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:

Landscape of prostate-specific membrane antigen heterogeneity and regulation in AR-positive and AR-negative metastatic prostate cancer

This article has been updated

Abstract

Tumor expression of prostate-specific membrane antigen (PSMA) is lost in 15–20% of men with castration-resistant prostate cancer (CRPC), yet the underlying mechanisms remain poorly defined. In androgen receptor (AR)-positive CRPC, we observed lower PSMA expression in liver lesions versus other sites, suggesting a role of the microenvironment in modulating PSMA. PSMA suppression was associated with promoter histone 3 lysine 27 methylation and higher levels of neutral amino acid transporters, correlating with 18F-fluciclovine uptake on positron emission tomography imaging. While PSMA is regulated by AR, we identified a subset of AR-negative CRPC with high PSMA. HOXB13 and AR co-occupancy at the PSMA enhancer and knockout models point to HOXB13 as an upstream regulator of PSMA in AR-positive and AR-negative prostate cancer. These data demonstrate how PSMA expression is differentially regulated across metastatic lesions and in the context of the AR, which may inform selection for PSMA-targeted therapies and development of complementary biomarkers.

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: Suppression of PSMA in prostate cancer liver metastases.
Fig. 2: Intra-tumoral heterogeneity of PSMA in liver lesions.
Fig. 3: Investigation of the PSMA cistrome across prostate cancer progression identifies a PSMA-positive subset of AR-negative NEPC tumors.
Fig. 4: HOXB13 binds to intronic and upstream enhancers of PSMA gene and regulates its expression.
Fig. 5: PSMA suppression is associated with H3K27ac and H3K27me at FOLH1 gene locus.
Fig. 6: 18F-fluciclovine PET delineates PSMA-suppressed CRPC and NEPC tumors.

Similar content being viewed by others

Data availability

All RNA-seq generated for the study, including LNCaP-WT (n = 3) and LNCaP-LMD (n = 3) cell lines and 22Rv1 metastatic xenograft model tumors (primary (n = 3), lymph nodes (n = 3) and liver (n = 3)) are accessible via GEO (accession no. GSE211452). H3K27ac ChIP-seq data of in vitro models are accessible via GEO accession no. GSE221613. Peak calls of HOXB13 ChIP were obtained from GEO accession no. GSE96652. HOXB13 ChIP peak intensity among healthy (n = 15), localized prostate cancer (n = 13) and CRPC samples (n = 15) were from GEO accession no. GSE130408. RNA-seq data from LNCaP and 22Rv1 cell lines with overexpression and knockdown of HOXB13 were obtained from GEO accession nos. GSE153585 (ref. 15) and GSE153586 (ref. 16). RNA-seq reads were aligned to the human reference genome (GRCh38). Source data have been provided as Source Data files. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

No customized code was used in the present study.

Change history

  • 22 May 2023

    In the version of this article initially published, the citation to Figure 6i originally cited Figure 5i, while the panel label for Figure 5h appeared originally as 5f; the errors have been corrected in the HTML and PDF versions of the article.

References

  1. Miyahira, A. K. et al. Meeting report from the Prostate Cancer Foundation PSMA theranostics state of the science meeting. Prostate https://doi.org/10.1002/pros.24056 (2020).

  2. Israeli, R. S., Powell, C. T., Corr, J. G., Fair, W. R. & Heston, W. D. Expression of the prostate-specific membrane antigen. Cancer Res. 54, 1807–1811 (1994).

    CAS  PubMed  Google Scholar 

  3. Narayan, V. et al. PSMA-targeting TGF-β-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat. Med. https://doi.org/10.1038/s41591-022-01726-1 (2022).

  4. Sartor, O. et al. Lutetium-177–PSMA-617 for metastatic castration-resistant prostate cancer. N. Engl. J. Med. 385, 1091–1103 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Paschalis, A. et al. Prostate-specific membrane antigen heterogeneity and DNA repair defects in prostate cancer. Eur. Urol. 76, 469–478 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hofman, M. S. et al. [(177)Lu]Lu-PSMA-617 versus cabazitaxel in patients with metastatic castration-resistant prostate cancer (TheraP): a randomised, open-label, phase 2 trial. Lancet 397, 797–804 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Evans, M. J. et al. Noninvasive measurement of androgen receptor signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen. Proc. Natl Acad. Sci. USA 108, 9578–9582 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Beltran, H. et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat. Med. 22, 298–305 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bakht, M. K. et al. Neuroendocrine differentiation of prostate cancer leads to PSMA suppression. Endocr.-Relat. Cancer 26, 131–146 (2019).

    Article  CAS  Google Scholar 

  10. Watt, F. et al. A tissue-specific enhancer of the prostate-specific membrane antigen gene, FOLH1. Genomics 73, 243–254 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Noss, K. R., Wolfe, S. A. & Grimes, S. R. Upregulation of prostate-specific membrane antigen/folate hydrolase transcription by an enhancer. Gene 285, 247–256 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Yu, J. et al. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell 17, 443–454 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Abida, W. et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc. Natl Acad. Sci. USA 116, 11428–11436 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Brady, L. et al. Inter- and intra-tumor heterogeneity of metastatic prostate cancer determined by digital spatial gene expression profiling. Nat. Commun. 12, 1426 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chen, Z. et al. Diverse AR-V7 cistromes in castration-resistant prostate cancer are governed by HoxB13. Proc. Natl Acad. Sci. USA 115, 6810–6815 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lu, X. et al. HOXB13 suppresses de novo lipogenesis through HDAC3-mediated epigenetic reprogramming in prostate cancer. Nat. Genet. https://doi.org/10.1038/s41588-022-01045-8 (2022).

  17. Kron, K. J. et al. TMPRSS2-ERG fusion co-opts master transcription factors and activates NOTCH signaling in primary prostate cancer. Nat. Genet. 49, 1336–1345 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Grasso, C. S. et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 487, 239–243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ross-Adams, H. et al. Integration of copy number and transcriptomics provides risk stratification in prostate cancer: a discovery and validation cohort study. eBioMedicine 2, 1133–1144 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chandrashekar, D. S. et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia 19, 649–658 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kasperzyk, J. L. et al. Prostate-specific membrane antigen protein expression in tumor tissue and risk of lethal prostate cancer. Cancer Epidemiol. Biomarkers Prev. 22, 2354–63 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Giambartolomei, C. et al. H3K27ac HiChIP in prostate cell lines identifies risk genes for prostate cancer susceptibility. Am. J. Hum. Genet. 108, 2284–2300 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tang, F. et al. Chromatin profiles classify castration-resistant prostate cancers suggesting therapeutic targets. Science 376, eabe1505 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Thang, S. P. et al. Poor outcomes for patients with metastatic castration-resistant prostate cancer with low prostate-specific membrane antigen (PSMA) expression deemed ineligible for 177Lu-labelled PSMA radioligand therapy. Eur. Urol. Oncol. https://doi.org/10.1016/j.euo.2018.11.007 (2019).

  25. Gusman, M. et al. Review of 18F-fluciclovine PET for detection of recurrent prostate cancer. Radiographics 39, 822–841 (2019).

    Article  PubMed  Google Scholar 

  26. Saarinen, I. et al. Correlation between 18F-1-amino-3-fluorocyclobutane-1-carboxylic acid (18F-fluciclovine) uptake and expression of alanine-serine-cysteine-transporter 2 (ASCT2) and L-type amino acid transporter 1 (LAT1) in primary prostate cancer. EJNMMI Res. 9, 50 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Okudaira, H. et al. Putative transport mechanism and intracellular fate of trans-1-amino-3-18F-fluorocyclobutanecarboxylic acid in human prostate cancer. J. Nucl. Med. 52, 822–829 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Bakht, M. K. et al. Differential expression of glucose transporters and hexokinases in prostate cancer with a neuroendocrine gene signature: a mechanistic perspective for FDG imaging of PSMA-suppressed tumors. J. Nucl. Med. 61, 904–910 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Beltran, H. et al. The role of lineage plasticity in prostate cancer therapy resistance. Clin. Cancer Res. 25, 6916–6924 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Khreish, F. et al. Response and outcome of liver metastases in patients with metastatic castration-resistant prostate cancer (mCRPC) undergoing 177Lu-PSMA-617 radioligand therapy. Eur. J. Nucl. Med. Mol. Imaging. 48, 103–112 (2021).

    Article  CAS  PubMed  Google Scholar 

  31. Ewing, C. M. et al. Germline mutations in HOXB13 and prostate-cancer risk. N. Engl. J. Med. 366, 141–149 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Norris, J. D. et al. The homeodomain protein HOXB13 regulates the cellular response to androgens. Mol. Cell 36, 405–416 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Pomerantz, M. M. et al. Prostate cancer reactivates developmental epigenomic programs during metastatic progression. Nat. Genet. 52, 790–799 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Epstein, J. I. et al. Proposed morphologic classification of prostate cancer with neuroendocrine differentiation. Am. J. Surg. Pathol. 38, 756–767 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Puca, L. et al. Patient-derived organoids to model rare prostate cancer phenotypes. Nat. Commun. 9, 2404 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Yang, X. et al. A public genome-scale lentiviral expression library of human ORFs. Nat. Methods 8, 659–661 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Baca, S. C. et al. Reprogramming of the FOXA1 cistrome in treatment-emergent neuroendocrine prostate cancer. Nat. Commun. 12, 1979 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Huang, Q. et al. A prostate cancer susceptibility allele at 6q22 increases RFX6 expression by modulating HOXB13 chromatin binding. Nat. Genet. 46, 126–135 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Kasashima, H. et al. An orthotopic implantation mouse model of hepatocellular carcinoma with underlying liver steatosis. STAR Protoc. 1, 100185 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Malviya, G. et al. 18F-Fluciclovine PET metabolic imaging reveals prostate cancer tumour heterogeneity associated with disease resistance to androgen deprivation therapy. EJNMMI Res. 10, 143 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. He, M. X. et al. Transcriptional mediators of treatment resistance in lethal prostate cancer. Nat. Med. 27, 426–433 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bakht, M. K. et al. Identification of alternative protein targets of glutamate-ureido-lysine associated with PSMA tracer uptake in prostate cancer cells. Proc. Natl Acad. Sci. USA 119, e2025710119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Beltran, H. et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 1, 487–495 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Torre, D., Lachmann, A. & Ma’ayan, A. BioJupies: automated generation of interactive notebooks for RNA-seq data analysis in the cloud. Cell Sys. 7, 556–561 (2018).

    Article  CAS  Google Scholar 

  46. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Wang, L., Wang, S. & Li, W. RSeQC: quality control of RNA-seq experiments. Bioinformatics 28, 2184–2185 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Crowe, A. R. & Yue, W. Semi-quantitative determination of protein expression using immunohistochemistry staining and analysis: an integrated protocol. Bio. Protoc. https://doi.org/10.21769/BioProtoc.3465 (2019).

  49. Zheng, R. et al. Cistrome Data Browser: expanded datasets and new tools for gene regulatory analysis. Nucleic Acids Res. 47, D729–D735 (2018).

    Article  PubMed Central  Google Scholar 

  50. Beltran, H. et al. Whole-exome sequencing of metastatic cancer and biomarkers of treatment response. JAMA Oncol. 1, 466–474 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work supported by the Prostate Cancer Foundation (to H.B.), US Department of Defense (W81XWH-17-1-0653 to H.B., W81XWH-22-1-0010 to M.K.B. and W81XWH-22-1-0197 to V.B.V.) and NIH National Cancer Institute (R37CA241486 and P50-CA211024 to H.B.). Y.Y. is supported by the Japan Society for the Promotion of Science. Partial support for the work was provided by NIH Center grant P30 CA08748 (Small Animal Imaging Core Facility and the Radiochemistry and Molecular Imaging Probe core). Support from NIH R35 CA232130 (to J.S.L.), DOD-IDEA Award grant W81XWH-19-1-0536 (to N.P.) and National Cancer Center (to V.B.V.) is acknowledged. We acknowledge support of Blue Earth Diagnostics for providing 18F-fluciclovine and 18F-rh-PSMA tracers. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

H.B. supervised the research. M.K.B. and H.B. conceived and designed the study. M.K.B and Y.Y. performed in vitro experiments. Y.Y. and S.K. developed animal models and M.K.B., Y.Y., S.K. and M.M.G. characterized animal models. M.K.B., V.B.V., J.S., H.W.L. and M.F. designed ChIP experiments and analyzed the data. J.A.K., T.M.K., N.P. and J.S.L. performed and analyzed 68Ga-PSMA PET imaging experiments. M.K.B., S.H.A., A.P.B. and Q.N. performed and analyzed 18F-rh-PSMA and 18F-fluciclovine PET imaging experiments. F.K. and O.E. shared samples and reviewed pathology from WCM. A.C. provided 18F-rh-PSMA and 18F-fluciclovine. M.K.B., K.M. and H.W.L. performed statistical and bioinformatics analyses. F.K., O.E., H.W.L., N.P., J.S.L., M.F., A.P.B., Q.N. and H.B. reviewed and approved data for publication. M.K.B. and H.B. wrote the first draft of the manuscript. All authors contributed to the writing and editing of the revised manuscript and approved the manuscript.

Corresponding author

Correspondence to Himisha Beltran.

Ethics declarations

Competing interests

H.B. participated in a virtual advisory board meeting with Blue Earth Diagnostics in 2021. A.C. is employed by Blue Earth Diagnostics. Blue Earth Diagnostics provided tracer for this study but had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. H.B. has also served as consultant/advisory board member for Janssen, Astellas, AstraZeneca, Merck, Pfizer, Foundation Medicine, Amgen, Bayer, Oncorus, LOXO, Daicchi Sankyo and Curie Therapeutics and has received research funding (to institution) from Janssen, AbbVie/Stemcentrx, Eli Lilly, Astellas, Millennium, Bristol Myers Squibb, Circle Pharma and Daicchi Sankyo. O.E. is supported by Janssen, J&J, AstraZeneca, Volastra and Eli Lilly research grants. He is scientific advisor and equity holder in Freenome, Owkin, Volastra Therapeutics, Pionyr Immunotherapeutics, Harmonic and One Three Biotech and a paid scientific advisor to Champions Oncology. All other authors have no competing interests.

Peer review

Peer review information

Nature Cancer thanks James Gulley, Amar Kishan and Li Xin 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 PSMA (FOLH1) gene expression is mostly correlated with AR and NEPC markers except in liver metastatic tumors with no NE features.

a, Heatmaps of the expression levels of PSMA gene (FOLH1), AR-markers and NE markers in metastatic CRPC samples from the International SU2C/PCF Dream Team dataset13. b, Expression of PSA gene (KLK3) and sites of metastases in the International SU2C/PCF Dream Team CRPC dataset13. c, AR score (left) and NEPC score (right) liver (n = 39), lymph node (n = 115) and bone (n = 73) metastatic CRPC samples in the International SU2C/PCF Dream Team dataset. The size of data points is proportional with the level of KLK3 gene expression in each sample. The lines and squares inside each box are the median and mean, respectively. The upper box border represents the 75th quartile, lower box border represents the 25th quartile and whiskers represent outliers by using the 1.5 interquartile range rule. In b, the data were analyzed by one-way ANOVA followed by Tukey’s multiple comparison tests.

Source data

Extended Data Fig. 2 PSMA heterogeneity in CRPC can be independent from AR score.

a, PSMA protein expression by IHC displaying heterogeneity in metastatic tumors from liver obtained at autopsy in two patients with CRPC. b, Evaluation of AR and NEPC markers in metastatic liver tumors of Mouse A01 (from Fig. 1g). c, Images of spontaneous metastatic tumors following orthotopic injection of 22Rv1 cell line in Mouse Model A02. Purple arrow points to a metastatic tumor in liver. d, Evaluation of AR and PSMA protein expression by IHC in Mouse A02. e, Western blot analyses of PSMA, AR and NKX3-1 protein levels in Mouse A02. The experiment in e was repeated 2 times with N = 2 independently collected samples with similar results. f, Representative images of spontaneous metastatic tumors in Mouse Model A03. g, Evaluation of AR and PSMA by IHC in Mouse A03. h, AR-score in primary and metastatic tissues of orthotopic 22Rv1 mouse models. LNCaP used as a reference. i, Heatmaps of the expression levels of PSMA gene (FOLH1), AR-markers and NE markers in primary and metastatic tissues of orthotopic 22Rv1 mouse models. j, Principal Component Analysis (PCA) identified global similarity patterns in N = 3 primary prostate tumors, N = 3 metastatic tumors from lymph nodes, and N = 6 PSMA-high metastatic tumors in liver. In b, d and g, IHC experiments were performed once using proper positive and negative controls.

Source data

Extended Data Fig. 3 Single-cell transcriptome analysis of the 22Rv1-WT cell line and 22Rv1 liver metastasis.

a, Uniform Manifold Approximation and Projection (UMAP) reduced dimension plots shows 22Rv1-WT consists of different clusters. The majority of 22Rv1 liver metastasis cells grouped as a single cluster, albeit this scRNA analysis was limited due to the number of viable cells of the sequenced liver metastasis (n = 197 cells) compared with 22Rv1-WT cell line (n = 3467 cells). b, Single-cell RNA expression of FOLH1, AR, HOXB13, LAT1 and NE markers over the UMAP representation of the map. The expression of LAT1 in Cluster 3 shows PSMA-low subpopulation of 22Rv1-WT cells are not LAT1 positive. This observation implies there is a heterogeneity within the PSMA-low cell populations. c, Unsupervised clusters were annotated as three clusters with distinct FOLH1 levels. d, Expression of FOLH1 in identified clusters. e, Stacked barplot displaying percentage of each cluster in 22Rv1-WT and 22Rv1 liver metastasis. Notably, only 10.9% of 22Rv1-WT belong to PSMA-low Cluster 3. However, more than 58% of 22Rv1 liver metastasis are within Cluster 3. In d, the data were analyzed by one-way ANOVA followed by Šídák’s multiple comparison tests. In a-c, data presented is based on data from a single experiment.

Source data

Extended Data Fig. 4 Estimation of clinically relevant FOLH1-positive regulators during progression from benign prostate to NEPC.

a, PSMA protein levels in prostate cancer models annotated by their FOLH1 mRNA expression levels obtained by RNA-seq. Since 22Rv1 xenograft tumors with RNA expression of 31.3 RPKM are pathologically considered as PSMA-positive xenografts and they are radiologically detectable with moderate PSMA PET uptake, we defined samples with FOLH1 expression levels more than 50 RPKM as FOLH1-high tumors and samples with FOLH1 expression levels less than 5 RPKM as FOLH1-low tumors. The column chart show mean ± s.e.m for N = 3 independently collected samples. b, Expression of FOLH1 during progression of prostate cancer toward NEPC. The incidence of FOLH1-high was at its maximum among primary prostate cancer samples. On the other hand, FOLH1-low was at its maximum among NEPC samples. c, A differential expression (DGE) performed on each cohort to determine which genes are expressed at FOLH1-high tumors. d, Short (1 kb), mid-range (10 kb) and long-range (100 kb) influence scores were calculated using Cistrome DB21. e, Schematic of generation of Venn diagram of overlapping differentially expressed genes in FOLH1-high cohorts with the estimated FOLH1 regulator to predict clinically relevant FOLH1-positive regulators.

Source data

Extended Data Fig. 5 Estimation of FOLH1-positive regulators among CRPC tumors with and without neuroendocrine (NE) features.

a, Heat map of the expression levels of PSMA gene (FOLH1), AR-markers, NE markers and projected PSMA regulators in metastatic CRPC samples from the International SU2C/PCF Dream Team dataset 13 (N = 224 tumors) a, Expression levels of FOLH1 in prostate tissue during progression from benign to NEPC. b, Volcano plot of DGE analysis in FOLH1-high vs. FOLH1-low among CRPC tumors with and without NE features. c, Venn diagrams illustrate the overlap of differentially expressed genes in FOLH1-high cohorts, with FOLH1 potential transcription factors estimated by Cistrome DB21.

Extended Data Fig. 6 HOXB13 is a positive regulator of PSMA (FOLH1).

a, Overexpression of WT-HOXB13 in LNCaP-shHOXB13 cells rescues FOLH1 expression while overexpression of G84E mutant-HOXB13 cannot rescue suppression of FOLH1. Data from GEO accession GSE15358516. b, Significant reduction in FOLH1 expression in LN95 (left) and 22Rv1 (right). Data from GEO accession GSE9937815. The boxes represent experimental replicates and samples with same treatment are labeled with same color. c, Bar charts of the expression levels of AR (top) and HOXB13 (bottom) in prostate tumors during progression from benign to NEPC. The bar colors represent FOLH1 levels in each sample. d, AR (top) and HOXB13 (bottom) ChIP-seq intensity in representative CRPC samples from GEO accession GSE13040850.

Extended Data Fig. 7 Gene expression of PSMA (FOLH1) in preclinical models and corresponding chromatin accessibility of its promoter and upstream enhancer are highly correlated.

a, Heat map of ATAC-seq intensity among prostate cancer models at the FOLH1 gene annotated with FOLH1 expression in each sample. Pearson correlation between the intensity of ATAC-seq peak and the expression of FOLH1 at promoter (b), close to upstream enhancer (c) and on upstream enhancer (d-e). Data from GEO accession GSE19919025. In b-e, the scatter plots show the intensity of ATAC-seq peak (y axis) and the expression of FOLH1 (x axis) for N = 18 preclinical prostate cancer models.

Source data

Extended Data Fig. 8 Elevation of LAT1 and ASCT2 gene expression in NEPC and low PSMA CRPC.

a, Tissue sections of CRPC and NEPC models stained with LAT1 and 4F2hc antibodies. Scale bar: 200 μm b, Western blot analyses of PSMA, LAT1 and ASCT2 protein levels of models. c, Tissue sections of NEPC model WCM1078 stained with ASCT2 antibody. Scale bar: 100 μm d, Evaluation of the expression of ASCT2 (SLC1A5) gene in Beltran8 dataset for N = 34 CRPC tumors and N = 15 NEPC tumors. The lines and squares inside each box are the median and mean, respectively. The upper box border represents the 75th quartile, lower box border represents the 25th quartile and whiskers represent the outlier by using the 1.5 interquartile range rule. e, Schematic illustration of anatomic sites of samples and expression levels of PSMA, LAT1 and ASCT2 in each sample. The representative images are shown for N = 3 (a-c) independently collected samples.

Source data

Extended Data Fig. 9 Proposed model of PSMA regulation in prostate cancer.

PSMA (FOLH1) expression is activated in prostate cancer via binding of both AR and its cofactor HOXB13 to the PSMA enhancer. Even in the absence of AR expression, a subset of AR-negative tumors will still express PSMA due to HOXB13 binding of the PSMA enhancer. CRPC tumors may suppress or lose PSMA expression either due to loss of AR/HOXB13 binding of the PSMA promotor and/or methylation of the PSMA promotor.

Supplementary information

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig 7

Statistical Source Data.

Source Data Extended Data Fig 8

Statistical Source Data.

Source Data Extended Data Fig 8

Unprocessed western blots.

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

Bakht, M.K., Yamada, Y., Ku, SY. et al. Landscape of prostate-specific membrane antigen heterogeneity and regulation in AR-positive and AR-negative metastatic prostate cancer. Nat Cancer 4, 699–715 (2023). https://doi.org/10.1038/s43018-023-00539-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00539-6

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