Combined BRAF, MEK, and CDK4/6 Inhibition Depletes Intratumoral Immune-Potentiating Myeloid Populations in Melanoma

Combined BRAF, MEK, and CDK4/6 inhibition is being tested in clinical trials for treating melanoma. The authors show this combination depletes tumor-associated myeloid cells in the tumor immune microenvironment, which renders tumors unresponsive to immune checkpoint blockade. Combined inhibition of BRAF, MEK, and CDK4/6 is currently under evaluation in clinical trials for patients with melanoma harboring a BRAFV600 mutation. While this triple therapy has potent tumor-intrinsic effects, the impact of this combination on antitumor immunity remains unexplored. Here, using a syngeneic BrafV600ECdkn2a−/−Pten−/− melanoma model, we demonstrated that triple therapy promoted durable tumor control through tumor-intrinsic mechanisms and promoted immunogenic cell death and T-cell infiltration. Despite this, tumors treated with triple therapy were unresponsive to immune checkpoint blockade (ICB). Flow cytometric and single-cell RNA sequencing analyses of tumor-infiltrating immune populations revealed that triple therapy markedly depleted proinflammatory macrophages and cross-priming CD103+ dendritic cells, the absence of which correlated with poor overall survival and clinical responses to ICB in patients with melanoma. Indeed, immune populations isolated from tumors of mice treated with triple therapy failed to stimulate T-cell responses ex vivo. While combined BRAF, MEK, and CDK4/6 inhibition demonstrates favorable tumor-intrinsic activity, these data suggest that collateral effects on tumor-infiltrating myeloid populations may impact antitumor immunity. These findings have important implications for the design of combination strategies and clinical trials that incorporate BRAF, MEK, and CDK4/6 inhibition with immunotherapy for the treatment of patients with melanoma.

[1]  E. Alnemri,et al.  Mutant BRAF and MEK inhibitors regulate the tumor immune microenvironment via pyroptosis. , 2019, Cancer discovery.

[2]  Howard Y. Chang,et al.  Clonal replacement of tumor-specific T cells following PD-1 blockade , 2019, Nature Medicine.

[3]  Rui Hou,et al.  scMatch: a single-cell gene expression profile annotation tool using reference datasets , 2019, Bioinform..

[4]  F. Hodi,et al.  Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade , 2019, Nature Immunology.

[5]  F. Souza-Fonseca-Guimaraes,et al.  A novel immunogenic mouse model of melanoma for the preclinical assessment of combination targeted and immune-based therapy , 2019, Scientific Reports.

[6]  Atul J. Butte,et al.  Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage , 2018, Nature Immunology.

[7]  Lai Guan Ng,et al.  Dimensionality reduction for visualizing single-cell data using UMAP , 2018, Nature Biotechnology.

[8]  M. Diamond,et al.  WDFY4 is required for cross-presentation in response to viral and tumor antigens , 2018, Science.

[9]  Christoph Hafemeister,et al.  Comprehensive integration of single cell data , 2018, bioRxiv.

[10]  Paul J. Hoover,et al.  Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma , 2018, Cell.

[11]  J. Allison,et al.  Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. , 2018, Cancer discovery.

[12]  R. Pearson,et al.  Palbociclib synergizes with BRAF and MEK inhibitors in treatment naïve melanoma but not after the development of BRAF inhibitor resistance , 2018, International journal of cancer.

[13]  Adrian V. Lee,et al.  An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics , 2018, Cell.

[14]  C. Paweletz,et al.  CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. , 2017, Cancer discovery.

[15]  J. Liu,et al.  Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor , 2017, Nature Immunology.

[16]  M. Ellis,et al.  CDK4/6 inhibition triggers anti-tumor immunity , 2017, Nature.

[17]  Jedd D. Wolchok,et al.  T-cell invigoration to tumour burden ratio associated with anti-PD-1 response , 2017, Nature.

[18]  Roland Eils,et al.  Complex heatmaps reveal patterns and correlations in multidimensional genomic data , 2016, Bioinform..

[19]  F. Ginhoux,et al.  Expansion and Activation of CD103(+) Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition. , 2016, Immunity.

[20]  Gianluca Bontempi,et al.  TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data , 2015, Nucleic acids research.

[21]  J. Utikal,et al.  Myeloid Cells and Related Chronic Inflammatory Factors as Novel Predictive Markers in Melanoma Treatment with Ipilimumab , 2015, Clinical Cancer Research.

[22]  A. Trautmann,et al.  Vaccine-induced tumor regression requires a dynamic cooperation between T cells and myeloid cells at the tumor site , 2015, Oncotarget.

[23]  J. Wolchok,et al.  Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. , 2015, The New England journal of medicine.

[24]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[25]  Jeffrey W Pollard,et al.  Tumor-associated macrophages: from mechanisms to therapy. , 2014, Immunity.

[26]  G. Freeman,et al.  Response to BRAF Inhibition in Melanoma Is Enhanced When Combined with Immune Checkpoint Blockade , 2014, Cancer Immunology Research.

[27]  T. Burke,et al.  The CDK4/6 Inhibitor LY2835219 Overcomes Vemurafenib Resistance Resulting from MAPK Reactivation and Cyclin D1 Upregulation , 2013, Molecular Cancer Therapeutics.

[28]  Sebastian Amigorena,et al.  Dissecting the Tumor Myeloid Compartment Reveals Rare Activating Antigen-Presenting Cells Critical for T Cell Immunity. , 2014, Cancer cell.

[29]  G. McArthur,et al.  The Cell-Cycle Regulator CDK4: An Emerging Therapeutic Target in Melanoma , 2013, Clinical Cancer Research.

[30]  R. Sullivan,et al.  BRAF Inhibition Is Associated with Enhanced Melanoma Antigen Expression and a More Favorable Tumor Microenvironment in Patients with Metastatic Melanoma , 2013, Clinical Cancer Research.

[31]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[32]  H. Mages,et al.  Expression of XCR1 Characterizes the Batf3-Dependent Lineage of Dendritic Cells Capable of Antigen Cross-Presentation , 2012, Front. Immun..

[33]  J. Wilmott,et al.  Selective BRAF Inhibitors Induce Marked T-cell Infiltration into Human Metastatic Melanoma , 2011, Clinical Cancer Research.

[34]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[35]  K. Flaherty,et al.  Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. , 2010, Cancer research.

[36]  E. Birney,et al.  Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt , 2009, Nature Protocols.

[37]  L. Schwartz,et al.  New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). , 2009, European journal of cancer.

[38]  L. Zitvogel,et al.  Calreticulin exposure dictates the immunogenicity of cancer cell death , 2007, Nature Medicine.

[39]  P. Grambsch,et al.  A Package for Survival Analysis in S , 1994 .

[40]  Elizabeth Brunk,et al.  Antigen receptor repertoire profiling from RNA-seq data , 2022 .