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:

Castration-mediated IL-8 promotes myeloid infiltration and prostate cancer progression

Abstract

Unlike several other tumor types, prostate cancer rarely responds to immune checkpoint blockade (ICB). To define tumor cell intrinsic factors that contribute to prostate cancer progression and resistance to ICB, we analyzed prostate cancer epithelial cells from castration-sensitive and -resistant samples using implanted tumors, cell lines, transgenic models and human tissue. We found that castration resulted in increased expression of interleukin-8 (IL-8) and its probable murine homolog Cxcl15 in prostate epithelial cells. We showed that these chemokines drove subsequent intratumoral infiltration of tumor-promoting polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), which was largely abrogated when IL-8 signaling was blocked genetically or pharmacologically. Targeting IL-8 signaling in combination with ICB delayed the onset of castration resistance and increased the density of polyfunctional CD8 T cells in tumors. Our findings establish a novel mechanism by which castration mediates IL-8 secretion and subsequent PMN-MDSC infiltration, and highlight blockade of the IL-8/CXCR2 axis as a potential therapeutic intervention.

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: ADT regulates Cxcl15 expression in murine prostate cancer cells.
Fig. 2: Androgen receptor signaling modulates IL-8 expression in human prostate cancer cells.
Fig. 3: Cxcl15 and IL-8 are upregulated in post-castration and castration-resistant prostate cancer.
Fig. 4: Castration-mediated IL-8 upregulation promotes PMN-MDSC infiltration.
Fig. 5: The Cxcl15/CXCR2 (IL-8/CXCR2) axis regulates the migration of PMN-MDSCs, but not their ability to suppress T cell function.
Fig. 6: CXCR2 blockade improves response to ICB following ADT.
Fig. 7: The therapeutic effect of the triple combination is associated with PMN-MDSC reduction.

Similar content being viewed by others

Data availability

Microarray data from this study have been deposited in the Gene Expression Omnibus under accession code GSE171491. Previously published microarray and ChIP-seq data that were reanalyzed here are available under accession codes GSE8466, GSE56288, GSE83860 and GSE55064. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Kantoff, P. W. et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 363, 411–422 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Kwon, E. D. et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 15, 700–712 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Beer, T. M. et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J. Clin. Oncol. 35, 40–47 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Topalian, S. L., Drake, C. G. & Pardoll, D. M. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 27, 450–461 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gao, J. et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat. Med. 23, 551–555 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Shen, Y. C. et al. Combining intratumoral Treg depletion with androgen deprivation therapy (ADT): preclinical activity in the Myc-CaP model. Prostate Cancer Prostatic Dis. 21, 113–125 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Obradovic, A. Z. et al. T-cell infiltration and adaptive Treg resistance in response to androgen deprivation with or without vaccination in localized prostate cancer. Clin. Cancer Res. 26, 3182–3192 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lu, X. et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature 543, 728–732 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lopez-Bujanda, Z. & Drake, C. G. Myeloid-derived cells in prostate cancer progression: phenotype and prospective therapies. J. Leukoc. Biol. 102, 393–406 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lopez-Bujanda, Z. A. et al. Robust antigen-specific CD8 T cell tolerance to a model prostate cancer neoantigen. OncoImmunology 9, 1809926 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Calcinotto, A. et al. IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature 559, 363–369 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Watson, P. A. et al. Context-dependent hormone-refractory progression revealed through characterization of a novel murine prostate cancer cell line. Cancer Res. 65, 11565–11571 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Ellwood-Yen, K. et al. Myc-driven murine prostate cancer shares molecular features with human prostate tumors. Cancer Cell 4, 223–238 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Rossi, D. L. et al. Lungkine, a novel CXC chemokine, specifically expressed by lung bronchoepithelial cells. J. Immunol. 162, 5490–5497 (1999).

    CAS  PubMed  Google Scholar 

  16. Schmitz, J. M., McCracken, V. J., Dimmitt, R. A. & Lorenz, R. G. Expression of CXCL15 (lungkine) in murine gastrointestinal, urogenital, and endocrine organs. J. Histochem. Cytochem. 55, 515–524 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Chen, R. et al. Telomerase deficiency causes alveolar stem cell senescence-associated low-grade inflammation in lungs. J. Biol. Chem. 290, 30813–30829 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Elliott, C. L., Allport, V. C., Loudon, J. A., Wu, G. D. & Bennett, P. R. Nuclear factor-kappa B is essential for up-regulation of interleukin-8 expression in human amnion and cervical epithelial cells. Mol. Hum. Reprod. 7, 787–790 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Fiedler, M. A., Wernke-Dollries, K. & Stark, J. M. Inhibition of TNF-α-induced NF-κB activation and IL-8 release in A549 cells with the proteasome inhibitor MG-132. Am. J. Respir. Cell Mol. Biol. 19, 259–268 (1998).

    Article  CAS  PubMed  Google Scholar 

  20. Khanjani, S., Terzidou, V., Johnson, M. R. & Bennett, P. R. NFκB and AP-1 drive human myometrial IL8 expression. Mediators Inflamm. 2012, 504952 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Mostaghel, E. A. et al. Intraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: therapeutic implications for castration-resistant prostate cancer. Cancer Res. 67, 5033–5041 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Antonarakis, E. S. et al. Neoadjuvant randomized trial of degarelix (Deg) ± cyclophosphamide/GVAX (Cy/GVAX) in men with high-risk prostate cancer (PCa) undergoing radical prostatectomy (RP). J. Clin. Oncol. 35, 5077 (2017).

    Article  Google Scholar 

  23. Cioni, B. et al. Loss of androgen receptor signaling in prostate cancer-associated fibroblasts (CAFs) promotes CCL2- and CXCL8-mediated cancer cell migration. Mol. Oncol. 12, 1308–1323 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Aytes, A. et al. ETV4 promotes metastasis in response to activation of PI3-kinase and Ras signaling in a mouse model of advanced prostate cancer. Proc. Natl Acad. Sci. USA 110, E3506–E3515 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Arriaga, J. M. et al. A MYC and RAS co-activation signature in localized prostate cancer drives bone metastasis and castration resistance. Nat. Cancer 1, 1082–1096 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Alshetaiwi, H. et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci. Immunol. https://doi.org/10.1126/sciimmunol.aay6017 (2020).

  27. Antonarakis, E. S. et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J. Clin. Oncol. 38, 395–405 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Schalper, K. A. et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat. Med. 26, 688–692 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yuen, K. C. et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat. Med. 26, 693–698 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kawahara, T. et al. Neutrophil-to-lymphocyte ratio predicts prostatic carcinoma in men undergoing needle biopsy. Oncotarget 6, 32169–32176 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Yin, X. et al. Prognostic role of neutrophil-to-lymphocyte ratio in prostate cancer: a systematic review and meta-analysis. Medicine (Baltimore) 95, e2544 (2016).

    Article  Google Scholar 

  32. Alfaro, C. et al. Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs). Clin. Cancer Res. 22, 3924–3936 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kumar, V. et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell 32, 654–668.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Patnaik, A. et al. Cabozantinib eradicates advanced murine prostate cancer by activating antitumor innate immunity. Cancer Discov. 7, 750–765 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. David, J., Dominguez, C., Hamilton, D. & Palena, C. The IL-8/IL-8R axis: a double agent in tumor immune resistance. Vaccines (Basel) 4, 22 (2016).

    Article  CAS  Google Scholar 

  37. Culig, Z. et al. Switch from antagonist to agonist of the androgen receptor bicalutamide is associated with prostate tumour progression in a new model system. Br. J. Cancer 81, 242–251 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rao, V. et al. A Hoxb13-driven reverse tetracycline transactivator system for conditional gene expression in the prostate. Prostate 72, 1045–1051 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wang, X. et al. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature 461, 495–500 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lopez-Bujanda, Z. A. et al. TGM4: an immunogenic prostate-restricted antigen. J. Immunother. Cancer 9, e001649 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Altschul, S. F. et al. Protein database searches using compositionally adjusted substitution matrices. FEBS J. 272, 5101–5109 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pevsner, J. Bioinformatics and Functional Genomics (John Wiley & Sons, 2015).

  43. Malinen, M., Niskanen, E. A., Kaikkonen, M. U. & Palvimo, J. J. Crosstalk between androgen and pro-inflammatory signaling remodels androgen receptor and NF-κB cistrome to reprogram the prostate cancer cell transcriptome. Nucleic Acids Res. 45, 619–630 (2017).

    Article  CAS  PubMed  Google Scholar 

  44. Chen, Y., Lun, A. T. L. & Smyth, G. K. From reads to genes to pathways: differential expression analysis of RNA-Seq experiments using Rsubread and the edgeR quasi-likelihood pipeline [version 2; peer review: 5 approved]. F1000Research https://doi.org/10.12688/f1000research.8987.2 (2016).

  45. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Asangani, I. A. et al. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature 510, 278–282 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pomerantz, M. M. et al. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat. Genet. 47, 1346–1351 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Haffner, M. C. et al. Androgen-induced TOP2B-mediated double-strand breaks and prostate cancer gene rearrangements. Nat. Genet. 42, 668–675 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Carlson, A. L. et al. Tracking single cells in live animals using a photoconvertible near-infrared cell membrane label. PLoS ONE https://doi.org/10.1371/journal.pone.0069257 (2013).

Download references

Acknowledgements

We thank members of the Drake laboratory for discussion and insightful comments; F. Veglia for advice with the in vitro suppression assays; K. C. Smith, A. Floratos and the Center for Computational Biology and Bioinformatics at Columbia University for the ChIP-seq analysis; S. Coley, T. Swayne, E. Munteanu and the Confocal and Specialized Microscopy Shared Resource at Columbia University for help with microscopy; L. Dasko-Vincent from the Sidney Kimmel Comprehensive Cancer Center Imaging Facility at Johns Hopkins University for support with the LCM; J. Pevsner for assistance with the protein homology analyses; and B. Johnson for help with the statistical analyses. This study was supported by the US Department of Defense (W81XWH-13-1-0369), US National Institutes of Health National Cancer Institute (R01: CA127153 and R01: CA183929-05), Patrick C. Walsh Fund, OneInSix Foundation and Prostate Cancer Foundation. Research reported in this publication was performed at the CCTI Flow Cytometry Core, supported in part by the Office of the Director, National Institutes of Health under award S10OD020056. Hematoxylin and eosin/IHC staining and image collection for this work was performed at the Molecular Pathology Shared Resource and Confocal and Specialized Microscopy Shared Resource of the Herbert Irving Comprehensive Cancer Center at Columbia University, supported by NIH grant P30 CA013696 (National Cancer Institute). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

Z.A.L.-B., M.C.H., M.G.C., N.C., N.J.V., A.O., R.A.P. and J.P.M. performed the experiments. C.S.H., J.J., C.J.B., P.J.H., M.J.S., A.J.K. and C.A.-S. contributed essential reagents. Z.A.L.-B., M.C.H., A.M.C. and C.G.D. designed and supervised the experiments. M.C.H., K.S.S. and A.M.D.M. coordinated the study on human samples. C.G.D. supervised the study. Z.A.L.-B. and C.G.D. wrote the manuscript. All authors edited the manuscript.

Corresponding author

Correspondence to Charles G. Drake.

Ethics declarations

Competing interests

C.G.D. has served as a consultant for Agenus, Dendreon, Janssen Oncology, Eli Lilly, Merck, AstraZeneca, MedImmune, Pierre Fabre, Genentech and Genocea Biosciences. A.M.C. is a shareholder of Aclaris Therapeutics and a consultant for Dermira and Aclaris Therapeutics. Columbia University has filed a US patent claiming the benefit of US provisional patent application number 62/809,060 (inventors C.G.D. and Z.A.L.-B.) on the use of IL-8/CXCR2 blockade of PMN-MDSC recruitment to the TME for the treatment of prostate cancer. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Christopher Barbieri, Vincenzo Bronte and James Gulley for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Cxcl15 Regulation Upon AR signaling Stimulation and Inflammatory Stimuli in Prostate Tumor Epithelial Cells.

a, Androgen responsive prostate tumor cells progressed from castration-sensitive (CS) to androgen responsive (pADT), and eventually developed castration-resistance (CR). CR was tumor size defined as ≥ 30% of nadir tumor volume. Top, tumor growth curve of MCRedAL tumors (n = 3 mice per group, repeated x2); bottom: treatment and harvest timeline. CTX: Castration. b, Upper: sorting strategy to isolate tumor epithelial cells from a based on their expression of mCherry and their CD45CD11bF4/80 phenotype; Lower: purity check of mCherry+ sorted cells. c, Histogram of log2 fold change comparisons (SD Log2 FC) between pADT and CS groups among all the microarray transcripts (n = 3 tumors per group). d, qRT-PCR quantification of Cxcl15 in Myc-CaP cells cultured at indicated concentrations of DHT for 8hrs and TNFα (50Units/ml) for 6hrs, cells cultured in androgen-free media for 48 hours before stimulation (n = 2 independently cultured replicates per condition, repeated x2). Expression levels normalized to mean ΔCT level in samples cultured in androgen free media without TNFα or DHT. e, qRT-PCR quantification of Cxcl15 in Myc-CaP WT cells expressing either nothing, scramble (Scr) shRNA, or an anti-AR shRNA (KD: knock-down) cultured in the presence of DHT (10 nM) for 8hrs and TNFα (50Units/ml) for 6hrs, cells cultured in androgen-free media for 24hrs before DHT stimulation (n = 2 independently cultured replicates per condition, repeated x2). Expression levels normalized to mean ΔCT level in WT samples cultured in androgen free media without TNFα or DHT. f, qRT-PCR quantification of Cxcl15 in Myc-CaP cells cultured at indicated concentrations of DHT for 8hrs and TNFα (50Units/ml) for 6hrs in the presence and absence of the AR blocker Enzalutamide (10 µM), cells cultured in androgen-free media for 48hrs before stimulation (n = 2 independently cultured replicates per condition, repeated x2). Bar plots represent means with SEM. Unpaired two-tailed (d-f) t-tests were performed, p-values ≤ 0.0001 (****); p-values ≥ 0.05 (ns).

Source data

Extended Data Fig. 2 IL-8 Regulation in Prostate Tumor Epithelial Cells.

a, Percentage input bound in ChIP-qPCR assays assessing binding of AR, pSer5 Pol II, and H3K9ac at the KLK3 (PSA) promoter (top) and upstream region (bottom) in LNCaP cells cultured at indicated concentrations of DHT for 8hrs and TNFα (50Units/ml) for 6hrs, cells cultured in androgen-free media for 72hrs before DHT stimulation (n = 2 independently cultured replicates per group). b, ChIP-Seq analysis of AR and NF-κB p65 subunit at the IL-8 (CXCL8) promoter in LNCaP cells cultured in the presence of either vehicle (DMSO), DHT (100 nM), or TNFα (1000U/ml) (n = 2 replicates per group; GSE83860). c, ChIP-Seq analysis at the IL-8 (CXCL8) promoter for AR and RNA pol II binding in LNCaP cells (top), and AR binding in VCaP cells (bottom). Both cell lines were cultured in the presence of either vehicle (DMSO) or DHT (10 nM) (GSE55064). For b, loci with significant differential binding (black bar) were identified as described in materials and methods. Blue shading marks AR binding site; while orange shading marks NF-κB p65 subunit binding site. Bar plots represent means with SEM. Unpaired one-tailed t-tests were performed, p-values ≤ 0.0001 (****); p-values ≥ 0.05 (ns).

Source data

Extended Data Fig. 3 Chemokine Regulation in Prostate Tumor Epithelial Cells.

a, ChIP-Seq enrichment of AR and NF-κB p65 subunit at the CXCL1, CXCL2, CXCL5, and CXCL12 promoters in LNCaP cells cultured in the presence of either vehicle (DMSO), AR signaling (DHT: 100 nM), or an inflammatory stimuli (TNFα: 1000U/ml) (n = 2 independently cultured replicates per group; GSE83860). Orange shading marks NF-κB p65 subunit binding site. b, ChIP-Seq enrichment of AR at the CXCL1, CXCL2, CXCL5, and CXCL12 promoters in primary human prostate cancers (n = 9 patients; GSE56288).

Extended Data Fig. 4 IL-8 Expression in Mouse and Human Prostate Cancer.

a, Representative images of positive and negative controls (cnts: murine and bacteria probe, respectively) in Myc-CaP tumors from indicated treatment groups highlight the specificity of RNA In situ hybridization independently of treatment groups. Tumors were harvested when tumor volume reached ~500mm3 (CS group) or at the time of castration-resistance (CR). Prostate tumor tissue sections were hybridized with CF568-labeled probe sets (white) to Cxcl15, CF640-labeled anti-PanCK antibody (red), and CF488-labeled anti-CD45 antibody (green). Nuclei were counterstained with DAPI (blue). b, qRT-PCR quantification of IL-8 in AR positive castration-sensitive (LNCaP, LAPC4, and VCaP) and AR independent castration-resistant (CWR22Rv1, DU145, and PC3) human prostate cancer cell lines (n = 2 independently cultured replicates per group, repeated x2). IL-8 expression levels were normalized to mean ΔCT level in DU145 samples. c, CXCL1, CXCL2, CXCL5, and CXCL12 protein expression in human AR positive castration-sensitive LNCaP cell line (CS) and it’s isogenic castration-resistant counterpart LNCaP-abl (CR) quantified by MSD (n = 3 independently cultured replicates per group, repeated x1). d, IL-8 protein expression in LNCaP and AR-independent PC3 human prostate cancer cell lines quantified by MSD, replicates as in c. e, Treatment scheme for GFP induction with Doxycycline (DOX) in transgenic Hoxb13-rtTA | TetO-H2BGFP mice for specific isolation of benign murine prostate epithelial cells from castration-sensitive (CS), androgen-deprivation treated (pADT) non-tumor bearing mice, and ADT-treated mice that received testosterone repletion (pADT + T). f, IL-8-RISH quantification in human prostate tumor specimens from untreated (n = 20 patients) or ADT-treated (n = 15 patients; NCT01696877). Quantification was restricted to tumor areas and represented as number of positive cells per mm2. Images representative of 3 independent experiments. RISH images are 60X magnification; scale bar=100 μm. For b, Tukey’s multiple comparisons test with a single pooled variance was performed, p-values ≤ 0.0001 (****). Bar plots represent means with SEM. Unpaired two-tailed (c-d) t-tests were performed, p-values ≤ 0.0001 (****); p-values ≥ 0.05 (ns). For f a Mann-Whitney U test was used due to the non-normal data distribution observed. The box plot includes 25th to 75th of all IL-8 positive cells/mm2 values with horizontal line indicating the median values. The whiskers represent the highest values, including outliers and extremes.

Source data

Extended Data Fig. 5 PMN-MDSCs Infiltration Relays on IL-8 (Cxcl15) / CXCR2 Signaling Following ADT.

a, PMN-MDSCs as a percentage of CD45+ cells in the TME of indicated human prostate tumors as determined by flow cytometry (n = 3 LNCaP and n = 6 PC3 tumors per group, 2 independent experiments). b, Representative H&E and immunohistochemistry (Ly6G and F4/80) sections of the indicated human prostate xenografts (repeated x3). c, Dorsal prostate transgenic NPK tumors (p = 0.0493) and d, Anterior prostate transgenic NPK tumor (p = 0.0006). Left: representative H&E and immunohistochemistry of Ly6G in transgenic NPK tamoxifen-inducible prostate tumors harvested from untreated (CS group) or ADT-treated mice (CR group). CTX (degarelix) was administrated 3 months after tamoxifen-induction, and tumors were harvest 30 days later; right: Ly6G positive counts per mm2 of tumor area as described in the methods section. e, PMN-MDSC density normalized to mg of tumor weight in transgenic NPK tamoxifen-inducible prostate tumors from mice treated as described in c-d (p = 0.0208; cells/mg; n = 4 CS & n = 3 CR tumors per group). f, Left: representative H&E and immunohistochemistry of Ly6G in non-cancerous murine prostate from castration-sensitive (CS), androgen-deprivation treated (pADT) non-tumor bearing mice, and ADT-treated mice that received testosterone repletion (pADT + T). Repeated x2; right: counts of Ly6G positive cells per field as described in the methods section. g, Representative histograms of protein expression determined by flow cytometry in PMN-MDSCs from indicated organs (repeated x2). h, Representative H&E and immunohistochemistry (Ly6G and F4/80) on CR-Myc-CaP allografts treated as indicated (repeated x3). i, Representative H&E and immunohistochemistry (Ly6G and F4/80) in PC3 tumor xenografts treated as indicated (repeated x3). H&E and IHC images are 40X magnification; scale bar=50 μm. Bar plots represent means with SEM. Unpaired two-tailed (a-e) t-tests were performed, p-values ≤ 0.0001 (****); p-values ≥ 0.05 (ns). For f, Tukey’s multiple comparisons test with a single pooled variance was performed.

Source data

Supplementary information

Source data

Source Data Fig. 1

Numerical source data.

Source Data Fig. 2

Numerical source data.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 5

Numerical source data.

Source Data Fig. 6

Numerical source data.

Source Data Fig. 7

Numerical source data.

Source Data Extended Data Fig. 1

Numerical source data.

Source Data Extended Data Fig. 2

Numerical source data.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 5

Numerical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lopez-Bujanda, Z.A., Haffner, M.C., Chaimowitz, M.G. et al. Castration-mediated IL-8 promotes myeloid infiltration and prostate cancer progression. Nat Cancer 2, 803–818 (2021). https://doi.org/10.1038/s43018-021-00227-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00227-3

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