Immuno-PET identifies the myeloid compartment as a key contributor to the outcome of the antitumor response under PD-1 blockade

Significance Immunotherapy, especially blockade of the PD-1/PD-L1 and CTLA-4 axes, has resulted in durable responses in a range of cancers. However, responses remain heterogeneous among patients. Treatment outcome results from changes in the tumor microenvironment imposed by such blockade. Here, we use immuno-PET and single-cell RNA sequencing to increase our understanding of the dynamics of immune cells and their functional status in the tumor microenvironment in response to PD-1 blockade. Our data provide insights into the dynamics of CD8+ T cells and the functional status of the myeloid compartment in response to PD-1 blockade. Immunotherapy using checkpoint-blocking antibodies against PD-1 has produced impressive results in a wide range of cancers. However, the response remains heterogeneous among patients. We used noninvasive immuno-positron emission tomography (PET), using 89Zr-labeled PEGylated single-domain antibody fragments (nanobodies or VHHs), to explore the dynamics and distribution of intratumoral CD8+ T cells and CD11b+ myeloid cells in response to anti–PD-1 treatment in the MC38 colorectal mouse adenocarcinoma model. Responding and nonresponding tumors showed consistent differences in the distribution of CD8+ and CD11b+ cells. Anti–PD-1 treatment mobilized CD8+ T cells from the tumor periphery to a more central location. Only those tumors fully infiltrated by CD8+ T cells went on to complete resolution. All tumors contained CD11b+ myeloid cells from the outset of treatment, with later recruitment of additional CD11b+ cells. As tumors grew, the distribution of intratumoral CD11b+ cells became more heterogeneous. Shrinkage of tumors in responders correlated with an increase in the CD11b+ population in the center of the tumors. The changes in distribution of CD8+ and CD11b+ cells, as assessed by PET, served as biomarkers to gauge the efficacy of anti–PD-1 treatment. Single-cell RNA sequencing of RNA from intratumoral CD45+ cells showed that CD11b+ cells in responders and nonresponders were markedly different. The responders exhibited a dominant population of macrophages with an M1-like signature, while the CD45+ population in the nonresponders displayed an M2-like transcriptional signature. Thus, by using immuno-PET and single-cell RNA sequencing, we show that anti–PD-1 treatment not only affects interactions of CD8+ T cells with the tumor but also impacts the intratumoral myeloid compartment.

[1]  E. Nduom,et al.  Biomarkers for immunotherapy for treatment of glioblastoma , 2020, Journal for immunotherapy of cancer.

[2]  P. Allavena,et al.  Current Strategies to Target Tumor-Associated-Macrophages to Improve Anti-Tumor Immune Responses , 2019, Cells.

[3]  T. Lawrence,et al.  Targeting STAT3 and STAT5 in Tumor-Associated Immune Cells to Improve Immunotherapy , 2019, Cancers.

[4]  Evan W. Newell,et al.  High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy , 2018, Cell.

[5]  Roger D Kamm,et al.  3D microfluidic ex vivo culture of organotypic tumor spheroids to model immune checkpoint blockade. , 2018, Lab on a chip.

[6]  Jennifer L. Guerriero Macrophages: The Road Less Traveled, Changing Anticancer Therapy. , 2018, Trends in molecular medicine.

[7]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[8]  G. Longmore,et al.  SNAIL1 action in tumor cells influences macrophage polarization and metastasis in breast cancer through altered GM-CSF secretion , 2018, Oncogenesis.

[9]  Zhi Wei,et al.  Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids. , 2018, Cancer discovery.

[10]  Yan Zhang,et al.  Upregulation of PD‐L1 by SPP1 mediates macrophage polarization and facilitates immune escape in lung adenocarcinoma , 2017, Experimental cell research.

[11]  R. Weinberg,et al.  Predicting the response to CTLA-4 blockade by longitudinal noninvasive monitoring of CD8 T cells , 2017, The Journal of experimental medicine.

[12]  J. Thiery,et al.  New insights into the role of EMT in tumor immune escape , 2017, Molecular oncology.

[13]  R. Weinberg,et al.  Epithelial-to-Mesenchymal Transition Contributes to Immunosuppression in Breast Carcinomas. , 2017, Cancer research.

[14]  Yoshiyuki Hizukuri,et al.  Guanylate-binding protein 5 is a marker of interferon-γ-induced classically activated macrophages , 2016, Clinical & translational immunology.

[15]  Matheus C. Bürger,et al.  Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy , 2016, Nature.

[16]  G. Freeman,et al.  Coinhibitory Pathways in Immunotherapy for Cancer. , 2016, Annual review of immunology.

[17]  Steven H. Liang,et al.  Enzyme-Mediated Modification of Single-Domain Antibodies for Imaging Modalities with Different Characteristics. , 2016, Angewandte Chemie.

[18]  A. Ribas,et al.  An Effective Immuno-PET Imaging Method to Monitor CD8-Dependent Responses to Immunotherapy. , 2016, Cancer research.

[19]  R. Weissleder,et al.  Use of 18F-2-Fluorodeoxyglucose to Label Antibody Fragments for Immuno-Positron Emission Tomography of Pancreatic Cancer , 2015, ACS central science.

[20]  Ralph Weissleder,et al.  Noninvasive imaging of immune responses , 2015, Proceedings of the National Academy of Sciences.

[21]  D. Fearon,et al.  T cell exclusion, immune privilege, and the tumor microenvironment , 2015, Science.

[22]  R. Ahmed,et al.  Reinvigorating Exhausted T Cells by Blockade of the PD-1 Pathway. , 2015, Forum on immunopathological diseases and therapeutics.

[23]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[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]  S. Gordon,et al.  The M1 and M2 paradigm of macrophage activation: time for reassessment , 2014, F1000prime reports.

[27]  H. Koblish,et al.  Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8+ T cells directly within the tumor microenvironment , 2014, Journal of Immunotherapy for Cancer.

[28]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[29]  J. Wolchok,et al.  Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti–CTLA-4 therapy against melanoma , 2013, The Journal of experimental medicine.

[30]  I. Kalomenidis,et al.  Beneficial Impact of CCL2 and CCL12 Neutralization on Experimental Malignant Pleural Effusion , 2013, PloS one.

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

[32]  Douglas Hanahan,et al.  Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment Prospects and Obstacles for Therapeutic Targeting of Function-enabling Stromal Cell Types , 2022 .

[33]  Peter Vogel,et al.  Microenvironment and Immunology Immune Inhibitory Molecules Lag-3 and Pd-1 Synergistically Regulate T-cell Function to Promote Tumoral Immune Escape , 2022 .

[34]  M. Sporn,et al.  Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. , 2011, The Journal of clinical investigation.

[35]  M. Nishimura,et al.  CD8+ tumor-infiltrating lymphocytes predict favorable prognosis in malignant pleural mesothelioma after resection , 2010, Cancer Immunology, Immunotherapy.

[36]  P. Jurek,et al.  Conjugation and radiolabeling of monoclonal antibodies with zirconium-89 for PET imaging using the bifunctional chelate p-isothiocyanatobenzyl-desferrioxamine , 2010, Nature Protocols.

[37]  P. Sinha,et al.  Myeloid-Derived Suppressor Cells: Linking Inflammation and Cancer 1 , 2009, The Journal of Immunology.

[38]  A. Gemma,et al.  Predominant infiltration of macrophages and CD8+ T Cells in cancer nests is a significant predictor of survival in stage IV nonsmall cell lung cancer , 2008, Cancer.

[39]  P. Allavena,et al.  The inflammatory micro-environment in tumor progression: the role of tumor-associated macrophages. , 2008, Critical reviews in oncology/hematology.

[40]  Loise M. Francisco,et al.  PD-1 and its ligands in T-cell immunity. , 2007, Current opinion in immunology.

[41]  Gerd Ritter,et al.  Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[42]  S. Segal,et al.  CD11b+/Gr-1+ Immature Myeloid Cells Mediate Suppression of T Cells in Mice Bearing Tumors of IL-1β-Secreting Cells1 , 2005, The Journal of Immunology.

[43]  R. Proia,et al.  The S1P-analog FTY720 differentially modulates T-cell homing via HEV: T-cell-expressed S1P1 amplifies integrin activation in peripheral lymph nodes but not in Peyer patches. , 2005, Blood.

[44]  R. Gibbs,et al.  Genomic segmental polymorphisms in inbred mouse strains , 2004, Nature Genetics.

[45]  W. Kuziel,et al.  CC Chemokine Ligand 3 (CCL3) Regulates CD8+-T-Cell Effector Function and Migration following Viral Infection , 2003, Journal of Virology.

[46]  S. Goerdt,et al.  Alternatively Activated Macrophages Differentially Express Fibronectin and Its Splice Variants and the Extracellular Matrix Protein βIG‐H3 , 2001, Scandinavian journal of immunology.

[47]  M. Bally,et al.  The role of tumor-associated macrophages in the delivery of liposomal doxorubicin to solid murine fibrosarcoma tumors. , 1997, The Journal of pharmacology and experimental therapeutics.

[48]  Mark M. Davis,et al.  T-cell antigen receptor genes and T-cell recognition , 1988, Nature.

[49]  A. Paulen A time for reassessment. , 1986, Cancer nursing.

[50]  L. Hood,et al.  The human t cell antigen receptor is encoded by variable, diversity, and joining gene segments that rearrange to generate a complete V gene , 1984, Cell.