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:

Obesity induces PD-1 on macrophages to suppress anti-tumour immunity

An Author Correction to this article was published on 09 July 2024

This article has been updated

Abstract

Obesity is a leading risk factor for progression and metastasis of many cancers1,2, yet can in some cases enhance survival3,4,5 and responses to immune checkpoint blockade therapies, including anti-PD-1, which targets PD-1 (encoded by PDCD1), an inhibitory receptor expressed on immune cells6,7,8. Although obesity promotes chronic inflammation, the role of the immune system in the obesity–cancer connection and immunotherapy remains unclear. It has been shown that in addition to T cells, macrophages can express PD-19,10,11,12. Here we found that obesity selectively induced PD-1 expression on tumour-associated macrophages (TAMs). Type I inflammatory cytokines and molecules linked to obesity, including interferon-γ, tumour necrosis factor, leptin, insulin and palmitate, induced macrophage PD-1 expression in an mTORC1- and glycolysis-dependent manner. PD-1 then provided negative feedback to TAMs that suppressed glycolysis, phagocytosis and T cell stimulatory potential. Conversely, PD-1 blockade increased the level of macrophage glycolysis, which was essential for PD-1 inhibition to augment TAM expression of CD86 and major histocompatibility complex I and II molecules and ability to activate T cells. Myeloid-specific PD-1 deficiency slowed tumour growth, enhanced TAM glycolysis and antigen-presentation capability, and led to increased CD8+ T cell activity with a reduced level of markers of exhaustion. These findings show that obesity-associated metabolic signalling and inflammatory cues cause TAMs to induce PD-1 expression, which then drives a TAM-specific feedback mechanism that impairs tumour immune surveillance. This may contribute to increased cancer risk yet improved response to PD-1 immunotherapy in obesity.

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: Obesity impairs anti-tumour immune cell function and metabolic signature.
Fig. 2: Obesity and obesity-associated signalling drive macrophage-specific PD-1.
Fig. 3: PD-1-mediated macrophage dysfunction is dependent on mTOR and MYC activation.
Fig. 4: Anti-PD-1 directly rescues macrophage anti-tumour function and metabolism.
Fig. 5: Targeting macrophage-specific PD-1 improves T cell-mediated anti-tumour immunity.

Similar content being viewed by others

Data availability

Materials are available upon request from the corresponding author with an appropriate material transfer agreement. scRNA-seq data are available at the Gene Expression Omnibus under accession number GSE179936. RNA-seq data are publicly available at the Gene Expression Omnibus under accession number GSE242839Source data are provided with this paper.

Change history

References

  1. Islami, F., Goding Sauer, A., Gapstur, S. M. & Jemal, A. Proportion of cancer cases attributable to excess body weight by US state, 2011–2015. JAMA Oncol. 5, 384–392 (2019).

    Article  PubMed  Google Scholar 

  2. Sung, H. et al. Global patterns in excess body weight and the associated cancer burden. CA Cancer J. Clin. 69, 88–112 (2019).

    Article  PubMed  Google Scholar 

  3. Choi, Y. et al. Body mass index and survival in patients with renal cell carcinoma: a clinical-based cohort and meta-analysis. Int. J. Cancer 132, 625–634 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Hakimi, A. A. et al. An epidemiologic and genomic investigation into the obesity paradox in renal cell carcinoma. J. Natl Cancer Inst. 105, 1862–1870 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schlesinger, S. et al. Postdiagnosis body mass index and risk of mortality in colorectal cancer survivors: a prospective study and meta-analysis. Cancer Causes Control 25, 1407–1418 (2014).

    Article  PubMed  Google Scholar 

  6. Wang, Z. et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat. Med. 25, 141–151 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Cortellini, A. et al. A multicenter study of body mass index in cancer patients treated with anti-PD-1/PD-L1 immune checkpoint inhibitors: when overweight becomes favorable. J. Immunother. Cancer 7, 57 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Assumpcao, J. A. F., Pasquarelli-do-Nascimento, G., Duarte, M. S. V., Bonamino, M. H. & Magalhaes, K. G. The ambiguous role of obesity in oncology by promoting cancer but boosting antitumor immunotherapy. J. Biomed. Sci. 29, 12 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gordon, S. R. et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 545, 495–499 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Huang, X. et al. PD-1 expression by macrophages plays a pathologic role in altering microbial clearance and the innate inflammatory response to sepsis. Proc. Natl Acad. Sci. USA 106, 6303–6308 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Strauss, L. et al. Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci. Immunol. 5, eaay1863 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yao, A. et al. Programmed death 1 deficiency induces the polarization of macrophages/microglia to the M1 phenotype after spinal cord injury in mice. Neurotherapeutics 11, 636–650 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Avgerinos, K. I., Spyrou, N., Mantzoros, C. S. & Dalamaga, M. Obesity and cancer risk: emerging biological mechanisms and perspectives. Metab. Clin. Exp. 92, 121–135 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Calle, E. E., Rodriguez, C., Walker-Thurmond, K. & Thun, M. J. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N. Engl. J. Med. 348, 1625–1638 (2003).

  15. Dai, Z., Xu, Y. C. & Niu, L. Obesity and colorectal cancer risk: a meta-analysis of cohort studies. World J. Gastroenterol. 13, 4199–4206 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Murphy, W. J. & Longo, D. L. The surprisingly positive association between obesity and cancer immunotherapy efficacy. JAMA 321, 1247–1248 (2019).

    Article  PubMed  Google Scholar 

  17. Donnelly, D. et al. The complex relationship between body mass index and response to immune checkpoint inhibition in metastatic melanoma patients. J. Immunother. Cancer 7, 222 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Naik, G. S. et al. Complex inter-relationship of body mass index, gender and serum creatinine on survival: exploring the obesity paradox in melanoma patients treated with checkpoint inhibition. J. Immunother. Cancer 7, 89 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Sanchez, A. et al. Transcriptomic signatures related to the obesity paradox in patients with clear cell renal cell carcinoma: a cohort study. Lancet Oncol. 21, 283–293 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Woodall, M. J., Neumann, S., Campbell, K., Pattison, S. T. & Young, S. L. The effects of obesity on anti-cancer immunity and cancer immunotherapy. Cancers 12, 1230 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Boi, S. K. et al. Obesity diminishes response to PD-1-based immunotherapies in renal cancer. J Immunother. Cancer 8, e000725 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Lennon, H., Sperrin, M., Badrick, E. & Renehan, A. G. The obesity paradox in cancer: a review. Curr. Oncol. Rep. 18, 56 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Le, C. T. et al. PD-1 blockade reverses obesity-mediated T cell priming impairment. Front. Immunol. 11, 590568 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ringel, A. E. et al. Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell 183, 1848–1866 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chakarov, S., Bleriot, C. & Ginhoux, F. Role of adipose tissue macrophages in obesity-related disorders. J. Exp. Med. 219, e20211948 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hamada, K. et al. Obesity strongly promotes growth of mouse MC38 colon cancer in an orthotopic-syngeneic C57BL/6 mouse model. In Vivo 36, 1643–1646 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Wagner, A. et al. Metabolic modeling of single Th17 cells reveals regulators of autoimmunity. Cell 184, 4168–4185 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Christofides, A. et al. SHP-2 and PD-1-SHP-2 signaling regulate myeloid cell differentiation and antitumor responses. Nat. Immunol. 24, 55–68 (2023).

    Article  CAS  PubMed  Google Scholar 

  29. Bally, A. P. et al. NF-kappaB regulates PD-1 expression in macrophages. J. Immunol. 194, 4545–4554 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Obradovic, A. et al. Single-cell protein activity analysis identifies recurrence-associated renal tumor macrophages. Cell 184, 2988–3005 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pelka, K. et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell 184, 4734–4752 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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 

  33. Wolf, E. M., Fingleton, B. & Hasty, A. H. The therapeutic potential of TREM2 in cancer. Front. Oncol. 12, 984193 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, W. et al. Correlation between PD-1/PD-L1 expression and polarization in tumor-associated macrophages: a key player in tumor immunotherapy. Cytokine Growth Factor Rev. 67, 49–57 (2022).

    Article  CAS  PubMed  Google Scholar 

  35. Schmidt, F. M. et al. Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS ONE 10, e0121971 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Kumar, R. et al. Association of leptin with obesity and insulin resistance. Cureus 12, e12178 (2020).

    PubMed  PubMed Central  Google Scholar 

  37. Zhao, L. et al. A panel of free fatty acid ratios to predict the development of metabolic abnormalities in healthy obese individuals. Sci. Rep. 6, 28418 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kono, Y. et al. Increased PD-1-positive macrophages in the tissue of gastric cancer are closely associated with poor prognosis in gastric cancer patients. BMC Cancer 20, 175 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Freemerman, A. J. et al. Myeloid Slc2a1-deficient murine model revealed macrophage activation and metabolic phenotype are fueled by GLUT1. J. Immunol. 202, 1265–1286 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang, W. et al. Diabetic hyperglycemia promotes primary tumor progression through glycation-induced tumor extracellular matrix stiffening. Sci. Adv. 8, eabo1673 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Dudzinski, S. O. et al. Leptin augments antitumor immunity in obesity by repolarizing tumor-associated macrophages. J. Immunol. 207, 3122–3130 (2021).

    Article  CAS  PubMed  Google Scholar 

  42. Reinfeld, B. I. et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 593, 282–288 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sorrelle, N. et al. Improved multiplex immunohistochemistry for immune microenvironment evaluation of mouse formalin-fixed, paraffin-embedded tissues. J. Immunol. 202, 292–299 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Zhang, L. et al. Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell 181, 442–459 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Rappold, P. M. et al. A targetable myeloid inflammatory state governs disease recurrence in clear-cell renal cell carcinoma. Cancer Discov. 12, 2308–2329 (2022).

Download references

Acknowledgements

We thank members of the laboratories of J.C.R. and W.K.R. for input; the laboratories of J. Balko, Y. Kim and P. Hurley for the use of their tumour dissociators; the Doran Lab (Vanderbilt University Medical Center) for providing the RAW264.7 cell line and the laboratory of B. Stanger (University of Pennsylvania) for providing the 6419c5 pancreatic cell line. We also thank A. Hakimi for his gift of LVRCC67 cells47. This work was supported by National Institutes of Health (NIH) grants K00 CA234920 (J.E.B.), F31 CA261049 (M.M.W.), F30 CA239367 (M.Z.M.), F30 CA247202 (B.I.R.), K00 CA253718 (E.N.A), F99 CA274695 (W.W.), T32 DK007314 (L.V.P.), R01 CA217987 (J.C.R.), K08 CA241351 (S.M.H.), R01CA238263 (V.A.B.), R01 DK105550 (J.C.R.), T32 GM007347 (M.Z.M. and B.I.R.), K12 CA090625 (K.E.B. and W.K.R.) and T32 DK101003 (K.S.), the American Association for Cancer Research (B.I.R. and W.K.R.), the Vanderbilt-Incyte Alliance (J.C.R. and W.K.R.), the Mark Foundation for Cancer Research (J.C.R. and A.H.H.) and pilot funding from the Vanderbilt Ingram Cancer Center (P30 CA068485; J.C.R.). The Vanderbilt VANTAGE Core, including A. Jones, provided technical assistance for this work. VANTAGE is supported in part by a Clinical and Translational Science Award grant (5UL1 RR024975-03), the Vanderbilt Ingram Cancer Center (P30 CA068485), the Vanderbilt Vision Center (P30 EY08126) and the NIH’s National Center for Research Resources (G20 RR030956). Flow-sorting experiments were carried out in the VUMC Flow Cytometry Shared Resource by D. K. Flaherty and B. K. Matlock and were supported by the Vanderbilt Ingram Cancer Center (P30 CA068485) and the Vanderbilt Digestive Disease Research Center (P30 DK058404). We acknowledge the Translational Pathology Shared Resource supported by NIH National Cancer Institute Cancer Center Support Grant 5P30 CA68485-19 and the Vanderbilt Mouse Metabolic Phenotyping Center Grant 2 U24 DK059637-16. The images of the needles, the tumour and the MACS set-up in Fig. 1a were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

J.E.B. and J.C.R. conceived and designed the study and composed the manuscript. K.E.B. provided clinical expertise and human ccRCC samples for flow cytometry analysis. M.D.L. and Z.H. assisted with collecting and processing of human patient samples. J.E.B., M.M.W., G.L.L.-T., B.I.R. and M.Z.M carried out DIO and tumour experiments and assisted with downstream analysis including flow cytometry. G.A.N., M.A.C. and X.Y. analysed scRNA-seq dataset. E.N.A. provided expertise in staining of tumour microarray samples that were collected by S.M.H. E.S.H. and K.K.S. assisted with experimental studies and editing the manuscript. A.C. and V.A.B. provided transgenic LysMCre Pdcd1fl/fl breeding pairs. E.E.F., A.B. and C.H.S. provided the immortalized BMDM cell line, the Pten-knockout BMDM cell line and processed the human blood monocytes from healthy donors and the patient with PTEN deficiency. L.V.P. and K.E.W. provided pancreatic tumour model expertise and the 6419c5 cell line and assisted with editing the manuscript. E.M.W. and A.H.H. provided the orthotopic breast cancer model and the EO771 cell line and assisted with editing the manuscript. M.E.B., K.S., W.W. and C.A.R.-K. carried out the experiments with the PyMT SZT mouse tumour model. S.M.H. provided clinical expertise and provided tumour microarray samples. D.S., C.A. and C.-H.L. provided endometrial biopsy samples. H.C. provided guidance on appropriate statistical analyses. J.C.R. and W.K.R. obtained funding for this study.

Corresponding author

Correspondence to Jeffrey C. Rathmell.

Ethics declarations

Competing interests

J.C.R. has held stock equity in Sitryx and in the past two years has received unrelated research support, travel and honoraria from Sitryx, Caribou, Nirogy, Kadmon, Calithera, Tempest, Merck, Mitobridge and Pfizer. In the past two years, W.K.R. has received unrelated clinical research support from Bristol-Meyers Squib, Merck, Pfizer, Peloton, Calithera and Incyte. Both J.C.R. and W.K.R. are divested of these interests at present. K.E.B. received funding to the institution for preclinical research from Bristol-Myers Squibb–International Association for the Study of Lung Cancer–Lung Cancer Foundation of America, funding to the institution for clinical trials from Arrowhead, Aravive, Aveo, BMS, Exelexis and Merck, and consulting fees from Alpine Immune Sciences, Aravive, AstraZeneca, Aveo, BMS, Exelexis, Merck, Seagen and Sanofi. V.A.B. has patents on the PD-1 pathway licensed by Bristol-Myers Squibb, Roche, Merck, EMD-Serono, Boehringer Ingelheim, AstraZeneca, Novartis and Dako.

Peer review

Peer review information

Nature thanks Christopher Garris 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 figures and tables

Extended Data Fig. 1 High Fat Diet feeding induces an obese phenotype.

a, Mouse body weights during 20 weeks of LFD or HFD feeding prior to MC38 tumor injection (n = 20 mice per diet). b, Mouse food intake during 20-week diet treatment. Food weighed weekly per cage and intake calculated as food weight difference between weeks (difference in food weight(g)/7 days/5 mice per cage X kcal of diet) (45% HFD = 4.73 kcal/g; LFD = 3.85 kcal/g) (n = 4 cages with 5 mice per cage). c-d, Blood glucose and blood insulin levels measured via tail bleed following a 4-hour fast after 20 weeks of diet treatment (n = 20 mice per diet). e-h, Representative tumor growth curves of mice inoculated with 5 × 105 MC38 cells following 20 weeks of diet treatment (n = 6-8 mice per group). i, Tumor weights on day 19 following subcutaneous inoculation. “X” indicates days of anti-PD-1 or IgG injection. Data points identified as “+” were pooled together at equal amounts of live cells for subsequent single cell RNA sequencing analysis. j, Percent change in tumor growth in response to a-PD-1 treatment within respective diets ((Tumor Diameter Day 9 LFD α-PD-1 – Diameter Day 19 LFD IgG) ÷ Diameter Day 19 LFD IgG × 100). P values were calculated using an unpaired two-tailed t-test (a-d, j) or two-way ANOVA using a Fishers LSD test for multiple comparisons (i). ns=p > 0.05; * p ≤ 0.05; ** p ≤ 0.01.

Source Data

Extended Data Fig. 2 Cell characterization of CD45+ and CD45- cells from MC38 tumors.

a-b, Cell counts of SingleR annotated clusters from scRNAseq of MACS enriched CD45- and CD45+ MC38 tumor samples. c, Heat map of identifying genes used to annotate CD45+ clusters. d, Representative gating strategy for quantifying T cell and macrophage cell populations within the spleen and tumor of MC38 tumor bearing mice. e-g, Quantification of tumor CD8+, CD4+ and TAMs, respectively, gated on live singlets in MC38 tumors. h-j, Quantification of percent positive and MFI of tumor PD-1+, Lag3+ and Tim3+ CD8 T cells, CD4 T cells and TAMs. k, Frequency of Pdcd1+ cells in TAM clusters of scRNAseq dataset. Data is representative of one experiment, n = 3 pooled tumors from each treatment group (a-b,k). Each data point represents a biological replicate; data are mean ± s.e.m. P values were calculated using a Two-way ANOVA with a Fishers LSD test for multiple comparisons. ns=p > 0.05; * p ≤ 0.05.

Source Data

Extended Data Fig. 3 HFD reduced clonality of CD8 T cells and impairs TAM function and metabolic signature with MC38 tumors.

a, UMAP of clone abundance within each treatment group from V(D)J Sequencing of T cell clusters with productive TCRs from scRNAseq dataset. b, Absolute number of T cells with paired TCR-a and TCR-b sequences. Data is representative of one experiment, n = 3 pooled tumors from each treatment group. c, Representative gating strategy for TAMs from MC38 tumors, identified as live singlets CD45+, CD8- CD11b+ and F480+. d, MFI of phrodo+ TAMs. e, MFI of MHCII in TAMs. f, Percent positive and MFI of CD80 in TAMs. MFI values were normalized within trials to IgG controls. g-i, Measurement of basal OCR, basal ECAR and OCR:ECAR ratio of MACS enriched CD11b+ cells sorted from MC38 tumors during a MitoStress Test measured using Agilent Seahorse extracellular flux analyzer. Each data point represents a biological replicate; For d-f MFI was normalized to the average of LFD IgG control within each independent trial and combined from 2 independent experiments. Data are mean ± s.e.m. P values were calculated using a Two-way ANOVA with a Fishers LSD test for multiple comparisons. * p ≤ 0.05.

Source Data

Extended Data Fig. 4 Pdcd1 is increased in TAMs in many types of cancers.

a, Pdcd1 expression within tumor immune cells subsets from scRNAseq within TISCH2 database. b-c, Pdcd1 expression in CD8 T effector and Monocyte/Macrophage identified cells in the TISCH2 scRNAseq database from various cancer datasets. d, Percent Pdcd1+ macrophages from human ccRCC tumor samples measured using gene expression from scRNAseq dataset (n = 11 patients) e, Percent Pdcd1+ macrophages of human CRC tumor samples quantified using gene expression from scRNAseq (n = 62 patients) f-g, Percent Pdcd1+ macrophage cells corresponding to pathological tumor grade compared to adjacent normal tissue from human CRC or human ccRCC tumor samples measured using gene expression from scRNAseq. h, Enrichment of KEGG metabolic signature scores of Pdcd1 versus Pdcd1+ TAMs from human CRC scRNAseq. Data are mean ± s.e.m. P values were calculated using an unpaired two-tailed t-test. *p ≤ 0.05, **p ≤ 0.01.

Source Data

Extended Data Fig. 5 Tumor Associated Macrophages express PD-1 in human tumor samples which is not directly associated with TREM2 expression.

a, Representative histogram and contour plot illustrating PD-1 expression within TAMs or CD8 T cells from human ccRCC patients. b, Frequency of live singlets CD45+ CD11b+ CD68+ and CD163+ TAMs and CD45+ CD8+ T cells from ccRCC patient PBMCs or tumor. c, MFI of PD-1 expression in TAMs and CD8 T cells from ccRCC patient samples. d, Representative flow gating strategy of MACS sorted CD11b+ and CD8+ cells from freshly isolated human ccRCC tumors or matched PBMCs. e, Histogram representing PD-1, TIM3 and LAG3 expression on human peripheral blood mononuclear cells (PBMCs) and matched ccRCC tumor in MACS sorted CD11b+ TAMs and CD8+ TILs. f, Representative image of Tumor Microarray (TMA) staining identifying PD-1 colocalizing with CD163+ and CD8+ cells at 40X magnification. Scale bar 50 mm. g, Frequency of PD-1+ TAMs or PD-1+ CD8 T cells from TMA staining with patient clinical data categorized by BMI status (lean BMI < 25; obese BMI 30-35) (n = 28 lean patients; n = 53 obese). h, Area of CD163+ or CD8+ staining taken from endometrial carcinoma tumor tissues obtained from 5 patients before and after at least a 10% body weight loss. i, Representative PD-1 and TREM2 expression from live CD11b+ CD163+ and CD68+ TAMs from human ccRCC tumor samples. j, Percent positive of PD-1+ cells in tumor immune cells from MC38 subcutaneous murine tumors. k, MFI of PD-1 expression within different immune cell subsets with representative overlay of histogram and contour plots with FMO control. Each data point represents individual patient samples. Data are mean ± s.e.m. P values were calculated using an unpaired two-tailed t-test (d,e,i) or a paired two tailed t-test (h). P values were calculated using a one way ANOVA compared to CD3- CD11b- cells (j-k).ns p > 0.05, *p ≤ 0.05, ****p ≤ 0.001.

Source Data

Extended Data Fig. 6 Obesity and metabolic dysfunction induce PD-1 expression on TAMs.

a, Tumor diameter over time of pancreatic tumor cells, 6419c5, injected 2 × 105 subcutaneously into mice fed a LFD or 45% HFD diet for 20–25 week. b, Tumor weight at Day17 post injection of pancreatic tumor cell line. c-d, Percent positive and MFI of PD-1 of TAMs and CD8 T cells respectively from pancreatic tumors. e-g, Percent positive and MFI of MHCII, CD80 and PDL1 of TAMs from pancreatic tumors. h, Percent of phrodo positive TAMs from pancreatic tumors. i) MFI of Bodipy C:16 uptake in TAMs from pancreatic tumors. j, Tumor diameter over time of subcutaneous kidney tumor cells, LVRCC, injected 2×106 subcutaneously into mice fed a LFD or 45% HFD diet for 20–25 week. k, Tumor weight at Day 28 post injection of kidney tumor cell line. l-m, Percent positive and MFI of PD-1 of TAMs and CD8 T cells respectively from kidney tumors. n-o, Percent positive PD-1 of TAMs and CD8 T cells respectively from 3 × 105 PD-L1 expressing EO771 cells injected in 4th mammary pad of female C57BL6/J mice ovariectomized at 6 weeks of age following 25 weeks of LFD or 60% HFD treatment. p, Tumor weight of FVB/MMTV-PyMT mice injected with or without streptozotocin (STZ) at 70 mg/kg and fed a 60% HFD for 12 weeks. q, Percent positive of PD-1 of TAMs from PYMT tumors. Each data point represents biological replicates. In n-o data were combined from 4 independent trials. In p-q data were combined from 4 independent trials and normalized to control within each trial. Individual tumors from PyMT mice with multiple tumors were processed separately. Dots of the same color indicate individual tumors collected from the same mouse. Data are mean ± s.e.m. P values were calculated using an unpaired two-tailed t-test. ns p > 0.05, *p ≤ 0.05, **p ≤ 0.01.

Source Data

Extended Data Fig. 7 Obesity induced PD-1 expression on TAMs independent of T cell presence.

a, Percent of CD11b+ F480+ macrophages within MC38 tumors and spleens following 2 weeks of LFD or 45% HFD treatment (acute diet). b, Percent of PD-1+ macrophages in tumor and spleen following 2-week acute diet treatment. c-e, Relative abundance of indicated immune cell populations in MC38 tumor (c) and spleen (d) with diet and αCD4/8 treatments (n = 3 mice/group) with representative flow cytometry plots (e) demonstrating T cell depletion. f-i, Tumor weights, PD-1+ expression, % phrodo+ cells, and MHCII expression on CD11b+/F480+ TAMs on day 14 of MC38 tumors following αCD4 and αCD8 T cell depletion in mice fed LFD or HFD for 20 weeks (n = 3-4 mice). j, Frequency of PD-1+ macrophages from spleen, epididymal adipose tissue, or tumors of s.c. pancreatic tumor-bearing mice following 20 weeks of diet treatment. k, Frequency of PD-1+ BMDMs following 4-, 8-, 24- and 48- hours exposure to IFNg (M1-like;Pro-inflammatory), IL-4 (M2-like; Anti-inflammatory) or tumor-conditioned media (TCM). l, MFI of CD274 (PD-L1) of BMDMs following 4-, 8-, 24- and 48-hour exposure to (M1), (M2) or (TCM) conditions. m, MFI of PD-1 of BMDMs following 24-hour exposure to LPS, IFNg, IL-4, TCM, IL-6, MCP-1, IL-1b, TNF-a, leptin, or insulin with representative Isotype and FMO control. n, MFI expression of PD-L1 and representative histograms of BMDMs following 24-hour stimulation. Data points indicate biological replicates. Data in E is one sample of 3-5 pooled spleens per treatment group. Data are mean ± s.e.m. P values were calculated using a Two-Way ANOVA followed by a Fishers LSD test for multiple comparisons (f-i) or an unpaired two-tailed t-test (j). P values were calculated using a one-way ANOVA compared to untreated M0 BMDMs (k-n) *p ≤ 0.05). *p ≤ 0.05). Data points indicate biological replicates (n = 3). Data are mean ± s.e.m.

Source Data

Extended Data Fig. 8 Pdcd1+ and PD-1 + TAMs have a unique gene signature.

a, GO Biological Process Pathway Analysis of differential gene expression from Pdcd1 versus Pdcd1+ TAMs from MC38 scRNAseq TAM clusters independent of diet or treatment. b, Differential Gene expression of select genes from Pdcd1 versus Pdcd1+ TAMs from MC38 scRNAseq (See Supplementary Table 1 for full list of significant genes). c, Quantification of Pdcd1 CPM expression from FACS sorted PD-1+ and PD-1low/negative populations. d, Bulk RNA sequencing of Hallmark Pathway analysis of DEGs from PD-1low/negative versus PD-1+ TAMs via FACS. Data points or squares represent biological replicates as mean ± s.e.m (n = 10). P values were calculated using a paired two-tailed t-test. ***p ≤ 0.001.

Source Data

Extended Data Fig. 9 PD-1 expression in macrophages is dependent on PI3K, mTOR, NF-κB and c-Myc activation.

a, Percent viability, MFI of PD-1 and percent PD-1+ of primary BMDMs stimulated with LPS (1ug/mL) and treated with c-Myc inhibitors (KJ Pyr 9 10ug/mL and 10058-F4 10ug/mL), mTOR inhibitor (Rapamycin 250 ng/mL), AMPK agonist (AICAR 2ug/mL) and NF-κB inhibitor (BMS-345541 1.2ug/mL) for 24 h. b, Percent viability, MFI of PD-1 and percent PD-1+ of immortalized cas9 BMDMs treated with c-Myc inhibitors (KJ Pyr 9 10ug/mL and 10058-F4 10ug/mL), mTOR inhibitor (Rapamycin 350 ng/mL), AMPK agonist (AICAR 5ug/mL) and NF-κB inhibitor (BMS-345541 300ug/mL) for 24 h. c, Percent viability, MFI of PD-1 and percent PD-1+ of RAW264.7 cells treated with c-Myc inhibitors (KJ Pyr 9 10ug/mL and 10058-F4 10ug/mL), mTOR inhibitor (Rapamycin 87.5 ng/mL), AMPK agonist (AICAR 52.5 g/mL) and NF-κB inhibitor (BMS-345541 150ug/mL) for 24 h. d, Western blot of PTEN expression of immortalized cas9 BMDMs following crispr mediated deletion of PTEN. Ctrl is the empty vector. All studies used PTEN 2 clone e, Pdcd1 expression of monocytes enriched from PBMCs of healthy donors or a PTEN haploinsufficient patient. Data points in a indicate biological replicates (n = 4). Data points in b-c indicate technical replicates (n = 4) and is representative of >3 independent trials. Data points in e represent 2 independent healthy donors and a PTEN deficient patient. Data are mean ± s.e.m. P values were calculated using a one-way ANOVA (A-C). ns p > 0.05, *p ≤ 0.05, **p ≤ 0.01, ****p ≤ 0.0001.

Source Data

Extended Data Fig. 10 Deletion of Pdcd1 suppresses tumor growth and promotes a pro-inflammatory, glycolytic phenotype in macrophages.

a, Tumor weight on Day 14 following 1×106 MC38 cells injected subcutaneously into WT or Pdcd1−/− age matched mice (n = 8-10mice). b, Percent phrodo+ of peritoneal macrophages from WT or Pdcd1−/ − tumor-exposed mice c, Percent positive and MFI of MHCII of peritoneal macrophages d) Percent positive and MFI of CD80 of peritoneal macrophages e, MFI of 2-NBDG in peritoneal macrophages following 45 min exposure to 2-NDBG. f-g, Tumor diameter over time and tumor weight on Day 14 following 5×106 MC38 cells injected subcutaneously into WT or Pdcd1−/ − age matched mice (n = 3-5mice). h-j, Frequency and MFI of MHCII, CD86 and Glut1 expression on TAMs from WT or Pdcd1−/ − mice. k, OCR and ECAR measurements over time from MitoStress Test of BMDMs isolated from WT or Pdcd1−/ − mice stimulated for 24 h with IFN-g (M1), IL-4 (M2), or 50% Tumor Conditioned Media (TCM). l, ECAR measurement over time from GlycoStress test of BMDMs with corresponding Glycolysis calculation. Data points in f-g represent 6 biological replicates run in triplicate. P values were calculated using an unpaired two-tailed t-test; ns= p > 0.05, *p ≤ 0.05, **p ≤ 0.05).

Source Data

Extended Data Fig. 11 Direct effects of anti-PD-1 treatment on tumor conditioned macrophage function and metabolism.

a, OCR from MitoStress Test of BMDM stimulated with TCM for 24 h followed by treatment with recombinant mouse PD-L1 and/or anti-PD-1 for an additional 24 h. b,c, Percent positive phrodo, and CD80 of TCM stimulated BMDM following rmPD-L1 and/or anti-PD-1 treatment. d-f, Percent positive phrodo, CD80 and MHCII of TAMs from MACS isolated CD11b+ cells from MC38 tumors ex-vivo treated α−PD-1 for 24 h. g, Fold change of MHCII, MHCI, Glut1, and iNOS expression of IgG/a-PD-1 ex-vivo treated TAMs from LFD versus HFD treated MC38 tumor bearing mice. h, Volcano plot of differentially expressed genes from MACS isolated CD11b+ cells from MC38 tumors ex-vivo treated α-PD-1 for 24 h. Data points indicated biological replicates. Data in a-c representative of ≥ three independent experiments with ≥ 3 mice per group run in triplicate. Data are mean ± s.e.m. P values were calculated using a One-way ANOVA (a-c), a paired two-tailed t-test (d-f), or an unpaired two tailed t-test g, *p ≤ 0.05, **p ≤ 0.01).

Source Data

Extended Data Fig. 12 Myeloid specific PD-1 depletion improves TAM anti-tumor function.

a, Frequency of immune cell subsets from MC38 tumors injected into WT and LysMcre Pdcd1fl/fl mice. b, Division index calculated from flow cytometry analysis of CTV staining CD11b+ isolated cells from MC38 tumors from WT vs LysMcre Pdcd1fl/fl mice cocultured with OT1 CD8+ isolated splenocytes. Cocultured cells treated in the presence of 1 mg/ml OVA protein for 5 days. c, Basal and maximal OCR measurements from MitoStress Test of CD11b+ isolated cells from MC38 tumors of WT and LysMcre Pdcd1fl/fl. d, Calculated glycolysis and glycolytic capacity following GlycoStress Test of CD11b+ isolated cells from MC38 tumors of WT and LysMcre Pdcd1fl/fl. e-h, Percent positive and MFI of PD-1, Lag3, TIM3 and CD69 expression on CD8 T cells from MC38 tumors of WT and LysMcre Pdcd1fl/fl. i, Representative gating following serial MACS isolations of Gr-1- cells followed by CD11b+ MACS isolation. j, Purity of GR-1- CD11b+ F480+ cells before and after MACS sorting. k-l, OCR and ECAR measurement over time from a MitoStress Test and ECAR measurement over time from a GlycoStress Test of MACS isolated GR-1- CD11b+ cells from MC38 tumors in WT vs LysMcre Pdcd1fl/fl male mice. m, Division index calculated from flow cytometry analysis of CellTrace violet staining GR-1- CD11b+ isolated cells from MC38 tumors from WT vs LysMcre Pdcd1fl/fl mice cocultured with OT1 CD8+ isolated splenocytes. Cocultured cells treated in the presence of 1 mg/ml OVA protein for 5 days. n, MFI of Granzyme B expression from OT1 CD8+ cells cocultured as indicated in in panel m. Data representative of ≥ three independent experiments with 3-6 mice per group. Data are mean ± s.e.m. P values were calculated using an unpaired two-tailed t-test. (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001).

Source Data

Supplementary information

Supplementary Figures

Supplementary Figs. 1–7.

Reporting Summary

Supplementary Table 1

Information on patients with endometrial cancer. Relative to data presented in Fig. 2a–c and Extended Data Fig. 5h.

Supplementary Table 2

Pathway enrichment in Pdcd1+/- macrophages. Relative to data presented in Extended Data Fig. 8a.

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

Bader, J.E., Wolf, M.M., Lupica-Tondo, G.L. et al. Obesity induces PD-1 on macrophages to suppress anti-tumour immunity. Nature 630, 968–975 (2024). https://doi.org/10.1038/s41586-024-07529-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07529-3

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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