Abstract
Prostate cancer remains a substantial health challenge, with >375,000 annual deaths amongst men worldwide. Most prostate cancer-related deaths are attributable to the development of resistance to standard-of-care treatments. Characterization of the diverse and complex surfaceome of treatment-resistant prostate cancer, combined with advances in drug development that leverage cell-surface proteins to enhance drug delivery or activate the immune system, have provided novel therapeutic opportunities to target advanced prostate cancer. The prostate cancer surfaceome, including proteins such as prostate-specific membrane antigen (PSMA), B7-H3, six transmembrane epithelial antigen of the prostate 1 (STEAP1), delta-like ligand 3 (DLL3), trophoblastic cell-surface antigen 2 (TROP2), prostate stem cell antigen (PSCA), HER3, CD46 and CD36, can be exploited as therapeutic targets, as regulatory mechanisms might contribute to the heterogeneity of expression of these proteins and subsequently affect treatment response and resistance. Specific treatment strategies targeting the surfaceome are in clinical development, including radionuclides, antibody–drug conjugates, T cell engagers and chimeric antigen receptor (CAR) T cells. Ultimately, biomarker development and clinical implementation of these agents will be informed and refined by further understanding of the biology of various targets; the target specificity and sensitivity of different agents; and off-target and toxic effects associated with these agents. Understanding the dynamic nature of cell-surface targets and non-overlapping expression patterns might also lead to future combinational strategies.
Key points
-
Surface proteins such as prostate-specific membrane antigen (PSMA), B7-H3, six transmembrane epithelial antigen of the prostate 1 (STEAP1), delta-like ligand 3 (DLL3), trophoblastic cell-surface antigen 2 (TROP2), prostate stem cell antigen (PSCA), HER3 and CD46 are a promising class of novel therapeutic targets in prostate cancer.
-
Radioligand therapies consist of antibodies or small molecules conjugated to radioactive isotopes to deliver targeted radiation. Advances in PSMA imaging and PSMA-targeted radioligands (such as [177Lu]-PSMA-617) have led to improved outcomes in metastatic castration-resistant prostate cancer. Other radioligand therapies are in development.
-
Antibody–drug conjugates consist of an antigen-specific antibody, a chemical linker and a cytotoxic payload, which enable targeted delivery of chemotherapy while minimizing systemic toxicity. Several antibody–drug conjugates are under investigation in prostate cancer, with challenges including stability, toxicity and resistance mechanisms.
-
Targeted T cell engager and chimeric antigen receptor T cell therapies are designed to overcome the limited efficacy of targeting non-specific immune checkpoints in prostate cancer. These new immune strategies are focused on balancing immune activation and antitumour activity while minimizing toxicity associated with excessive immune stimulation.
-
Broad challenges in targeting cell-surface proteins include tumour heterogeneity and dynamic target expression; patient-specific tumour, host and drug features that might limit efficacy; and the development of acquired resistance mechanisms including loss of target expression.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout


Similar content being viewed by others
References
Paschalis, A. & de Bono, J. S. Prostate cancer 2020: ‘the times they are a’changing’. Cancer Cell 38, 25–27 (2020).
Posdzich, P. et al. Metastatic prostate cancer — a review of current treatment options and promising new approaches. Cancers 15, 461 (2023).
Collins, D. M., Bossenmaier, B., Kollmorgen, G. & Niederfellner, G. Acquired resistance to antibody-drug conjugates. Cancers 11, 394 (2019).
Coleman, N., Yap, T. A., Heymach, J. V., Meric-Bernstam, F. & Le, X. Antibody-drug conjugates in lung cancer: dawn of a new era? NPJ Precis. Oncol. 7, 5 (2023).
Wängler, B., Schirrmacher, R., Bartenstein, P. & Wängler, C. Chelating agents and their use in radiopharmaceutical sciences. Mini Rev. Med. Chem. 11, 968–983 (2011).
Nasr, D. et al. Radioimmunoconjugates in the age of modern immuno-oncology. Life Sci. 310, 121126 (2022).
Witzig, T. E. et al. Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin’s lymphoma. J. Clin. Oncol. 20, 2453–2463 (2002).
Hussain, M. et al. Differential effect on bone lesions of targeting integrins: randomized phase II trial of abituzumab in patients with metastatic castration-resistant prostate cancer. Clin. Cancer Res. 22, 3192–3200 (2016).
Goebeler, M.-E. & Bargou, R. C. T cell-engaging therapies — BiTEs and beyond. Nat. Rev. Clin. Oncol. 17, 418–434 (2020).
Archer, S. et al. CB307: a dual targeting costimulatory Humabody VH therapeutic for treating PSMA-positive tumors. Clin. Cancer Res. 30, 1595–1606 (2024).
Leclercq, G. et al. Dissecting the mechanism of cytokine release induced by T-cell engagers highlights the contribution of neutrophils. Oncoimmunology 11, 2039432 (2022).
Morris, E. C., Neelapu, S. S., Giavridis, T. & Sadelain, M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat. Rev. Immunol. 22, 85–96 (2022).
de Spéville, B. D. & Moreno, V. Antidrug antibodies and drug development: challenges in the immunotherapy era. Clin. Cancer Res. 27, 2669–2671 (2021).
Dorff, T. B. et al. PSCA-CAR T cell therapy in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat. Med. 30, 1636–1644 (2024).
Sutherland, S. I. M., Ju, X., Horvath, L. G. & Clark, G. J. Moving on from sipuleucel-T: new dendritic cell vaccine strategies for prostate cancer. Front. Immunol. 12, 641307 (2021).
Rastogi, I., Muralidhar, A. & McNeel, D. G. Vaccines as treatments for prostate cancer. Nat. Rev. Urol. 20, 544–559 (2023).
Esapa, B. et al. Target antigen attributes and their contributions to clinically approved antibody-drug conjugates (ADCs) in haematopoietic and solid cancers. Cancers 15, 1845 (2023).
Vogel, C. L. et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J. Clin. Oncol. 20, 719–726 (2002).
Modi, S. et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N. Engl. J. Med. 387, 9–20 (2022).
Banerji, U. et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 20, 1124–1135 (2019).
Curigliano, G. et al. Trastuzumab deruxtecan (T-DXd) vs physician’s choice of chemotherapy (TPC) in patients (pts) with hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)-low or HER2-ultralow metastatic breast cancer (mBC) with prior endocrine therapy (ET): primary results from DESTINY-Breast06 (DB-06). J. Clin. Oncol. 42, LBA1000–LBA1000 (2024).
Majzner, R. G. et al. Tuning the antigen density requirement for CAR T-cell activity. Cancer Discov. 10, 702–723 (2020).
Powles, T. et al. Enfortumab vedotin and pembrolizumab in untreated advanced urothelial cancer. N. Engl. J. Med. 390, 875–888 (2024).
Powles, T. B. et al. 1966MO EV-302: exploratory analysis of nectin-4 expression and response to 1L enfortumab vedotin (EV) + pembrolizumab (P) in previously untreated locally advanced or metastatic urothelial cancer (la/mUC). Ann. Oncol. 35, S1137–S1138 (2024).
Klümper, N. et al. Membranous NECTIN-4 expression frequently decreases during metastatic spread of urothelial carcinoma and is associated with enfortumab vedotin resistance. Clin. Cancer Res. 29, 1496–1505 (2023).
Aggen, D. H., Chu, C. E. & Rosenberg, J. E. Scratching the surface: NECTIN-4 as a surrogate for enfortumab vedotin resistance. Clin. Cancer Res. 29, 1377–1380 (2023).
Sartor, O. et al. Lutetium-177–PSMA-617 for metastatic castration-resistant prostate cancer. N. Engl. J. Med. 385, 1091–1103 (2021).
Vlachostergios, P. J. et al. Imaging expression of prostate-specific membrane antigen and response to PSMA-targeted β-emitting radionuclide therapies in metastatic castration-resistant prostate cancer. Prostate 81, 279–285 (2021).
Kuo, P. et al. [68Ga]Ga-PSMA-11 PET baseline imaging as a prognostic tool for clinical outcomes to [177Lu]Lu-PSMA-617 in patients with mCRPC: a VISION substudy. J. Clin. Oncol. 40, 5002 (2022).
Buteau, J. P. et al. PSMA and FDG-PET as predictive and prognostic biomarkers in patients given [177Lu]Lu-PSMA-617 versus cabazitaxel for metastatic castration-resistant prostate cancer (TheraP): a biomarker analysis from a randomised, open-label, phase 2 trial. Lancet Oncol. 23, 1389–1397 (2022).
Dhatchinamoorthy, K., Colbert, J. D. & Rock, K. L. Cancer immune evasion through loss of MHC class I antigen presentation. Front. Immunol. 12, 636568 (2021).
Carter, P., Smith, L. & Ryan, M. Identification and validation of cell surface antigens for antibody targeting in oncology. Endocr. Relat. Cancer 11, 659–687 (2004).
Reily, C., Stewart, T. J., Renfrow, M. B. & Novak, J. Glycosylation in health and disease. Nat. Rev. Nephrol. 15, 346–366 (2019).
Matsumoto, Y., Jia, N., Heimburg-Molinaro, J. & Cummings, R. D. Targeting Tn-positive tumors with an afucosylated recombinant anti-Tn IgG. Sci. Rep. 13, 5027 (2023).
Wang, M., Zhu, J., Lubman, D. M. & Gao, C. Aberrant glycosylation and cancer biomarker discovery: a promising and thorny journey. Clin. Chem. Lab. Med. 57, 407–416 (2019).
Anczuków, O. & Krainer, A. R. Splicing-factor alterations in cancers. RNA 22, 1285–1301 (2016).
Kaucká, M. et al. Post-translational modifications regulate signalling by Ror1. Acta Physiol. 203, 351–362 (2011).
Dwek, R. A. Biological importance of glycosylation. Dev. Biol. Stand. 96, 43–47 (1998).
Meacham, C. E. & Morrison, S. J. Tumour heterogeneity and cancer cell plasticity. Nature 501, 328–337 (2013).
Cordani, M., Dando, I., Ambrosini, G. & González-Menéndez, P. Signaling, cancer cell plasticity, and intratumor heterogeneity. Cell Commun. Signal. 22, 255 (2024).
Rübben, A. & Araujo, A. Cancer heterogeneity: converting a limitation into a source of biologic information. J. Transl. Med. 15, 190 (2017).
Marusyk, A. & Polyak, K. Tumor heterogeneity: causes and consequences. Biochim. Biophys. Acta 1805, 105–117 (2010).
Gu, X., Wei, S. & Lv, X. Circulating tumor cells: from new biological insights to clinical practice. Signal. Transduct. Target. Ther. 9, 226 (2024).
Zhu, X., Maier, G. & Panda, S. Learning from circadian rhythm to transform cancer prevention, prognosis, and survivorship care. Trends Cancer 10, 196–207 (2024).
Carter, P. Improving the efficacy of antibody-based cancer therapies. Nat. Rev. Cancer 1, 118–129 (2001).
Glennie, M. J. & van de Winkel, J. G. J. Renaissance of cancer therapeutic antibodies. Drug. Discov. Today 8, 503–510 (2003).
Ross, J. et al. Antibody-based therapeutics in oncology. Expert. Rev. Anticancer Ther. 3, 107–121 (2003).
Staudacher, A. H. & Brown, M. P. Antibody drug conjugates and bystander killing: is antigen-dependent internalisation required? Br. J. Cancer 117, 1736–1742 (2017).
Jang, A., Kendi, A. T., Johnson, G. B., Halfdanarson, T. R. & Sartor, O. Targeted alpha-particle therapy: a review of current trials. Int. J. Mol. Sci. 24, 11626 (2023).
Bonin, S. & Stanta, G. Pre-analytics and tumor heterogeneity. N. Biotechnol. 55, 30–35 (2020).
Ceze, N. et al. [Target antigens for therapeutic antibodies in oncology: many candidates, few successes]. Bull. Cancer 94, F129–F136 (2007).
Di Fiore, P. P. et al. erbB-2 is a potent oncogene when overexpressed in NIH/3T3 cells. Science 237, 178–182 (1987).
Hudziak, R. M., Schlessinger, J. & Ullrich, A. Increased expression of the putative growth factor receptor p185HER2 causes transformation and tumorigenesis of NIH 3T3 cells. Proc. Natl Acad. Sci. USA 84, 7159–7163 (1987).
Sun, L. et al. Shed antigen-induced blocking effect on CAR-T cells targeting Glypican-3 in hepatocellular carcinoma. J. Immunother. Cancer 9, e001875 (2021).
Shi, X. et al. Integrative molecular analyses define correlates of high B7-H3 expression in metastatic castrate-resistant prostate cancer. NPJ Precis. Oncol. 6, 80 (2022).
Bakht, M. K. & Beltran, H. Biological determinants of PSMA expression, regulation and heterogeneity in prostate cancer. Nat. Rev. Urol. 22, 26–45 (2024).
Hope, T. A. et al. 68Ga-PSMA-11 PET imaging of response to androgen receptor inhibition: first human experience. J. Nucl. Med. 58, 81–84 (2017).
Sheehan, B. et al. Prostate-specific membrane antigen expression and response to DNA damaging agents in prostate cancer. Clin. Cancer Res. 28, 3104–3115 (2022).
Sosa, M. S., Bragado, P. & Aguirre-Ghiso, J. A. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat. Rev. Cancer 14, 611–622 (2014).
Baldominos, P. et al. Quiescent cancer cells resist T cell attack by forming an immunosuppressive niche. Cell 185, 1694–1708.e19 (2022).
Zhang, X. et al. Induction of mitochondrial dysfunction as a strategy for targeting tumour cells in metabolically compromised microenvironments. Nat. Commun. 5, 3295 (2014).
Rehman, S. K. et al. Colorectal cancer cells enter a diapause-like DTP state to survive chemotherapy. Cell 184, 226–242.e21 (2021).
McHugh, D. et al. COPI vesicle formation and N-myristoylation are targetable vulnerabilities of senescent cells. Nat. Cell Biol. 25, 1804–1820 (2023).
Gabrilovich, D. I., Ostrand-Rosenberg, S. & Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 12, 253–268 (2012).
Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 (2016).
Semenza, G. L. Hypoxia-inducible factors in physiology and medicine. Cell 148, 399–408 (2012).
Brinkmann, V. et al. Neutrophil extracellular traps kill bacteria. Science 303, 1532–1535 (2004).
Cools-Lartigue, J. et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Invest. 123, 3446–3458 (2013).
Demers, M. et al. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc. Natl Acad. Sci. USA 109, 13076–13081 (2012).
Park, J. et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl. Med. 8, 361ra138 (2016).
Albrengues, J. et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 361, eaao4227 (2018).
Meads, M. B., Gatenby, R. A. & Dalton, W. S. Environment-mediated drug resistance: a major contributor to minimal residual disease. Nat. Rev. Cancer 9, 665–674 (2009).
Dizdaroglu, M., Jaruga, P., Birincioglu, M. & Rodriguez, H. Free radical-induced damage to DNA: mechanisms and measurement. Free. Radic. Biol. Med. 32, 1102–1115 (2002).
Azzam, E. I., Jay-Gerin, J.-P. & Pain, D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 327, 48–60 (2012).
Piranfar, A. et al. Radiopharmaceutical transport in solid tumors via a 3-dimensional image-based spatiotemporal model. NPJ Syst. Biol. Appl. 10, 39 (2024).
Cui, C. et al. Synthesis and evaluation of [64Cu]PSMA-617 targeted for prostate-specific membrane antigen in prostate cancer. Am. J. Nucl. Med. Mol. Imaging 7, 40–52 (2017).
Institute of Medicine (US) Committee to Study the Feasibility of, and Need for, Epidemiologic Studies of Adverse Reproductive Outcomes in the Families of Atomic Veterans. Adverse Reproductive Outcomes in Families of Atomic Veterans: The Feasibility of Epidemiologic Studies (National Academies Press, 1995).
Khazaei Monfared, Y. et al. DNA damage by radiopharmaceuticals and mechanisms of cellular repair. Pharmaceutics 15, 2761 (2023
Ku, A., Facca, V. J., Cai, Z. & Reilly, R. M. Auger electrons for cancer therapy — a review. EJNMMI Radiopharm. Chem. 4, 27 (2019).
Holik, H. A. et al. The chemical scaffold of theranostic radiopharmaceuticals: radionuclide, bifunctional chelator, and pharmacokinetics modifying linker. Molecules 27, 3062 (2022).
Hupe, M. C. et al. Expression of prostate-specific membrane antigen (PSMA) on biopsies is an independent risk stratifier of prostate cancer patients at time of initial diagnosis. Front. Oncol. 8, 623 (2018).
Hofman, M. S. et al. [177Lu]-PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): a single-centre, single-arm, phase 2 study. Lancet Oncol. 19, 825–833 (2018).
FDA approves Pluvicto for metastatic castration-resistant prostate cancer. J. Nucl. Med. 63, 13N (2024).
Morris, M. J. et al. 177Lu-PSMA-617 versus a change of androgen receptor pathway inhibitor therapy for taxane-naive patients with progressive metastatic castration-resistant prostate cancer (PSMAfore): a phase 3, randomised, controlled trial. Lancet 404, 1227–1239 (2024).
Sathekge, M. M. et al. Actinium-225-PSMA radioligand therapy of metastatic castration-resistant prostate cancer (WARMTH Act): a multicentre, retrospective study. Lancet Oncol. 25, 175–183 (2024).
Tagawa, S. T. et al. Prostate-specific membrane antigen-targeting alpha emitter via antibody delivery for metastatic castration-resistant prostate cancer: a phase I dose-escalation study of 225Ac-J591. J. Clin. Oncol. 42, 842–851 (2024).
Griffiths, M. R. et al. First-in-human 212Pb-PSMA-targeted α-therapy SPECT/CT imaging in a patient with metastatic castration-resistant prostate cancer. J. Nucl. Med. 65, 664 (2024).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04726033 (2024).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04868604 (2024).
Hammer, S. et al. Preclinical efficacy of a PSMA-targeted thorium-227 conjugate (PSMA-TTC), a targeted alpha therapy for prostate cancer. Clin. Cancer Res. 26, 1985–1996 (2020).
Bakht, M. K. 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).
Seifert, R. et al. Analysis of PSMA expression and outcome in patients with advanced prostate cancer receiving 177Lu-PSMA-617 radioligand therapy. Theranostics 10, 7812–7820 (2020).
Mannweiler, S. et al. Heterogeneity of prostate-specific membrane antigen (PSMA) expression in prostate carcinoma with distant metastasis. Pathol. Oncol. Res. 15, 167–172 (2009).
Stuparu, A. D. et al. Mechanisms of resistance to prostate-specific membrane antigen-targeted radioligand therapy in a mouse model of prostate cancer. J. Nucl. Med. 62, 989–995 (2021).
De Bono, J. S. et al. Baseline ctDNA analyses and associations with outcomes in taxane-naive patients with mCRPC treated with 177Lu-PSMA-617 versus change of ARPI in PSMAfore. J. Clin. Oncol. 42, 5008–5008 (2024).
Bollito, E. et al. Relationship between neuroendocrine features and prognostic parameters in human prostate adenocarcinoma. Ann. Oncol. 12, S159–S164 (2001).
Luboldt, W. et al. Visualization of somatostatin receptors in prostate cancer and its bone metastases with Ga-68-DOTATOC PET/CT. Mol. Imaging Biol. 12, 78–84 (2010).
Yasukawa, R. et al. Excellent response to 177Lu-DOTATATE peptide receptor radionuclide therapy in a patient with treatment-related neuroendocrine prostate cancer with urinary retention and rectal obstruction: a case report. Yonago Acta Med. 67, 266–269 (2024).
Assadi, M. et al. 177Lu-PSMA and 177Lu-DOTATATE therapy in a patient with metastatic castration-resistant prostate cancer and neuroendocrine differentiation. Clin. Nucl. Med. 44, 978–980 (2019).
Nagasaki, S. et al. Immunohistochemical analysis of gastrin-releasing peptide receptor (GRPR) and possible regulation by estrogen receptor βcx in human prostate carcinoma. Neoplasma 59, 224–232 (2012).
Beer, M. et al. Profiling gastrin-releasing peptide receptor in prostate tissues: clinical implications and molecular correlates. Prostate 72, 318–325 (2012).
Zhang, H. et al. Synthesis and evaluation of bombesin derivatives on the basis of pan-bombesin peptides labeled with indium-111, lutetium-177, and yttrium-90 for targeting bombesin receptor-expressing tumors. Cancer Res. 64, 6707–6715 (2004).
García Garayoa, E. et al. New [99mTc]bombesin analogues with improved biodistribution for targeting gastrin releasing-peptide receptor-positive tumors. Q. J. Nucl. Med. Mol. Imaging 51, 42–50 (2007).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT06379217 (2025).
Greifenstein, L. et al. From automated synthesis to in vivo application in multiple types of cancer-clinical results with [68Ga]Ga-DATA5m.SA.FAPi. Pharmaceuticals 15, 1000 (2022).
Vlachostergios, P. J., Karathanasis, A. & Tzortzis, V. Expression of fibroblast activation protein is enriched in neuroendocrine prostate cancer and predicts worse survival. Genes 13, 135 (2022).
Hintz, H. M. et al. Imaging fibroblast activation protein alpha improves diagnosis of metastatic prostate cancer with positron emission tomography. Clin. Cancer Res. 26, 4882–4891 (2020).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05963386 (2024).
Wen, X. et al. Evans blue-modified radiolabeled fibroblast activation protein inhibitor as long-acting cancer therapeutics. Theranostics 12, 422–433 (2022).
Sidrak, M. M. A. et al. Fibroblast activation protein inhibitor (FAPI)-based theranostics — where we are at and where we are heading: a systematic review. Int. J. Mol. Sci. 24, 3863 (2023).
Laudicella, R. et al. Preliminary findings of the role of FAPi in prostate cancer theranostics. Diagnostics 13, 1175 (2023).
Vargas, H. A. et al. Bone metastases in castration-resistant prostate cancer: associations between morphologic CT patterns, glycolytic activity, and androgen receptor expression on PET and overall survival. Radiology 271, 220–229 (2014).
Fox, J. J. et al. Positron emission tomography/computed tomography-based assessments of androgen receptor expression and glycolytic activity as a prognostic biomarker for metastatic castration-resistant prostate cancer. JAMA Oncol. 4, 217–224 (2018).
Bobba, K. N. et al. Development of CD46 targeted alpha theranostics in prostate cancer using 134Ce/225Ac-Macropa-PEG4-YS5. Theranostics 14, 1344–1360 (2024).
Li, J. et al. CD46 targeted 212Pb alpha particle radioimmunotherapy for prostate cancer treatment. J. Exp. Clin. Cancer Res. 42, 61 (2023).
Dubrovska, A. et al. CXCR4 expression in prostate cancer progenitor cells. PLoS ONE 7, e31226 (2012).
Morgat, C. et al. Neurotensin receptor-1 expression in human prostate cancer: a pilot study on primary tumors and lymph node metastases. Int. J. Mol. Sci. 20, 1721 (2019).
Skovgaard, D., Persson, M. & Kjaer, A. PET imaging of urokinase-type plasminogen activator receptor (uPAR) in prostate cancer: current status and future perspectives. Clin. Transl. Imaging 4, 457–465 (2016).
Yu, S. et al. The therapeutic efficacy of I131-PSCA-mAb in orthotopic mouse models of prostate cancer. Eur. J. Med. Res. 18, 56 (2013).
Bäck, T. A. et al. Targeted alpha therapy with astatine-211-labeled anti-PSCA A11 minibody shows antitumor efficacy in prostate cancer xenografts and bone microtumors. EJNMMI Res. 10, 10 (2020).
Korsen, J. A. et al. Delta-like ligand 3-targeted radioimmunotherapy for neuroendocrine prostate cancer. Proc. Natl Acad. Sci. USA 119, e2203820119 (2022).
Akashi, T. et al. Chemokine receptor CXCR4 expression and prognosis in patients with metastatic prostate cancer. Cancer Sci. 99, 539–542 (2008).
Rebello, R. J. et al. Prostate cancer. Nat. Rev. Dis. Prim. 7, 9 (2021).
Wang, J. H. & Kiess, A. P. PSMA-targeted therapy for non-prostate cancers. Front. Oncol. 13, 1220586 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05867615 (2024).
Pomykala, K. L. et al. Next generation radiotheranostics promoting precision medicine. Ann. Oncol. 34, 507–519 (2023).
Pillow, T. H. et al. Antibody conjugation of a chimeric BET degrader enables in vivo activity. ChemMedChem 15, 17–25 (2020).
Ostuni, E. & Taylor, M. R. G. Commercial and business aspects of alpha radioligand therapeutics. Front. Med. 9, 1070497 (2022).
Fu, Z., Li, S., Han, S., Shi, C. & Zhang, Y. Antibody drug conjugate: the ‘biological missile’ for targeted cancer therapy. Signal. Transduct. Target. Ther. 7, 93 (2022).
Sheyi, R., de la Torre, B. G. & Albericio, F. Linkers: an assurance for controlled delivery of antibody-drug conjugate. Pharmaceutics 14, 396 (2022).
Bargh, J. D., Isidro-Llobet, A., Parker, J. S. & Spring, D. R. Cleavable linkers in antibody-drug conjugates. Chem. Soc. Rev. 48, 4361–4374 (2019).
Diamantis, N. & Banerji, U. Antibody-drug conjugates — an emerging class of cancer treatment. Br. J. Cancer 114, 362–367 (2016).
McCombs, J. R. & Owen, S. C. Antibody drug conjugates: design and selection of linker, payload and conjugation chemistry. AAPS J. 17, 339–351 (2015).
Dan, N. et al. Antibody-drug conjugates for cancer therapy: chemistry to clinical implications. Pharmaceuticals 11, 32 (2018).
Beck, A., Goetsch, L., Dumontet, C. & Corvaïa, N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat. Rev. Drug Discov. 16, 315–337 (2017).
Petrylak, D. P. et al. PSMA ADC monotherapy in patients with progressive metastatic castration-resistant prostate cancer following abiraterone and/or enzalutamide: efficacy and safety in open-label single-arm phase 2 study. Prostate 80, 99–108 (2020).
Milowsky, M. I. et al. Phase 1/2 multiple ascending dose trial of the prostate-specific membrane antigen-targeted antibody drug conjugate MLN2704 in metastatic castration-resistant prostate cancer. Urol. Oncol. 34, 530.e15–530.e21 (2016).
Li, C. et al. 718 AMG 509, a STEAP1 x CD3 bispecific XmAb® 2+1 immune therapy, exhibits avidity-driven binding and preferential killing of high STEAP1-expressing prostate and Ewing sarcoma cancer cells. J. Immunother. Cancer 8, A430 (2020).
Danila, D. C. et al. Phase I study of DSTP3086S, an antibody-drug conjugate targeting six-transmembrane epithelial antigen of prostate 1, in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 37, 3518–3527 (2019).
Sperger, J. M. et al. Expression and therapeutic targeting of TROP-2 in treatment-resistant prostate cancer. Clin. Cancer Res. 29, 2324–2335 (2023).
Bardia, A. et al. Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer. N. Engl. J. Med. 380, 741–751 (2019).
Rugo, H. S. et al. Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer (TROPiCS-02): a randomised, open-label, multicentre, phase 3 trial. Lancet 402, 1423–1433 (2023).
Tagawa, S. T. et al. TROPHY-U-01: a phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors. J. Clin. Oncol. 39, 2474–2485 (2021).
Bardia, A. et al. Sacituzumab govitecan, a Trop-2-directed antibody-drug conjugate, for patients with epithelial cancer: final safety and efficacy results from the phase I/II IMMU-132-01 basket trial. Ann. Oncol. 32, 746–756 (2021).
Oaknin, A. et al. 714MO Datopotamab deruxtecan (Dato-DXd) in patients with endometrial (EC) or ovarian cancer (OC): results from the phase II TROPION-PanTumor03 study. Ann. Oncol. 35, S547–S548 (2024).
TROPION-PanTumor03: phase 2, multicenter study of datopotamab deruxtecan (Dato-DXd) as monotherapy and in combination with anticancer agents in patients (pts) with advanced/metastatic solid tumors. J. Clin. Oncol. https://ascopubs.org/doi/10.1200/JCO.2023.41.16_suppl.TPS3153 (2023).
SKB264 (TROP2-ADC) for the treatment of patients with advanced NSCLC: efficacy and safety data from a phase 2 study. J. Clin. Oncol. https://ascopubs.org/doi/10.1200/JCO.2023.41.16_suppl.9114 (2023).
Cardone, J., Le Friec, G. & Kemper, C. CD46 in innate and adaptive immunity: an update. Clin. Exp. Immunol. 164, 301–311 (2011).
Aggarwal, R. R. et al. Phase 1a/1b study of FOR46, an antibody drug conjugate (ADC), targeting CD46 in metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 40, 3001 (2022).
Puca, L. et al. Delta-like protein 3 expression and therapeutic targeting in neuroendocrine prostate cancer. Sci. Transl. Med. 11, eaav0891 (2019).
Ahn, M.-J. et al. Tarlatamab for patients with previously treated small-cell lung cancer. N. Engl. J. Med. 389, 2063–2075 (2023).
US Food and Drug Administration. FDA grants accelerated approval to tarlatamab-dlle for extensive-stage small cell lung cancer. FDA https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-tarlatamab-dlle-extensive-stage-small-cell-lung-cancer (2024).
Rudin, C. M. et al. Emerging therapies targeting the delta-like ligand 3 (DLL3) in small cell lung cancer. J. Hematol. Oncol. 16, 66 (2023).
Mansfield, A. S. et al. A phase I/II study of rovalpituzumab tesirine in delta-like 3-expressing advanced solid tumors. NPJ Precis. Oncol. 5, 74 (2021).
Shenderov, E. et al. Neoadjuvant enoblituzumab in localized prostate cancer: a single-arm, phase 2 trial. Nat. Med. 29, 888–897 (2023).
Guo, C. et al. B7-H3 as a therapeutic target in advanced prostate cancer. Eur. Urol. 83, 224–238 (2023).
Doi, T. et al. 453O DS-7300 (B7-H3 DXd antibody-drug conjugate [ADC]) shows durable antitumor activity in advanced solid tumors: extended follow-up of a phase I/II study. Ann. Oncol. 33, S744–S745 (2022).
de Bono, J. S. et al. 1654P TAMARACK: randomized phase II trial of the B7-H3 targeting antibody drug conjugate (ADC) vobramitamab duocarmazine (vobra duo) in metastatic castration-resistant prostate cancer (mCRPC). Ann. Oncol. 35, S996–S997 (2024).
Monteiro, M. R. et al. Antibody-drug conjugates in breast cancer: a comprehensive review of how to selectively deliver payloads. Breast Cancer 16, 51–70 (2024).
Autio, K. A., Boni, V., Humphrey, R. W. & Naing, A. Probody therapeutics: an emerging class of therapies designed to enhance on-target effects with reduced off-tumor toxicity for use in immuno-oncology. Clin. Cancer Res. 26, 984–989 (2020).
Singh, S. et al. Nonclinical efficacy and safety of CX-2029, an anti-CD71 probody-drug conjugate. Mol. Cancer Ther. 21, 1326–1336 (2022).
Fang, S. et al. Design and characterization of immune-stimulating imidazo[4,5-c]quinoline antibody-drug conjugates. Mol. Pharm. 19, 3228–3241 (2022).
Zhou, Z. Z. et al. A dual-payload antibody-drug conjugate targeting CD276/B7-H3 elicits cytotoxicity and immune activation in triple-negative breast cancer. Cancer Res. 84, 3848–3863 (2024).
Ma, X. et al. Advances in research based on antibody-cell conjugation. Front. Immunol. 14, 1310130 (2023).
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).
Antonarakis, E. S. et al. Pembrolizumab III KEYLYNK-010 plus olaparib for patients with previously treated and biomarker-unselected metastatic castration-resistant prostate cancer: the randomized, open-label, phase trial. J. Clin. Oncol. 41, 3839–3850 (2023).
Sharma, P. et al. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the checkmate 650 trial. Cancer Cell 38, 489–499.e3 (2020).
Powles, T. et al. Atezolizumab with enzalutamide versus enzalutamide alone in metastatic castration-resistant prostate cancer: a randomized phase 3 trial. Nat. Med. 28, 144–153 (2022).
Koinis, F. et al. Myeloid-derived suppressor cells in prostate cancer: present knowledge and future perspectives. Cells 11, 20 (2021).
Guo, C. et al. Targeting myeloid chemotaxis to reverse prostate cancer therapy resistance. Nature 623, 1053–1061 (2023).
Omer, M. H. et al. Bispecific antibodies in hematological malignancies: a scoping review. Cancers 15, 4550 (2023).
Schram, A. M. et al. Zenocutuzumab, a HER2xHER3 bispecific antibody, is effective therapy for tumors driven by NRG1 gene rearrangements. Cancer Discov. 12, 1233–1247 (2022).
Lee, S. C. et al. A PSMA-targeted bispecific antibody for prostate cancer driven by a small-molecule targeting ligand. Sci. Adv. 7, eabi8193 (2021).
Hummel, H.-D. et al. Pasotuxizumab, a BiTE® immune therapy for castration-resistant prostate cancer: phase I, dose-escalation study findings. Immunotherapy 13, 125–141 (2021).
Lim, E. A. et al. Phase 1 study of safety and preliminary clinical activity of JNJ-63898081, a PSMA and CD3 bispecific antibody, for metastatic castration-resistant prostate cancer. Clin. Genitourin. Cancer 21, 366–375 (2023).
Dorff, T. et al. A phase I study of acapatamab, a half-life extended, PSMA-targeting bispecific T-cell engager for metastatic castration-resistant prostate cancer. Clin. Cancer Res. 30, 1488–1500 (2024).
Kelly, W. K. et al. Xaluritamig, a STEAP1 × CD3 XmAb 2+1 immune therapy for metastatic castration-resistant prostate cancer: results from dose exploration in a first-in-human study. Cancer Discov. 14, 76–89 (2024).
Wermke, M. et al. Phase I trial of the DLL3/CD3 bispecific T-cell engager BI 764532 in DLL3-positive small-cell lung cancer and neuroendocrine carcinomas. Future Oncol. 18, 2639–2649 (2022).
Beltran, H. et al. Interim results from a phase 1/2 study of HPN328, a tri-specific, half-life (T1/2) extended DLL3-targeting T-cell engager, in patients (pts) with neuroendocrine prostate cancer (NEPC) and other neuroendocrine neoplasms (NEN). J. Clin. Oncol. 42, 121 (2024).
Aggarwal, R. R. et al. Phase 1b study of tarlatamab in de novo or treatment-emergent neuroendocrine prostate cancer (NEPC). J. Clin. Oncol. 42, 5012 (2024).
Beltran, H. et al. Updated results from a phase 1/2 study of HPN328, a tri-specific, half-life (T1/2) extended DLL3-targeting T-cell engager in patients (pts) with small cell lung cancer (SCLC) and other neuroendocrine cancers (NEC). J. Clin. Oncol. 42, 8090 (2024).
Mikami, H. et al. Engineering CD3/CD137 dual specificity into a DLL3-targeted T-cell engager enhances T-cell infiltration and efficacy against small-cell lung cancer. Cancer Immunol. Res. 12, 719–730 (2024).
Dutcher, J. P. et al. High dose interleukin-2 (Aldesleukin) — expert consensus on best management practices — 2014. J. Immunother. Cancer 2, 26 (2014).
Haber, L. et al. Generation of T-cell-redirecting bispecific antibodies with differentiated profiles of cytokine release and biodistribution by CD3 affinity tuning. Sci. Rep. 11, 14397 (2021).
Geiger, M. et al. Protease-activation using anti-idiotypic masks enables tumor specificity of a folate receptor 1-T cell bispecific antibody. Nat. Commun. 11, 3196 (2020).
Hackenbruch, C. et al. Clinical development of the bispecific PSMAxCD3 antibody CC-1. J. Clin. Oncol. https://ascopubs.org/doi/10.1200/JCO.2023.41.16_suppl.2534 (2023).
Mehra, N. et al. Phase I/II trial of LAVA-1207, a novel bispecific gamma-delta T-cell engager alone, or with low dose IL-2 or pembrolizumab, in metastatic castration resistant prostate cancer (mCRPC). J. Clin. Oncol. 42, TPS2672 (2024).
Chen, T. T. Conditionally active T cell engagers for the treatment of solid tumors: rationale and clinical development. Expert. Opin. Biol. Ther. 22, 955–963 (2022).
Baeuerle, P. A. & Wesche, H. T-cell-engaging antibodies for the treatment of solid tumors: challenges and opportunities. Curr. Opin. Oncol. 34, 552–558 (2022).
Nathan, P. et al. Overall survival benefit with tebentafusp in metastatic uveal melanoma. N. Engl. J. Med. 385, 1196–1206 (2021).
Hsiue, E. H.-C. et al. Targeting a neoantigen derived from a common TP53 mutation. Science 371, eabc8697 (2021).
van de Donk, N. W. C. J. & Zweegman, S. T-cell-engaging bispecific antibodies in cancer. Lancet 402, 142–158 (2023).
You, G. et al. B7-H3×4-1BB bispecific antibody augments antitumor immunity by enhancing terminally differentiated CD8+ tumor-infiltrating lymphocytes. Sci. Adv. 7, eaax3160 (2021).
Hashimoto, K. CD137 as an attractive T cell co-stimulatory target in the TNFRSF for immuno-oncology drug development. Cancers 13, 2288 (2021).
Zhu, H. et al. 4-1BBL has a possible role in mediating castration-resistant conversion of prostate cancer via up-regulation of androgen receptor. J. Cancer 10, 2464–2471 (2019).
Zorko, N. A. et al. Natural killer cell infiltration in prostate cancers predict improved patient outcomes. Prostate Cancer Prostatic Dis. 28, 129–137 (2025).
Zorko, N. et al. Effect of camelid B7-H3 tri-specific killer engagers on natural killer cells in patients with prostate cancer. J. Clin. Oncol. 42, 155 (2024).
Sterner, R. C. & Sterner, R. M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11, 69 (2021).
Thompson-Elliott, B., Johnson, R. & Khan, S. A. Alterations in TGFβ signaling during prostate cancer progression. Am. J. Clin. Exp. Urol. 9, 318–328 (2021).
Jin, C. et al. Cellular senescence in metastatic prostate cancer: a therapeutic opportunity or challenge (Review). Mol. Med. Rep. 30, 162 (2024).
Narayan, V. et al. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat. Med. 28, 724–734 (2022).
Amor, C. et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583, 127–132 (2020).
Zhang, Y. et al. A signature for pan-cancer prognosis based on neutrophil extracellular traps. J. Immunother. Cancer 10, e004210 (2022).
McKean, M. et al. Safety and early efficacy results from a phase 1, multicenter trial of PSMA-targeted armored CAR T cells in patients with advanced mCRPC. J. Clin. Oncol. 40, 94 (2022).
Stein, M. N. et al. PSCA-targeted BPX-601 CAR T cells with pharmacological activation by rimiducid in metastatic pancreatic and prostate cancer: a phase 1 dose escalation trial. Nat. Commun. 15, 10743 (2024).
Sloas, C., Gill, S. & Klichinsky, M. Engineered CAR-macrophages as adoptive immunotherapies for solid tumors. Front. Immunol. 12, 783305 (2021).
Zhang, L. et al. Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions. J. Hematol. Oncol. 13, 153 (2020).
Page, A., Chuvin, N., Valladeau-Guilemond, J. & Depil, S. Development of NK cell-based cancer immunotherapies through receptor engineering. Cell Mol. Immunol. 21, 315–331 (2024).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT03692663 (2022).
Chen, H. et al. Delivery of CD47 blocker SIRPα-Fc by CAR-T cells enhances antitumor efficacy. J. Immunother. Cancer 10, e003737 (2022).
Hiltensperger, M. & Krackhardt, A. M. Current and future concepts for the generation and application of genetically engineered CAR-T and TCR-T cells. Front. Immunol. 14, 1121030 (2023).
Tousley, A. M. et al. Co-opting signalling molecules enables logic-gated control of CAR T cells. Nature 615, 507–516 (2023).
Kloss, C. C., Condomines, M., Cartellieri, M., Bachmann, M. & Sadelain, M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 31, 71–75 (2013).
Kantoff, P. W. et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 363, 411–422 (2010).
Fletcher, E. et al. Abstract 4160: promoting immunogenicity of synthetic long peptide vaccines based on in vivo IgG complex formation: preclinical evaluation and clinical entry of the TET platform. Cancer Res. 82, 4160 (2022).
Autio, K. A. et al. First-in-human, phase 1 study of PF-06753512, a vaccine-based immunotherapy regimen (VBIR), in non-metastatic hormone-sensitive biochemical recurrence and metastatic castration-resistant prostate cancer (mCRPC). J. Immunother. Cancer 11, e005702 (2023).
Stenzl, A. et al. Results of the randomized, placebo-controlled phase I/IIB trial of CV9104, an mRNA based cancer immunotherapy, in patients with metastatic castration-resistant prostate cancer (mCRPC). Ann. Oncol. 28, v408–v409 (2017).
Li, X.-Y., Li, T., Li, X.-J., Wang, J.-N. & Chen, Z. TSG-6 induces apoptosis of human hypertrophic scar fibroblasts via activation of the Fas/FasL signalling pathway. Folia Biol. 64, 173–181 (2018).
Peel, R. et al. Zinc in preventing the progression of pre-diabetes (ZIPPeD Study) — study protocol for a randomised placebo-controlled trial in Australia. Trials 20, 219 (2019).
Kübler, H. et al. Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/IIa study. J. Immunother. Cancer 3, 26 (2015).
Bissett, S. M., Presseau, J., Rapley, T. & Preshaw, P. M. Uptake of best practice recommendations in the management of patients with diabetes and periodontitis: a cross-sectional survey of dental clinicians. Br. Dent. J. 226, 131–137 (2019).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04382898 (2024).
Simmons, S. J. et al. GM-CSF as a systemic adjuvant in a phase II prostate cancer vaccine trial. Prostate 39, 291–297 (1999).
Waeckerle-Men, Y. et al. Dendritic cell-based multi-epitope immunotherapy of hormone-refractory prostate carcinoma. Cancer Immunol. Immunother. 55, 1524–1533 (2006).
Yeh, K. H., Yeh, S. H., Chang, Y. S. & Cheng, A. L. Minimal toxicity to myeloid progenitor cells of weekly 24-hr infusion of high-dose 5-fluorouracil: direct evidence from colony forming unit-granulocyte and monocyte (CFU-GM) clonogenic assay. Pharmacol. Toxicol. 86, 122–124 (2000).
Dixon, K. J., Wu, J. & Walcheck, B. Engineering anti-tumor monoclonal antibodies and fc receptors to enhance ADCC by human NK cells. Cancers 13, 312 (2021).
Tatard, V. M., Xiang, C., Biegel, J. A. & Dahmane, N. ZNF238 is expressed in postmitotic brain cells and inhibits brain tumor growth. Cancer Res. 70, 1236–1246 (2010).
Zhou, J.-K. et al. ROR1 expression as a biomarker for predicting prognosis in patients with colorectal cancer. Oncotarget 8, 32864–32872 (2017).
Ma, F. et al. Autocrine canonical Wnt signaling primes noncanonical signaling through ROR1 in metastatic castration-resistant prostate cancer. Cancer Res. 82, 1518–1533 (2022).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05156905 (2024).
de Bono, J. S. et al. 687P A CRUK phase I/IIA, first in human dose-escalation and expansion trial of HMBD-001 (an anti-HER3 antibody) in patients with advanced HER3 positive solid tumours. Ann. Oncol. 34, S479–S480 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05057013 (2024).
Atiq, M. O. et al. Combining IL-12 immunocytokine (M9241) with docetaxel in metastatic prostate cancer: a phase I study. J. Clin. Oncol. 40, e17033 (2022).
SunHo announces first patient dosed in phase I/II clinical trial of a potential first-in-class immunocytokine IBB0979. PR Newswire https://www.prnewswire.com/news-releases/sunho-announces-first-patient-dosed-in-phase-iii-clinical-trial-of-a-potential-first-in-class-immunocytokine-ibb0979-301883084.html (21 July 2023).
Mahalingam, D. et al. First-in-human phase I dose escalation trial of the first-in-class tumor microenvironment modulator VT1021 in advanced solid tumors. Commun. Med. 4, 10 (2024).
Bourque, P. R., Brooks, J., McCudden, C. R., Warman-Chardon, J. & Breiner, A. Age matters: impact of data-driven CSF protein upper reference limits in Guillain-Barré syndrome. Neurol. Neuroimmunol. Neuroinflamm. 6, e576 (2019).
Watt, M. J. et al. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer. Sci. Transl. Med. 11, eaau5758 (2019).
Zhou, X. et al. CD36: the bridge between lipids and tumors. Molecules 29, 531 (2024).
Guerrero-Rodríguez, S. L., Mata-Cruz, C., Pérez-Tapia, S. M. & Velasco-Velázquez, M. A. Role of CD36 in cancer progression, stemness, and targeting. Front. Cell Dev. Biol. 10, 1079076 (2022).
Carpi, S. et al. AM251 induces apoptosis and G2/M cell cycle arrest in A375 human melanoma cells. Anticancer Drugs 26, 754–762 (2015).
Zhang, B. et al. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature 537, 553–557 (2016).
Tannock, I. F. et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. Med. 351, 1502–1512 (2004).
Cohen, L., Assaraf, Y. G. & Livney, Y. D. Novel selectively targeted multifunctional nanostructured lipid carriers for prostate cancer treatment. Pharmaceutics 14, 88 (2021).
Nordquist, L. T. et al. Final results from a phase I clinical trial evaluating the safety, immunogenicity, and anti-tumor activity of SNS-301 in men with biochemically relapsed prostate cancer. Ann. Oncol. 29, viii136 (2018).
Moorthy, G. et al. Comprehensive clinical pharmacology characterization of AZD4635 in healthy participants to support dosing considerations. Br. J. Clin. Pharmacol. 89, 2775–2787 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT02740985 (2023).
Wang, B. et al. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct. Target. Ther. 9, 1–65 (2024).
Govindarajan, B. et al. Adaptor proteins mediate CXCR4 and PI4KA crosstalk in prostate cancer cells and the significance of PI4KA in bone tumor growth Sci. Rep. 13, 20634 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04227275 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT03245736 (2021).
Acknowledgements
The authors apologize to the many researchers whose work we were unable to cite because of space limitations. We thank members of the Beltran and de Bono lab for discussion of topics relevant to this Review. V.B.V. is supported by the DoD PCRP Early Investigator Research Award (W81XWH2210197), the National Cancer Center Postdoctoral Fellowship Award and the Prostate Cancer Foundation Young Investigator Award (23YOUN15). H.B. is supported by the Prostate Cancer Foundation, the DoD PCRP (W81XWH-17-1-0653) and the NIH/NCI (R37CA241486-01A1, DF/HCC SPORE P50 CA272390-01, WCM SPORE P50 CA211024-01A1). The de Bono laboratory acknowledges funding from Cancer Research UK, Prostate Cancer UK, the Prostate Cancer Foundation, the Medical Research Council and the US Department of Defense.
Author information
Authors and Affiliations
Contributions
The authors contributed equally to all aspects of the article.
Corresponding authors
Ethics declarations
Competing interests
H.B. has served as consultant/advisory board member for Janssen, AstraZeneca, Merck, Pfizer, Amgen, Bayer, Daiichi Sankyo and Novartis, and has received research funding (to their institution) from Janssen, Bristol Myers Squibb, Circle Pharma, Daiichi Sankyo and Novartis. J.d.B. has served as an adviser for Daiichi Sankyo, AstraZeneca, MSD, Pfizer, Amgen, Bayer, Merck Serono, Janssen, GSK, Macrogenics, Actinium Pharma, Astellas, Amunix and Sanofi-Aventis. The other authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Urology thanks Lawrence True, Naga Vara Kishore Pillarsetty 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.
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.
About this article
Cite this article
Boixareu, C., Taha, T., Venkadakrishnan, V.B. et al. Targeting the tumour cell surface in advanced prostate cancer. Nat Rev Urol 22, 569–589 (2025). https://doi.org/10.1038/s41585-025-01014-w
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s41585-025-01014-w
This article is cited by
-
Recent advances and future prospects of immunotherapeutic approaches in prostate cancer
Journal of Translational Medicine (2026)
-
State of the art — biomarkers in advanced prostate cancer
Nature Reviews Urology (2025)


