Deterministic reprogramming of neutrophils within tumors

Neutrophils are increasingly recognized as key players in the tumor immune response and are associated with poor clinical outcomes. Despite recent advances characterizing the diversity of neutrophil states in cancer, common trajectories and mechanisms governing the ontogeny and relationship between these neutrophil states remain undefined. Here, we demonstrate that immature and mature neutrophils that enter tumors undergo irreversible epigenetic, transcriptional, and proteomic modifications to converge into a distinct, terminally differentiated dcTRAIL-R1+ state. Reprogrammed dcTRAIL-R1+ neutrophils predominantly localize to a glycolytic and hypoxic niche at the tumor core and exert pro-angiogenic function that favors tumor growth. We found similar trajectories in neutrophils across multiple tumor types and in humans, suggesting that targeting this program may provide a means of enhancing certain cancer immunotherapies. Editor’s summary Neutrophils are the most abundant white blood cell population in the body. Although different types of neutrophils frequently gather in the tumor microenvironment, it is currently unclear how they might coordinate to support tumor growth. Using an experimental model of pancreatic cancer, Ng et al. report populations of tumor-infiltrating neutrophils that converged to develop into a single long-lived “T3” cell subset. T3 neutrophils prompted the growth of new blood vessels, which enhanced tumor survival in areas with low oxygen and limited nutrients. Depleting T3 neutrophils or inhibiting their angiogenic function reduced tumor growth. The researchers suggest that neutrophils could be reprogrammed within the tumor microenvironment into a single functional state, and modulation of protumoral neutrophil responses may have therapeutic potential. —Priscilla N. Kelly Reprogramming of tumor-infiltrating neutrophils drives their convergent differentiation within the tumor microenvironment. INTRODUCTION Neutrophils are the first responders to infection and injury and are rapidly recruited to affected tissues in large numbers to enact their protective function. As such, neutrophils were historically perceived as a homogeneous and transient population. Recently, however, a diverse array of neutrophil states has been reported in cancer, varying in their maturation, surface marker expression, and transcript profiles. The relationship between these neutrophil states and their organization into a unified protumoral response have yet to be elucidated, limiting the therapeutic targeting of neutrophils in cancer. RATIONALE To identify the mechanisms by which disparate neutrophil states are coordinated into a concerted protumoral response, we used single-cell RNA sequencing and ATACseq (assay for transposable chromatin sequencing) on neutrophils from various organs and tumors in a murine orthotopic model of pancreatic cancer. Tumor neutrophil states identified from these analyses were validated by multiparametric flow cytometry, and spatial mapping at the RNA and protein levels were performed to reveal their localization within the pancreatic tumors. In vitro and in vivo approaches were then used to examine how the tumor environment shapes neutrophil phenotype, lifespan, and protumoral functions. RESULTS We identified three distinct neutrophil states within the tumor microenvironment, T1, T2, and T3, which were epigenetically and transcriptionally distinct from neutrophils in the bone marrow, spleen, and blood. By assessing nuclear morphology and maturation status, we determined that immature and mature neutrophils infiltrating the tumor differentiated into transitional T1 and T2 populations, respectively. T1 and T2 neutrophils underwent further reprogramming to converge into the T3 neutrophil state, which was terminally differentiated and expressed the surface marker dcTRAIL-R1. dcTRAIL-R1 up-regulation in tumor-naïve neutrophils could be induced by exposure to tumor-conditioned medium in vitro or entry into the tumor in vivo, and was accompanied by the expression of T3-specific genes. More importantly, this phenomenon was independent of their initial maturation phenotype. These findings thus underscore the capability of neutrophils to adopt a new functional phenotype, overlaying it onto their existing differentiation stage. The T3 phenotype was strongly correlated with a prolonged lifespan, with dcTRAIL-R1+ neutrophils persisting for more than 5 days within the tumor. Furthermore, T3 neutrophils were mainly localized to a unique hypoxic-glycolytic niche within the tumor, where they optimally exerted their pro-angiogenic function. This indicated that neutrophil reprogramming plays a critical role in enabling their survival under hypoxia, oxidative stress, and metabolic perturbations within the tumor microenvironment. Specifically, T3 neutrophils expressed high levels of vascular endothelial growth factor alpha (VEGFα) and substantially enhanced blood vessel formation within the tumor core, and only coinjection of T3 neutrophils with tumor cells accelerated tumor growth. Therefore, the ablation of either T3 neutrophils or VEGFα inhibits this growth enhancement. Finally, all three tumor neutrophil states were observed across mouse models and in multiple human cancers, with the T3 signature predicting poorer patient outcomes in two independent human pancreatic cancer cohorts and other solid tumor types. CONCLUSION By examining neutrophils in the context of their ontogeny, we uncovered their intrinsic flexibility in adapting to environmental signals regardless of their initial maturation stage. This implies that neutrophils infiltrating a tissue niche follow a common path, merging their different functional states into a single terminal phenotype as guided by the tissue. Within the tumor, this deterministic program likely ensures a continual supply of pro-angiogenic T3 neutrophils that fuel tumor growth. Our findings thus demonstrate how short-lived effector cells such as neutrophils effectively tailor their functions to accommodate tissue requirements, highlighting the untapped possibilities of targeting the local neutrophil response as immunotherapy. Tumor-infiltrating neutrophils undergo convergent reprogramming into pro-angiogenic neutrophils that support tumor growth. In cancer, both immature and mature neutrophils infiltrate the tumor. After entering the tumor microenvironment, these neutrophils undergo differentiation, leading to the formation of transitional populations. Through reprogramming, these populations ultimately converge into a terminal neutrophil state. Reprogrammed neutrophils strongly express VEGFα and localize to a unique hypoxic-glycolytic niche near the tumor core. This places them in an optimal position to exert their pro-angiogenic function within hypoxic and nutrient-poor tumor regions, thereby promoting tumor growth. The emergence of tumor reprogramming reflects the adaptability of neutrophils to environmental cues, allowing them to consolidate their protumoral responses.

[1]  T. Speed,et al.  Spatial analysis with SPIAT and spaSim to characterize and simulate tissue microenvironments , 2023, Nature communications.

[2]  Yongmei Yin,et al.  Proteolytic regulation of CD73 by TRIM21 orchestrates tumor immunogenicity , 2023, Science advances.

[3]  D. Lazarević,et al.  Cellular and transcriptional dynamics of human neutrophils at steady state and upon stress , 2022, Nature Immunology.

[4]  Jinyan Huang,et al.  Single-cell RNA-seq analysis reveals BHLHE40-driven pro-tumour neutrophils with hyperactivated glycolysis in pancreatic tumour microenvironment , 2022, Gut.

[5]  T. Kang,et al.  PERK reprograms hematopoietic progenitor cells to direct tumor-promoting myelopoiesis in the spleen , 2022, The Journal of experimental medicine.

[6]  M. Gerstung,et al.  Cell2location maps fine-grained cell types in spatial transcriptomics , 2022, Nature Biotechnology.

[7]  F. Ginhoux,et al.  High-throughput single-cell quantification of hundreds of proteins using conventional flow cytometry and machine learning , 2021, Science advances.

[8]  A. Bosio,et al.  MACSima imaging cyclic staining (MICS) technology reveals combinatorial target pairs for CAR T cell treatment of solid tumors , 2021, Scientific Reports.

[9]  E. Kenigsberg,et al.  Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells , 2021, Nature.

[10]  B. Walzog,et al.  Distinct transcription factor networks control neutrophil-driven inflammation , 2021, Nature Immunology.

[11]  Raphael Gottardo,et al.  Spatial transcriptomics at subspot resolution with BayesSpace , 2021, Nature Biotechnology.

[12]  B. Janela,et al.  Targeting Glycolysis in Macrophages Confers Protection Against Pancreatic Ductal Adenocarcinoma , 2021, International journal of molecular sciences.

[13]  Jason D. Buenrostro,et al.  The neutrotime transcriptional signature defines a single continuum of neutrophils across biological compartments , 2021, Nature Communications.

[14]  D. Soto-Pantoja,et al.  Functions of Thrombospondin-1 in the Tumor Microenvironment , 2021, International journal of molecular sciences.

[15]  Santiago J. Carmona,et al.  UCell: Robust and scalable single-cell gene signature scoring , 2021, bioRxiv.

[16]  S. Peters,et al.  GLUT1 Expression in Tumor-Associated Neutrophils Promotes Lung Cancer Growth and Resistance to Radiotherapy , 2021, Cancer Research.

[17]  M. Nees,et al.  CD73 facilitates EMT progression and promotes lung metastases in triple-negative breast cancer , 2021, Scientific Reports.

[18]  H. Dweep,et al.  Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice , 2021, The Journal of experimental medicine.

[19]  A. MacKinnon,et al.  The therapeutic potential of galectin-3 inhibition in fibrotic disease. , 2020, The international journal of biochemistry & cell biology.

[20]  Jessica S. Yu,et al.  Multimodal mapping of the tumor and peripheral blood immune landscape in human pancreatic cancer , 2020, Nature Cancer.

[21]  A. Dopazo,et al.  Co-option of Neutrophil Fates by Tissue Environments , 2020, Cell.

[22]  Raphael Gottardo,et al.  Integrated analysis of multimodal single-cell data , 2020, Cell.

[23]  Allon M. Klein,et al.  Tumor-Promoting Ly-6G+ SiglecFhigh Cells Are Mature and Long-Lived Neutrophils , 2020, Cell Reports.

[24]  A. Giatromanolaki,et al.  Ectonucleotidase CD73 and CD39 expression in non-small cell lung cancer relates to hypoxia and immunosuppressive pathways. , 2020, Life sciences.

[25]  P. Hegde,et al.  Suppressing neutrophil-dependent angiogenesis abrogates resistance to anti-VEGF antibody in a genetic model of colorectal cancer , 2020, Proceedings of the National Academy of Sciences.

[26]  Huy Q. Dinh,et al.  Coexpression of CD71 and CD117 Identifies an Early Unipotent Neutrophil Progenitor Population in Human Bone Marrow. , 2020, Immunity.

[27]  F. Ginhoux,et al.  Combinatorial Single-Cell Analyses of Granulocyte-Monocyte Progenitor Heterogeneity Reveals an Early Uni-potent Neutrophil Progenitor. , 2020, Immunity.

[28]  Yvan Saeys,et al.  A scalable SCENIC workflow for single-cell gene regulatory network analysis , 2020, Nature Protocols.

[29]  T. Cheng,et al.  Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection , 2020, Nature Immunology.

[30]  David E. Muench,et al.  Mouse models of neutropenia reveal progenitor-stage-specific defects , 2020, Nature.

[31]  Bjoern H Menze,et al.  Cellular and Molecular Probing of Intact Human Organs , 2020, Cell.

[32]  S. Jalkanen,et al.  Compartmentalization of adenosine metabolism in cancer cells and its modulation during acute hypoxia , 2020, Journal of Cell Science.

[33]  F. Ginhoux,et al.  Single-Cell Analysis of Human Mononuclear Phagocytes Reveals Subset-Defining Markers and Identifies Circulating Inflammatory Dendritic Cells. , 2019, Immunity.

[34]  Halli E. Miller,et al.  Immune-Checkpoint Protein VISTA Regulates Antitumor Immunity by Controlling Myeloid Cell–Mediated Inflammation and Immunosuppression , 2019, Cancer Immunology Research.

[35]  K. Kessenbrock,et al.  Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics , 2019, Science Immunology.

[36]  Anshul Kundaje,et al.  The ENCODE Blacklist: Identification of Problematic Regions of the Genome , 2019, Scientific Reports.

[37]  E. Ma,et al.  Immature Low-Density Neutrophils Exhibit Metabolic Flexibility that Facilitates Breast Cancer Liver Metastasis. , 2019, Cell reports.

[38]  B. Eynde Faculty Opinions recommendation of Fatty acid transport protein 2 reprograms neutrophils in cancer. , 2019, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[39]  Virginia Savova,et al.  Single-Cell Transcriptomics of Human and Mouse Lung Cancers Reveals Conserved Myeloid Populations across Individuals and Species. , 2019, Immunity.

[40]  W. Lu,et al.  Epithelial-Mesenchymal Plasticity in Cancer Progression and Metastasis. , 2019, Developmental cell.

[41]  N. Hockstein,et al.  Fatty acid transporter 2 reprograms neutrophils in cancer , 2019, Nature.

[42]  L. Ng,et al.  Heterogeneity of neutrophils , 2019, Nature Reviews Immunology.

[43]  T. Zander,et al.  Tumour-infiltrating neutrophils counteract anti-VEGF therapy in metastatic colorectal cancer , 2018, British Journal of Cancer.

[44]  Huy Q. Dinh,et al.  Identification of an Early Unipotent Neutrophil Progenitor with Pro-tumoral Activity in Mouse and Human Bone Marrow. , 2018, Cell reports.

[45]  Erik Sundström,et al.  RNA velocity of single cells , 2018, Nature.

[46]  Wenjie Zhu,et al.  Spleen mediates a distinct hematopoietic progenitor response supporting tumor-promoting myelopoiesis , 2018, The Journal of clinical investigation.

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

[48]  Inna Kuperstein,et al.  Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer. , 2018, Cancer cell.

[49]  Jun Yu,et al.  ATACseqQC: a Bioconductor package for post-alignment quality assessment of ATAC-seq data , 2018, BMC Genomics.

[50]  F. Ginhoux,et al.  Developmental Analysis of Bone Marrow Neutrophils Reveals Populations Specialized in Expansion, Trafficking, and Effector Functions , 2018, Immunity.

[51]  Omar K. Yaghi,et al.  Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhigh neutrophils , 2017, Science.

[52]  Nicholas A. Sinnott-Armstrong,et al.  An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues , 2017, Nature Methods.

[53]  George A. Dominguez,et al.  Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients , 2016, Science Immunology.

[54]  Peter J. Murray,et al.  Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards , 2016, Nature Communications.

[55]  Fabian J Theis,et al.  Diffusion pseudotime robustly reconstructs lineage branching , 2016, Nature Methods.

[56]  Fabian J. Theis,et al.  destiny: diffusion maps for large-scale single-cell data in R , 2015, Bioinform..

[57]  Ash A. Alizadeh,et al.  Abstract PR09: The prognostic landscape of genes and infiltrating immune cells across human cancers , 2015 .

[58]  D. Tuveson,et al.  TLR9 ligation in pancreatic stellate cells promotes tumorigenesis , 2015, The Journal of experimental medicine.

[59]  Ronit Vogt Sionov,et al.  Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. , 2015, Cell reports.

[60]  H. Moses,et al.  Role of TGF-β Signaling in Generation of CD39+CD73+ Myeloid Cells in Tumors , 2014, The Journal of Immunology.

[61]  Fidel Ramírez,et al.  deepTools: a flexible platform for exploring deep-sequencing data , 2014, Nucleic Acids Res..

[62]  B. Kuster,et al.  Clec12a is an inhibitory receptor for uric acid crystals that regulates inflammation in response to cell death. , 2014, Immunity.

[63]  N. Takakura,et al.  CD44 Expressed on Cancer‐Associated Fibroblasts Is a Functional Molecule Supporting the Stemness and Drug Resistance of Malignant Cancer Cells in the Tumor Microenvironment , 2014, Stem cells.

[64]  Robert Gentleman,et al.  Software for Computing and Annotating Genomic Ranges , 2013, PLoS Comput. Biol..

[65]  Avi Ma'ayan,et al.  Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool , 2013, BMC Bioinformatics.

[66]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[67]  J. Borghans,et al.  In vivo labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days. , 2010, Blood.

[68]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[69]  S. Weiss,et al.  Neutrophils responsive to endogenous IFN-beta regulate tumor angiogenesis and growth in a mouse tumor model. , 2010, The Journal of clinical investigation.

[70]  Davis J. McCarthy,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[71]  Gerhard Christofori,et al.  The angiogenic switch in carcinogenesis. , 2009, Seminars in cancer biology.

[72]  Fu-Tong Liu,et al.  Galectin-3 is an important mediator of VEGF- and bFGF-mediated angiogenic response , 2009, The Journal of experimental medicine.

[73]  Michelle Collazo,et al.  Subsets of Myeloid-Derived Suppressor Cells in Tumor-Bearing Mice1 , 2008, The Journal of Immunology.

[74]  J. Talmadge,et al.  Chemokine-mediated rapid turnover of myeloid-derived suppressor cells in tumor-bearing mice. , 2008, Blood.

[75]  F. Peale,et al.  Bv8 regulates myeloid-cell-dependent tumour angiogenesis , 2007, Nature.

[76]  Christopher Chiu,et al.  Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis , 2006, Proceedings of the National Academy of Sciences.

[77]  R. Hruban,et al.  Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. , 2005, Cancer cell.

[78]  B. Fingleton,et al.  Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. , 2004, Cancer cell.

[79]  M. Maio,et al.  Endoglin (CD105): a powerful therapeutic target on tumor-associated angiogenetic blood vessels , 2003, Oncogene.

[80]  D. Beis,et al.  Matrigel Plug Assay for In Vivo Evaluation of Angiogenesis. , 2019, Methods in molecular biology.

[81]  Heng Li,et al.  BIOINFORMATICS ORIGINAL PAPER , 2022 .