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
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Cancer Immunology Essentials
    • Collections
      • COVID-19 & Cancer Resource Center
      • Toolbox: Coding and Computation
      • Toolbox: Signatures and Cells
      • "Best of" Collection
      • Editors' Picks
  • 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
Cancer Immunology Research
Cancer Immunology Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Cancer Immunology Essentials
    • Collections
      • COVID-19 & Cancer Resource Center
      • Toolbox: Coding and Computation
      • Toolbox: Signatures and Cells
      • "Best of" Collection
      • Editors' Picks
  • 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

Research Articles

Response to Programmed Cell Death-1 Blockade in a Murine Melanoma Syngeneic Model Requires Costimulation, CD4, and CD8 T Cells

Blanca Homet Moreno, Jesse M. Zaretsky, Angel Garcia-Diaz, Jennifer Tsoi, Giulia Parisi, Lidia Robert, Katrina Meeth, Abibatou Ndoye, Marcus Bosenberg, Ashani T. Weeraratna, Thomas G. Graeber, Begoña Comin-Anduix, Siwen Hu-Lieskovan and Antoni Ribas
Blanca Homet Moreno
1Division of Hematology/Oncology, Department of Medicine, University of California (UCLA), Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jesse M. Zaretsky
1Division of Hematology/Oncology, Department of Medicine, University of California (UCLA), Los Angeles, California.
2Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Angel Garcia-Diaz
1Division of Hematology/Oncology, Department of Medicine, University of California (UCLA), Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jennifer Tsoi
2Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Giulia Parisi
1Division of Hematology/Oncology, Department of Medicine, University of California (UCLA), Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lidia Robert
1Division of Hematology/Oncology, Department of Medicine, University of California (UCLA), Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Katrina Meeth
3Departments of Immunobiology, Dermatology, and Pathology, Yale University School of Medicine, New Haven, Connecticut.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Abibatou Ndoye
5Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marcus Bosenberg
3Departments of Immunobiology, Dermatology, and Pathology, Yale University School of Medicine, New Haven, Connecticut.
4Howard Hughes Medical Institute, Chevy Chase, Maryland.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ashani T. Weeraratna
5Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thomas G. Graeber
2Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.
6Jonsson Comprehensive Cancer Center (JCCC) at UCLA, Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Begoña Comin-Anduix
6Jonsson Comprehensive Cancer Center (JCCC) at UCLA, Los Angeles, California.
7Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Siwen Hu-Lieskovan
1Division of Hematology/Oncology, Department of Medicine, University of California (UCLA), Los Angeles, California.
6Jonsson Comprehensive Cancer Center (JCCC) at UCLA, Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: aribas@mednet.ucla.edu shu-lieskovan@mednet.ucla.edu
Antoni Ribas
1Division of Hematology/Oncology, Department of Medicine, University of California (UCLA), Los Angeles, California.
2Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.
6Jonsson Comprehensive Cancer Center (JCCC) at UCLA, Los Angeles, California.
7Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: aribas@mednet.ucla.edu shu-lieskovan@mednet.ucla.edu
DOI: 10.1158/2326-6066.CIR-16-0060 Published October 2016
  • 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.

    Enhanced in vivo antitumor activity with anti–PD-1 or anti–PD-L1 in MC38 and YUMM2.1 tumor models compared with YUMM1.1. Tumor growth curves of MC38 (A), YUMM2.1 (B), and YUMM1.1 C, with 4 mice in each group (mean ± SD) after anti–PD-1, anti–PD-L1, or isotype control. The arrow indicates the day when treatment with anti–PD-1, anti–PD-L1, or isotype control was started. *, P < 0.001 by unpaired t test on day 20, anti–PD-1 versus isotype control, anti–PD-L1 versus isotype control in MC38, anti–PD-1 versus isotype control, anti–PD-L1 versus isotype control in YUMM2.1 tumors.

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

    IFNγ modulates PD-L1 expression in MC38, YUMM2.1, and YUMM1.1. A, Western blot analysis of PD-L1. MC38, YUMM2.1, and YUMM1.1 cells were cultured with or without IFNγ for 24 hours. B, expression of PD-L1 by flow cytometry on MC38, YUMM2.1, and YUMM1.1 cells at baseline and after 24 hours of stimulation with IFNγ. C, chromosomal copy-number variation in MC38, YUMM2.1, and YUMM1.1 cell lines. Y-axis represents Log2 depth ratio vs. matched normal.

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

    Both CD8 and CD4 cells mediate response to PD-1 blockade in MC38 and YUMM2.1. Tumor growth curves of MC38 (A) and YUMM2.1 (B) after anti–PD-1 and either anti-CD8 (anti–PD-1aCD8), anti-CD4 (anti–PD-1aCD4), anti-CD8 + anti-CD4 (anti–PD-1aCD8/4) or isotype control; 4 mice in each group, mean ± SD. (*, P < 0.001 isotype control, anti–PD-1aCD8, anti–PD-1aCD4, anti–PD-1aCD8/4 versus anti–PD-1 in MC38, P < 0.001 isotype control, anti–PD-1aCD4, anti–PD-1aCD8/4 versus anti–PD-1 in YUMM2.1, unpaired t test, n = 4); *, P = 0.003 anti–PD-1aCD8 versus anti–PD-1, unpaired t test, n = 4. The arrow indicates the day treatment with anti–PD-1 or isotype control was started. This experiment was performed in triplicate. On days 3 (d3) and 10 (d10) after treatment with anti–PD-1 or isotype control was started, MC38 and YUMM2.1 tumors were isolated and stained with fluorescent-labeled antibodies, analyzed by FACS. C and D, percentage of CD3+CD8+ (CD8 T cells) and CD3+CD4+ (CD4 T cells) in MC38 (C) and YUMM2.1. (D), tumors are shown (mean ± SD). *, P = 0.03 anti–PD-1 d10 versus control d10 in MC38; P = 0.03 anti–PD-1 d10 versus control d10 in YUMM2.1 (unpaired t test, n = 4). Results were consistent in 6 replicate experiments. E and F, statistical analysis of the 2C total number of CD8 T cells per gram of tumor in MC38 (E) and (F) YUMM2.1 tumors. *, P = 0.05 anti–PD-1 d10 versus control d10 in MC38, P = 0.02 anti–PD-1 d10 versus control d10 in YUMM2.1, unpaired t test, n = 8). G, representative immunofluorescence of CD8 T cells stained in YUMM2.1 tumors and spleens d10 after treatment with anti–PD-1 or isotype control was started.

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

    Wnt/β-catenin pathway is not involved in CD8 T-cell decrease or anti–PD-1 antitumor response in YUMM2.1 tumor model. A, Western blot analysis of β-catenin in YUMM2.1 cells transduced with shRNA without β-catenin (sh YUMM2.1) or with shβ-catenin (shβ-catenin YUMM2.1) and YUMM1.1 cells transduced with shRNA without β-catenin (sh YUMM1.1) or with shβ-catenin (shβ-catenin YUMM1.1). B, quantification of CD3+CD8+ (CD8 T cells). Tumor cells harvested on days 3 and 10 after anti–PD-1 or isotype control were counted and analyzed by flow cytometry for CD3/CD8 staining; 3 mice in each group (mean ± SD). *, P = 0.003 anti–PD-1 d10 versus control d10 in sh-control YUMM2.1 tumors. C, shβ-catenin YUMM2.1 tumors. *, P = 0.008 anti–PD-1 d10 versus control d10 in shβ-catenin YUMM2.1 tumors, unpaired t test, n = 4. (D ) in vivo sh and shβ-catenin YUMM2.1 and (E) sh and shβ-catenin YUMM1.1 tumor growth curves with 3 to 4 mice in each group (mean ± SD) after anti–PD-1 or isotype control.

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

    Increased antigen-presenting DCs in anti–PD-1-treated YUMM2.1 tumors. A, tumor growth curves of CD28KO or C57BL/6 mice bearing YUMM2.1 treated with anti–PD-1 or isotype control. B, tumor growth curves of CD80/86KO or C57BL/6 mice bearing YUMM2.1 treated with anti–PD-1 or isotype control. Four mice in each group (mean ± SD). The arrow indicates the day treatment with anti–PD-1 or isotype control was initiated. C, on day 10 after starting treatment, MC38, YUMM2.1, and YUMM1.1 tumors were isolated and stained with fluorescent-labeled antibodies and analyzed by FACS, with 3 mice in each group (mean ± SD). B220− and B220+ cells presented as percentage of CD11c+ cells. *, P = 0.04 anti–PD-1 versus isotype control, CD11c+B220− cells in MC38 tumors, unpaired t test, n = 3. D, B220−CD8+ and B220−CD103+ presented as percentage of CD11c+ cells. E, in vivo YUMM2.1 growth curve after anti–PD-1 ± anti-CD103 or isotype control ± anti-CD103, 4 mice in each group (mean ± SD). The arrow indicates the day anti–PD-1 or isotype control treatment was started. F, CD11b+ and CD11b+MHC-IIhigh DCs presented as percentage of CD11c+ cells. *, P = 0.04 anti–PD-1 versus control, P = 0.01 anti–PD-1 versus control in YUMM2.1 tumors, unpaired t test, n = 3.

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

    Modulation of the tumor microenvironment by anti–PD-1 in MC38, YUMM2.1, and YUMM1.1. On day 10 after anti–PD-1 or isotype control, MC38, YUMM2.1, and YUMM1.1 tumors were isolated and stained with fluorescent-labeled antibodies and analyzed by FACS, with 3 mice in each group (mean ± SD). A, analysis of TAMs (CD11b+F4/80+). B, TAMs MHC-IIhigh (M1 TAMs, CD11b+F4/80+MHC-IIhigh) and TAMs MHC-IIlow (M2 TAMs, CD11b+F4/80+MHC-IIlow). *, P = 0.04 anti–PD-1 d10 versus control d10 TAMs; P = 0.02 anti–PD-1 d10 versus control d10 TAMs MHC-IIhigh in YUMM2.1 tumors, unpaired t test, n = 3. C, MO-MDSC (CD11b+Ly6ChighLy6Glow) and PMN-MDSC (CD11b+Ly6ClowLy6Ghigh) presented as percentage of CD11b+ cells. D, analysis of Tregs (CD4+CD25+FOXp3+). E, representative FACS plots in tumors.

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

    YUMM2.1 is more inherently immune permissive than YUMM1.1. A, Gene Set Enrichment Analysis (GSEA) curves for YUMM2.1 versus YUMM1.1 enriched pathways involved in immune response, cytokine production, and inflammatory response. B, corresponding Normalized Enrichment Score (NES), P values, and False Discovery Rate (FDR) of the GSEA plots.

Additional Files

  • Figures
  • Supplementary Data

    • Supplementary figure legends - Supplementary figure legends
    • Supplementary Figure 1 - IGV plot exome sequencing from YUMM1.1, YUMM1.7 and YUMM2.1, tumor growth curve of YUMM1.7 and B16 with 4 mice in each group, analysis of the non-synonymous mutational load compared to a strain-matched normal with known dbSNP variants excluded.
    • Supplementary Figure 2 - Quantification of CD3+CD8+ (CD8 T-cells) and (B) CD3+CD4+ (CD4 T-cells) in both tumors and spleens in YUMM2.1; gating strategy after exclusion of dead cells of CD3+CD8+ and CD3+CD4+ cells after gating for CD3+ cells; quantification of CD3+CD8+ (CD8 T-cells) in MC38 and YUMM2.1 spleens; CD3+CD4+ (CD4 T-cells) in MC38 and YUMM 2.1 spleens CD8 T-cells in both tumors and spleens in YUMM1.1.
    • Supplementary Figure 3 - IGV plot of RNA-Seq from YUMM1.1, YUMM1.7 and YUMM2.1; western blot analysis of cytoplasmic and nuclear beta-catenin in YUMM1.7 and YUMM2.1 cell lines with or without exposure to 10 uM 4HT for 48 hours; top-flash activity of total beta-catenin in YUMM1.7 and YUMM2.1 with or without exposure to 10uM 4HT for 48 hours; representative immunofluorescence of beta-catenin stained non-treated tumors.
    • Supplementary Figure 4 - Gating strategy of CD11c+B220-, CD11c+B220+, CD11c+B220-CD8+ and CD11c+B220-CD103+ cells; gating strategy of CD11b+MHC-IIhigh DCs in CD11c+ cells; gating strategy of CD11b+F4/80+TAMs, CD11b+F4/80+MHC-IIlow TAMs and CD11b+F4/80+MHC-IIhigh TAMs; gating strategy of MO-MDSC (CD11b+Ly6ChighLy6Glow) and PMN-MDSC (CD11b+Ly6ClowLy6Ghigh); gating strategy of Tregs (CD4+CD25+FoxP3+); corresponding normalized enrichment scores (NES), P values and false discovery rates (FDR) of the GSEA plots for YUMM2.1 versus YUMM1.1 enriched pathways involved in immune response, cytokine production and inflammatory response.
PreviousNext
Back to top
Cancer Immunology Research: 4 (10)
October 2016
Volume 4, Issue 10
  • 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 Cancer Immunology 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.
Response to Programmed Cell Death-1 Blockade in a Murine Melanoma Syngeneic Model Requires Costimulation, CD4, and CD8 T Cells
(Your Name) has forwarded a page to you from Cancer Immunology Research
(Your Name) thought you would be interested in this article in Cancer Immunology Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Response to Programmed Cell Death-1 Blockade in a Murine Melanoma Syngeneic Model Requires Costimulation, CD4, and CD8 T Cells
Blanca Homet Moreno, Jesse M. Zaretsky, Angel Garcia-Diaz, Jennifer Tsoi, Giulia Parisi, Lidia Robert, Katrina Meeth, Abibatou Ndoye, Marcus Bosenberg, Ashani T. Weeraratna, Thomas G. Graeber, Begoña Comin-Anduix, Siwen Hu-Lieskovan and Antoni Ribas
Cancer Immunol Res October 1 2016 (4) (10) 845-857; DOI: 10.1158/2326-6066.CIR-16-0060

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Response to Programmed Cell Death-1 Blockade in a Murine Melanoma Syngeneic Model Requires Costimulation, CD4, and CD8 T Cells
Blanca Homet Moreno, Jesse M. Zaretsky, Angel Garcia-Diaz, Jennifer Tsoi, Giulia Parisi, Lidia Robert, Katrina Meeth, Abibatou Ndoye, Marcus Bosenberg, Ashani T. Weeraratna, Thomas G. Graeber, Begoña Comin-Anduix, Siwen Hu-Lieskovan and Antoni Ribas
Cancer Immunol Res October 1 2016 (4) (10) 845-857; DOI: 10.1158/2326-6066.CIR-16-0060
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
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Lenalidomide Turns Myeloma-Associated Macrophage Tumoricidal
  • Inflammasome-Independent IL1β Inhibits Antitumor Immunity
  • EDB-CAR T Cells for Solid Tumors
Show more Research Articles
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook   Twitter   LinkedIn   YouTube   RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Cancer Immunology Essentials

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Immunology Research

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

Copyright © 2021 by the American Association for Cancer Research.

Cancer Immunology Research
eISSN: 2326-6074
ISSN: 2326-6066

Advertisement