Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Anti-CTLA-4 antibodies drive myeloid activation and reprogram the tumor microenvironment through FcγR engagement and type I interferon signaling

An Author Correction to this article was published on 14 November 2022

This article has been updated

Abstract

Despite the clinical success of checkpoint inhibitors, a substantial gap still exists in our understanding of their mechanism of action. While antibodies to cytotoxic T lymphocyte-associated protein-4 (CTLA-4) were developed to block inhibitory signals in T cells, several recent studies have demonstrated that Fcγ receptor (FcγR)-dependent depletion of regulatory T cells (Treg) is critical for antitumor activity. Here, using single-cell RNA sequencing, we dissect the impact of anti-CTLA-4-blocking, Treg cell-depleting and FcR-engaging activity on the immune response within tumors. We observed a rapid remodeling of the innate immune landscape as early as 24 h after treatment. Using genetic Treg cell ablation models, we show that immune remodeling was not driven solely by Treg cell depletion or CTLA-4 blockade but mainly through FcγR engagement, downstream activation of type I interferon signaling and reduction of suppressive macrophages. Our findings indicate that FcγR engagement and innate immune remodeling are involved in successful anti-CTLA-4 treatment, supporting the development of optimized immunotherapy agents bearing these features.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Characterization of immune dynamics in the TME following anti-CTLA-4 treatments.
Fig. 2: Anti-CTLA-4 m2a treatment drives rapid and broad rearrangement of the adaptive and innate immune TME.
Fig. 3: Molecular dissection of the immediate response to mAbs to CTLA-4.
Fig. 4: FcγR-dependent activation underlies myeloid reprogramming following anti-CTLA-4 m2a treatment.
Fig. 5: FcγR engagement drives a robust activation of type I IFN signaling.

Similar content being viewed by others

Data availability

Raw and processed scRNA-seq data can be downloaded from NCBI Gene Expression Omnibus with accession number GSE186484. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

Custom code has been deposited to GitHub and can be accessed at https://github.com/tomerlan/paper_aCTLA4_MOA.

Change history

References

  1. Sharma, P. et al. The next decade of immune checkpoint therapy. Cancer Discov. 11, 838–857 (2021).

    Article  CAS  PubMed  Google Scholar 

  2. Weiss, S. A., Wolchok, J. D. & Sznol, M. Immunotherapy of melanoma: facts and hopes. Clin. Cancer Res. 25, 5191–5201 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hegde, P. S. & Chen, D. S. Top 10 challenges in cancer immunotherapy. Immunity 52, 17–35 (2020).

    Article  CAS  PubMed  Google Scholar 

  4. Walker, L. S. K. & Sansom, D. M. Confusing signals: recent progress in CTLA-4 biology. Trends Immunol. 36, 63–70 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Walker, L. S. K. Treg and CTLA-4: two intertwining pathways to immune tolerance. J. Autoimmun. 45, 49–57 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 (1996).

    Article  CAS  PubMed  Google Scholar 

  7. Simpson, T. R. et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 210, 1695–1710 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Arce Vargas, F. et al. Fc effector function contributes to the activity of human anti-CTLA-4 antibodies. Cancer Cell 33, 649–663 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bulliard, Y. et al. Activating Fcγ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J. Exp. Med. 210, 1685–1693 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Selby, M. J. et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol. Res. 1, 32–42 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Quezada, S. A. & Peggs, K. S. Lost in translation: deciphering the mechanism of action of anti-human CTLA-4. Clin. Cancer Res. 25, 1130–1132 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Yofe, I., Dahan, R. & Amit, I. Single-cell genomic approaches for developing the next generation of immunotherapies. Nat. Med. 26, 171–177 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Giladi, A. & Amit, I. Single-cell genomics: a stepping stone for future immunology discoveries. Cell 172, 14–21 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Mulder, K. et al. Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity 54, 1883–1900 (2021).

    Article  CAS  PubMed  Google Scholar 

  15. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Ledergor, G. et al. Single cell dissection of plasma cell heterogeneity in symptomatic and asymptomatic myeloma. Nat. Med. 24, 1867–1876 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Cohen, Y. C. et al. Identification of resistance pathways and therapeutic targets in relapsed multiple myeloma patients through single-cell sequencing. Nat. Med. 27, 491–503 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Katzenelenbogen, Y. et al. Coupled scRNA-seq and intracellular protein activity reveal an immunosuppressive role of TREM2 in cancer. Cell 182, 872–885 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Giladi, A. et al. Dissecting cellular crosstalk by sequencing physically interacting cells. Nat. Biotechnol. 38, 629–637 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Nimmerjahn, F. & Ravetch, J. V. Fcγ receptors as regulators of immune responses. Nat. Rev. Immunol. 8, 34–47 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Jeong, H. J. et al. Myeloid-derived suppressor cells mediate inflammation resolution in humans and mice with autoimmune uveoretinitis. J. Immunol. 200, 1306–1315 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Veglia, F., Sanseviero, E. & Gabrilovich, D. I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 21, 485–498 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mehta, A. K., Kadel, S., Townsend, M. G., Oliwa, M. & Guerriero, J. L. Macrophage biology and mechanisms of immune suppression in breast cancer. Front. Immunol. 12, 643771 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Shao, X. et al. CellTalkDB: a manually curated database of ligand–receptor interactions in humans and mice. Brief. Bioinform. 22, bbaa269 (2021).

    Article  PubMed  Google Scholar 

  25. Molgora, M. et al. TREM2 modulation remodels the tumor myeloid landscape enhancing anti-PD-1 immunotherapy. Cell 182, 886–900 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Deczkowska, A., Weiner, A. & Amit, I. The physiology, pathology, and potential therapeutic applications of the TREM2 signaling pathway. Cell 181, 1207–1217 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Nagarsheth, N., Wicha, M. S. & Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 17, 559–572 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Clark, N. M. et al. Regulatory T cells support breast cancer progression by opposing IFN-γ-dependent functional reprogramming of myeloid cells. Cell Rep. 33, 108482 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim, J. M., Rasmussen, J. P. & Rudensky, A. Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8, 191–197 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Dhodapkar, K. M. et al. Selective blockade of the inhibitory Fcγ receptor (FcγRIIB) in human dendritic cells and monocytes induces a type I interferon response program. J. Exp. Med. 204, 1359–1369 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Riaz, N. et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 171, 934–949 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Dolgin, E. Antibody engineers seek optimal drug targeting TIGIT checkpoint. Nat. Biotechnol. 38, 1007–1009 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Romano, E. et al. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc. Natl Acad. Sci. USA 112, 6140–6145 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mujal, A. M. et al. Holistic characterization of tumor monocyte-to-macrophage differentiation integrates distinct immune phenotypes in kidney cancer. Cancer Immunol. Res. 10, 403–419 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sharma, A. et al. Anti-CTLA-4 immunotherapy does not deplete FOXP3+ regulatory T cells (Tregs) in human cancers. Clin. Cancer Res. 25, 1233–1238 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Smith, P., DiLillo, D. J., Bournazos, S., Li, F. & Ravetch, J. V. Mouse model recapitulating human Fcγ receptor structural and functional diversity. Proc. Natl Acad. Sci. USA 109, 6181–6186 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Prinz, M. et al. Distinct and nonredundant in vivo functions of IFNAR on myeloid cells limit autoimmunity in the central nervous system. Immunity 28, 675–686 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Deczkowska, A. et al. XCR1+ type 1 conventional dendritic cells drive liver pathology in non-alcoholic steatohepatitis. Nat. Med. 27, 1043–1054 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Karlsson, F., Robinson-Jackson, S. A., Gray, L., Zhang, S. & Grisham, M. B. Ex vivo generation of regulatory T cells: characterization and therapeutic evaluation in a model of chronic colitis. Methods Mol. Biol. 677, 47–61 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Keren-Shaul, H. et al. MARS-seq2.0: an experimental and analytical pipeline for indexed sorting combined with single-cell RNA sequencing. Nat. Protoc. 14, 1841–1862 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 1–15 (2019).

    Article  Google Scholar 

  45. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wilkerson, D. M. & Hayes, N. D. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinform. 26, 1572–1573 (2010).

    Article  CAS  Google Scholar 

  47. Korotkevich, G., Sukhov, V. & Sergushichev, A. (2019). Fast gene set enrichment analysis. bioRxiv. http://biorxiv.org/content/early/2016/06/20/060012

  48. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinform. 27, 1739–1740 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank N.D. Geller from the Scientific Illustration unit of the Weizmann Institute for artwork. I.Y. is a Cancer Research Institute Irvington Fellow supported by the Cancer Research Institute. I.A. is an Eden and Steven Romick Professorial Chair, supported by Merck KGaA, Darmstadt, Germany, the Chan Zuckerberg Initiative, the HHMI International Scholar award, the European Research Council Consolidator grant (ERC-COG) 724471- HemTree2.0, an SCA award of the Wolfson Foundation and Family Charitable Trust, an MRA Established Investigator Award (509044), the Helen and Martin Kimmel award for innovative investigation, the NeuroMac DFG/Transregional Collaborative Research Center grant, a Cancer Research Institute Technology Impact Award (CRI Award CRI3250), an International Progressive MS Alliance/NMSS PA-1604 08459 and an Adelis Foundation grant. S.A.Q. is funded by a Cancer Research UK (CRUK) Senior Cancer Research Fellowship (C36463/A22246) and a CRUK Biotherapeutic Program grant (C36463/A20764). K.S.P. received funding from the NIHRBTRU for Stem Cells and Immunotherapies (167097). C.C. is funded by Becas Chile. Part of this work was undertaken at UCL with support from the CRUK-UCL Centre (C416/A18088) and the Cancer Immuno-therapy Accelerator Award (CITA-CRUK) (C33499/A20265).

Author information

Authors and Affiliations

Authors

Contributions

I.Y., T.L., A.Y., S.A.Q. and I.A. conceived the project, designed the experiments, analyzed the data and wrote the manuscript. I.Y., T.L., A.Y., I.S., C.C. and D.F.D. performed the experiments, E.D. contributed to data analysis, M.S., C.B., O.B. and I.M. contributed experimentally and K.S.P., S.A.Q. and I.A. supervised the project.

Corresponding authors

Correspondence to Sergio A. Quezada or Ido Amit.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Genevieve Boland, Falk Nimmerjahn and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Flow cytometry gating strategy and scRNAseq data pre-processing steps.

(a) Gating strategy used to sort CD45+ cells or TCRb+ (T) cells. (b) Violin plot depicting the distribution of detected genes per cell per sample. (c) Violin plot depicting the distribution of unique molecular identifiers (UMIs) per cell per sample. (d) Violin plot depicting the distribution of percentage of UMIs coming from mitochondrial genes per cell per sample. (e) Explained variance (Y) as function of principal components (PCs, X) for PCA of normalized expression values of all cells and 3,000 most variable genes (Methods). UMAP of all cells before (left) and after (right) batch correction by CCA (Methods).

Extended Data Fig. 2 Characterization of immune dynamics in the TME following anti-CTLA-4 treatments.

(a) Principle component analysis (PCA) of the relative abundance of each cell type in each sample, or the mean gene expression of genes with UMI > 2 in cells > 5 in each sample. Performed separately on lymphoid and myeloid cells. Pointes are samples color coded for treatment arm. Dashed line separates control and anti-CTLA-4 m1 from anti-CTLA-4 m2a. (b) Comparison of the fractions of the indicated lymphoid cell types in individual mice in each treatment group. n = 3, 2 and 3 biologically independent samples for Ctrl, m1 and m2a groups respectively were used for statistical analysis. (c) Comparison of the fractions of the indicated myeloid cell types in individual mice in each treatment group. n = 5, 3 and 5 biologically independent samples for Ctrl, m1 and m2a groups respectively were used for statistical analysis. Bars represent mean ± SE of mice per treatment arm. Two-tailed Student’s t test was used.

Extended Data Fig. 3 Anti-CTLA-4 m2a treatment drives rapid and broad rearrangement of the adaptive and innate immune TME.

(a) UMAP of scRNA-seq data from day 1 post-treatment tumor infiltrating immune cells. Color code for cell type assignment is indicated in the legend. (b) UMAP projections of relative cell density per treatment, or relative CTLA-4 expression. (c) Comparison of the fractions of the indicated lymphoid cell types in individual mice in each treatment group. n = 3, 6 and 7 biologically independent samples for Ctrl, m1 and m2a groups respectively were used for statistical analysis. (d) Comparison of the fractions of the indicated myeloid cell types in individual mice in each treatment group. n = 2, 6 and 7 biologically independent samples for Ctrl, m1 and m2a groups respectively were used for statistical analysis. (e) Consensus hierarchical clustering (n clusters = 2, Methods) of cellular makeup across samples with Spearman correlation as distance metric. Color coded for P co-clustering after 500 iterations. (f) Histogram of CD4+ draining lymph node-derived cells frequency per FOXP3 protein expression, as measured by flow cytometry. n = 5 per group. Average percentage to the right of the dashed line is stated. Day 4 post-treatment. (g) As in (f) for day 1 post-treatment. (h) Growth curves of MCA-205 tumors in mice untreated, or treated with 50 µg/mouse anti-CTLA-4 m2a at days 6, 9, 12, with or without 100 µg/mouse anti-Gr1 at days 5 and 13. n = 5 mice per arm. In (c) and (d), bars represent mean ± SE of mice per treatment arm. Two-way ANOVA (fraction ~ treatment * batch) was used.

Extended Data Fig. 4 Molecular dissection of the immediate response to anti-CTLA-4 mAbs.

(a) 12 leading DEGs for myeloid cell populations. Log2 fold-change is relative to all other cells. (b) Number of DEGs per metacluster in anti-CTLA-4 m1 vs. control. (c) Scatterplot comparing anti-CTLA-4 m2a (Y) vs. Control (X) gene expression changes in monocytes. Leading DEGs are annotated. (d) Scatterplot comparing anti-CTLA-4 m2a (Y) vs. Control (X) gene expression changes in TAMs. Leading DEGs are annotated. (e) Scatterplot comparing anti-CTLA-4 m2a (Y) vs. Control (X) gene expression changes in monocytes and TAMs combined. Leading DEGs are annotated. Differential gene expression is estimated using two-sided Wilcoxon rank-sum test, Benjamini-Hochberg adjusted p-value < 0.05, FC > 1.25.

Source data

Extended Data Fig. 5 FcγR dependent activation underlies myeloid reprograming following anti-CTLA-4 m2a treatment.

(a) Enrichment of lymphoid cell type frequencies in DT vs control in Foxp3DTR background, or anti-CTLA-4 m2a versus control in WT background. Values are log2 fold change of the mean frequency of each cell type across mice, dot size indicates the average frequency of the cell population between treatments, and color the p-value of two-tailed Student’s t test between each treatment and control. (b) Comparison of the fractions of the indicated lymphoid cell types in individual mice in each treatment group. n = 4 and 5 biologically independent samples for Ctrl and DT groups respectively were used for statistical analysis. (c) As in (a), but for myeloid cells. (d) As in (b), but for myeloid cells. n = 4 and 5 biologically independent samples for Ctrl and DT groups respectively were used for statistical analysis. (e) Enrichment of lymphoid cell type frequencies in anti-CTLA-4 m2a versus anti-CTLA-4 m1 in FCGR KO background, or anti-CTLA-4 m2a versus anti-CTLA-4 m1 in WT background. Values are log2 fold change of the mean frequency of each cell type across mice, dot size indicates the average frequency of the cell population between treatments, and color the p-value of two-tailed Student’s t test between each treatment and control. (f) Comparison of the fractions of the indicated lymphoid cell types in individual mice in each treatment group. n = 7 and 4 biologically independent samples for m1 and m2a groups respectively were used for statistical analysis. (g) As in (e), but for myeloid cells. (h) As in (f), but for myeloid cells. n = 6 and 6 biologically independent samples for m1 and m2a groups respectively were used for statistical analysis. In (b), (d), (f), and (h), Bars represent mean ± SE of mice per treatment arm. Two-tailed Student’s t test was used.

Source data

Extended Data Fig. 6 FcγR engagement drives a robust activation of type-I IFN signaling.

(a) Comparison of the fractions of the indicated cell populations in individual mice in each genotype and treatment group. n = 6, 4, and 3 biologically independent samples for each of Ctrl, m1 and m2a in WT, FCGR KO and IFNAR KO groups respectively, were used for statistical analysis. (b) Scatterplot comparing anti-CTLA-4 m2a WT (Y) vs. anti-CTLA-4 m2a FCGR KO (X) gene expression changes in BMDM cells. Leading DEGs are annotated. (c) Scatterplot comparing anti-CTLA-4 m2a WT (Y) vs. anti-CTLA-4 m2a IFNAR KO (X) gene expression changes in BMDM cells. Leading DEGs are annotated. (d) Scatterplot comparing anti-CTLA-4 m2a FCGR KO (Y) vs. anti-CTLA-4 m2a IFNAR KO (X) gene expression changes in BMDM cells. Leading DEGs are annotated. (e) Percent of proliferating T cells and interferon gamma secretion in idle T cells, activated T cells, and activated T cells co-cultured with Cd11b+ cells purified from MCA-205 tumors treated with anti-CTLA-4 m2a in WT or IFNAR KO mice. n = 4 biologically independent samples for each group were used for statistical analysis. In (a), bars represent mean ± SE of mice per treatment arm. Two-way ANOVA (fraction ~ treatment * batch) was used. Differential gene expression is estimated using two-sided Wilcoxon rank-sum test, Benjamini-Hochberg adjusted p-value < 0.05, FC > 1.25.

Source data

Extended Data Fig. 7 melanoma patients data provides mechanistic link between anti-tumor activity of anti-CTLA-4 treatment in mice and humans.

(ad) Gene set enrichment analysis (GSEA) for responder vs non-responder patients. Green line shows running enrichment score for indicated gene ontology (GO) terms. All gene log2 fold-changes are ranked (bottom, grey) and genes belonging to the gene set are shown in black (top).

Supplementary information

Reporting Summary

Supplementary Table 1

Supplementary Tables 1–6.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yofe, I., Landsberger, T., Yalin, A. et al. Anti-CTLA-4 antibodies drive myeloid activation and reprogram the tumor microenvironment through FcγR engagement and type I interferon signaling. Nat Cancer 3, 1336–1350 (2022). https://doi.org/10.1038/s43018-022-00447-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-022-00447-1

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer