 18, 22, and 26 with either dual treatment with anti-PD-1 (200 μg/mouse) and anti-CTLA-4 (150 μg/mouse; P+C) or isotype control (ISO; 200 μg/mouse). Data shown as the mean ± SEM of 6–8 mice per group of a representative experiment (n > 3). B, Mouse survival is shown as combined data of two independent experiments. ****, P < 0.0001 (determined by two-way ANOVA and Mantel–Cox test). C and D, Seven days posttreatment, tumors were harvested (n = 3 per group) and gene expression was analyzed by NanoString and presented as an MA plot (C). D, Tumors were cultured in PBS for 4 hours at 37°C (2 μL per mg of tissue) and chemokine expression determined by cytometric bead array. *, P < 0.05; **, P < 0.005; multiple t test. N = 18–19 mice per group. E–H, Analysis of patient datasets as described in ref. 23. Differential gene expression analysis comparing responders (n = 6) to nonresponders (n = 3) in baseline (E) and on-treatment biopsies from patients with melanoma receiving combination nivolumab/pembrolizumab and ipilimumab (F). Benjamini–Hochberg Padj values shown. G, Kaplan–Meier curves showing overall survival of 72 patients with melanoma treated with anti-PD-1 or anti-PD-1 and anti-CTLA-4 therapy stratified by high and low expression of indicated chemokines. Survival curves were compared with the log-rank test. H, Correlation of expression of indicated chemokine and numbers of intratumoral CD8+ T cells in 56 biopsies of metastatic melanoma taken prior to checkpoint blockade, with each case annotated by response to therapy. Correlation tested with Pearson product moment correlation coefficient.

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

    CXCR3 blockade abrogates therapeutic efficacy of dual PD-1 and CTLA-4 blockade. A, C57BL/6 mice were injected subcutaneously with 5 × 105 AT-3ova cells and allowed to establish for 14 days before dual treatment with anti-PD-1 (200 μg/mouse) and anti-CTLA-4 (150 μg/mouse) ± anti-CXCR3 (200 μg/mouse). CXCR3 blockade was given on days 13, 14, 18, 22, and 26. Tumor growth (B) and survival (C) was assessed. Data shown as the mean ± of 6–8 mice/group of a representative experiment of n = 3. Survival is shown as combined data of two independent experiments. **, P < 0.005; *, P < 0.05 (determined by two-way ANOVA and Mantel–Cox test). Data points for ISO and P+C treated are included in Fig. 1B. D and E, Representative sizes (D) and weights (E) of tumors harvested 7 days posttreatment. A representative experiment of n > 3 is shown. F–H, C57BL/6 mice were injected with 3 × 105 MC38 cells (s.c.) and allowed to establish for 12 days (n = 6 mice per group). A total of 1.5 × 105 E0771 cells (intra mammary fat pad) and allowed to establish for 3 days (n = 5–8 per group of representative experiment n = 2; G) or 2 × 105 B16F10-OVAdim cells were injected (s.c.; H) and allowed to establish for 4 days (n = 16–17 mice per group) before dual treatment with anti-PD-1/anti-CTLA-4 ± anti-CXCR3, as per B, except for MC38, which received only two doses of combined therapy.

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

    CXCR3 blockade inhibits dual PD-1 and CTLA-4 blockade-induced CD8+ T-cell migration and activation. C57BL/6 mice were injected subcutaneously with 5 × 105 AT-3ova cells and allowed to establish for 14 days before dual treatment with anti-PD-1 and anti-CTLA-4 ± anti-CXCR3 as per Fig. 2. On day 21 (7 days posttreatment) TIL infiltrate was analyzed as a percentage of CD45+ cells; CD8+ T cells (A), CD4+foxp3− (B), NK1.1+ cells (C), and CD4+foxp3+ cells (D). E, T-cell infiltrate quantified by immunofluorescence staining. Shown are representative images and quantification of three tumors per group. F, IFNγ and TNFα expression in CD8+ and CD4+foxp3− T cells was assessed. Shown are concatenated data from 6 mice (left) and quantification from two independent experiments (right). G, Ki67, PD-1, and TIM3 expression was determined in CD8+ and CD4+foxp3− T cells. A–D, F, and G, Data represent combined data of two independent experiments with n = 11–15 per group. H, Where indicated, mice were dosed with either the S1PR1 inhibitor FTY720 (25 mg/kg) or DMSO control on days 13 and 14 post tumor inoculation, and every 2 days subsequently. Right, numbers of CD8+ and CD4+ T cells collected from blood three days post initial FTY720 administration. Data are represented as the mean ± SEM of 6–7 mice per group (****, P < 0.0001; ***, P < 0.001; **, P < 0.01 one-way; n.s., not significant, one-way ANOVA or two-way ANOVA).

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

    CXCR3 and its ligands modulate T-cell priming within the tumor DLNs. DLNs were harvested from AT-3ova tumor bearing C57BL/6 mice following treatment with dual anti-PD-1 and anti-CTLA-4 ± anti-CXCR3 as per Fig. 2. A, Indicated immune cell populations were isolated using FACS and CXCL9, CXCL10, and CXCR3 expression was assessed by qRT-PCR. Expression is shown relative to the Rpl27 housekeeping gene and is representative of triplicate qRT-PCR reactions pooled from n = 4 mice per group. B–F, CD8+ and CD4+foxp3− T cells were assessed by flow cytometry for expression of CXCR3 (B), IFNγ (C), Ki67 (D), Tbet (E), and PD-1 [F;*, P < 0.01; **, P < 0.01; n.s., not significant, one-way ANOVA (C–F) or unpaired t test (B)]. Data are representative of n = 12 per group from two pooled experiments.

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

    Macrophages are the major source of intratumoral CXCL9 and CXCL10 following dual combination therapy. C57BL/6 mice were injected subcutaneously with either 5 × 105 control AT-3ova (Mock KO) or CXCL9/CXCL10 double KO AT-3ova cells and allowed to establish for 14 days (A). Mice were then treated with either dual anti-PD-1 (200 μg/mouse) and anti-CTLA-4 (150 μg/mouse) or isotype control (200 μg/mouse). Treatment was repeated on days 18, 22, and 26. On day 21, tumors were collected from mice and CXCL9 and CXCL10 production assessed by chemokine bead array as per Fig. 1A and B. Data shown are the mean ± SEM of 5–6 mice per group, of a representative experiment (n = 2). *, P < 0.05, n.s., not significant assessed by one-way ANOVA. C, D, and F, Intracellular CXCL9 staining was performed ex vivo in the presence of golgi plug/golgi stop for 3 hours at 37°C. n = 3 mice per group, two representative experiments. C, CXCL9 production per cell type before and after therapy. D, CXCL9 staining in indicated populations shown as representative histograms. E, Tissue enrichment scores for the top 200 genes upregulated in tumors treated with anti-PD-1 and anti-CTLA-4 relative to isotype control treated mice. F, Pie charts indicating the source of CXCL9 by cell type. Each pie represents one mouse from a representative experiment. G, C57BL/76 mice were injected with MC38 subcutaneously and 19 days postinoculation expression of CXCL9 by tumor-infiltrating myeloid cells was determined. H, Expression of Cxcl9, Cxcl10, Adgrel (F4/80), Fcgr1 (CD64), and Itgae (CD103) in CT26 tumor-infiltrating immune cells as determined by single-cell RNA-seq (35). UMAP embedding of single cells in macrophage cluster (larger population) and fibroblast cluster (smaller population bottom left) identified as per the original study are shown, with color intensity representing normalized gene expression level.

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

    IFNγ is sufficient to induce CXCL9 expression from intratumoral macrophages. A, Correlation of IFNγ or TNFα and CXCL9/CXCL10 production as determined by cytometric bead array of tumor samples obtained as per Fig. 1. Data is represented as n = 6 mice per group from two independent experiments. R2 values determined by linear regression analysis. B, IFNγ and TNFα gene signature enrichment scores for CXCL9- and CXCL10-high tumors in patients with urothelial cancer treated with atezolizumab (ImVigor210 trial). C, AT-3ova tumor-bearing mice were treated as per Fig. 5A with the additional treatment of anti-IFNγ or anti-TNFα (250 μg/mouse) on days 14 and 18. Expression of CXCL9 in MoMacs and CD103+ dendritic cells determined in n = 6 mice per group. D and E, AT-3ova tumor-bearing C57BL/6 wild-type or BATF3−/− mice were treated as per Fig. 5A. Seven days posttreatment, tumor-infiltrating lymphocytes were dissociated and stimulated for 3 hours with 1 ng/mL IFNγ and/or TNFα in the presence of Golgi Plug/Stop. Expression of CXCL9 in Mo/Macs or CD103+ DCs was determined by flow cytometry (*, P < 0.05; **, P < 0.01; ***, P < 0.001, not significant, one-way ANOVA).

  • Figure 7.
    • Download figure
    • Open in new tab
    • Download powerpoint
    Figure 7.

    Generation of CXCR3 ligands by macrophages is critical for the efficacy of immune checkpoint blockade. A–C, C57BL/6 mice were injected subcutaneously with 5 × 105 AT-3ova cells and allowed to establish for 14 days before dual treatment with anti-PD-1, anti-CTLA-4 and/or anti-F4/80 (100 μg/mouse) as per Fig. 2. A, Tumor size (left) and survival (right) of n = 6 mice per group from a representative experiment of n = 2. B, CXCL9 production by tumors ex vivo following therapy, n = 3–7 per group. C, FACS plots from concatenated samples (top) and individual data points (n = 8–14 per group; bottom) are shown. Expression of CXCL9, CXCL10, CXCL11, CD68, and CLEC9a by cell subset in all patients at baseline in a cohort of patients with metastatic melanoma (D) and a cohort of patients with lung cancer (E) that underwent single-cell RNASeq (melanoma; 5,928 cells from 19 patients, lung carcinoma; Violin plots are shown with single-cell expression values overlayed). F, Single-cell differential gene expression analysis in macrophages in baseline melanoma samples from responders versus nonresponders. G, Box plot of CXCL9, CXCL10, and CXCL11 expression level in individual macrophages from baseline melanoma cases in relation to immune checkpoint blockade response. Significance determined by unpaired t test. H, Proportion of macrophages displaying chemokine signature in responders and nonresponders.

Additional Files

  • Figures
  • Supplementary Data

    • Supplementary Figure Legends - Supplementary Figure Legends
    • Figure S1 - Figure S1 shows the response of AT-3 ova tumors to single agent anti-PD-1 or anti-CTLA-4 and the upregulation of chemokine receptors or chemokines following dual or single agent therapy.
    • Figure S2 - Figure S2 shows the expression of chemokines following single agent anti-PD-1 therapy in patients and their association with patient prognosis and immune cell infiltrate.
    • Figure S3 - Figure S3 shows the association between the expression of chemokines/ chemokine receptors and patient prognosis and immune cell infiltrate.
    • Figure S4 - Figure S4 shows the effect of CXCR3 blockade on the number and phenotype of tumor-infiltrating immune cells post combination therapy
    • Figure S5 - Figure S5 shows the expression of CXCL9 and CXCL10 mRNA and protein by AT-3ova tumor cells following stimulation with IFNgamma and TNFalpha
    • Figure S6 - Figure S6 shows the phenotype and expression of CXCL9 and CXCL10 by tumor-infiltrating immune cells following treatment with anti-PD-1 and anti-CTLA-4
    • Figure S7 - Figure S7 shows the gene expression profile of macrophages and other immune cell subsets in cancer patients
PreviousNext
Back to top
Clinical Cancer Research: 26 (2)
January 2020
Volume 26, Issue 2
  • 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.
Macrophage-Derived CXCL9 and CXCL10 Are Required for Antitumor Immune Responses Following Immune Checkpoint Blockade
(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
Macrophage-Derived CXCL9 and CXCL10 Are Required for Antitumor Immune Responses Following Immune Checkpoint Blockade
Imran G. House, Peter Savas, Junyun Lai, Amanda X.Y. Chen, Amanda J. Oliver, Zhi L. Teo, Kirsten L. Todd, Melissa A. Henderson, Lauren Giuffrida, Emma V. Petley, Kevin Sek, Sherly Mardiana, Tuba N. Gide, Camelia Quek, Richard A. Scolyer, Georgina V. Long, James S. Wilmott, Sherene Loi, Phillip K. Darcy and Paul A. Beavis
Clin Cancer Res January 15 2020 (26) (2) 487-504; DOI: 10.1158/1078-0432.CCR-19-1868

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Macrophage-Derived CXCL9 and CXCL10 Are Required for Antitumor Immune Responses Following Immune Checkpoint Blockade
Imran G. House, Peter Savas, Junyun Lai, Amanda X.Y. Chen, Amanda J. Oliver, Zhi L. Teo, Kirsten L. Todd, Melissa A. Henderson, Lauren Giuffrida, Emma V. Petley, Kevin Sek, Sherly Mardiana, Tuba N. Gide, Camelia Quek, Richard A. Scolyer, Georgina V. Long, James S. Wilmott, Sherene Loi, Phillip K. Darcy and Paul A. Beavis
Clin Cancer Res January 15 2020 (26) (2) 487-504; DOI: 10.1158/1078-0432.CCR-19-1868
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

  • OxC Accurately Identified Poor Prognosis in Ovarian Cancer
  • FGFR Inhibition Overcomes Gefitinib Resistance in cSCCs
  • Stem Cells Deliver Virotherapy to DIPG
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