1
|
Cerra B, Gioiello A. Discovery and development of steroidal enzyme inhibitors as anti-cancer drugs: state-of-the-art and future perspectives. J Enzyme Inhib Med Chem 2025; 40:2483818. [PMID: 40172115 PMCID: PMC11967001 DOI: 10.1080/14756366.2025.2483818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025] Open
Abstract
Steroidal compounds have emerged as effective therapeutic agents in oncology. Beyond natural-occurring and synthetic steroids that act as cytotoxic anti-tumoral agents, steroidal derivatives can be designed to mime the endogenous substrates of key metabolic enzymes in steroidogenesis, thus reducing the circulating levels of relevant oestrogenic and androgenic hormones responsible for cancer survival and proliferation. Therefore, enzyme inhibition represents an intriguing endocrine approach for the treatment of hormone-dependent tumours, such as breast and prostate cancer, with well-known approved drugs and several pre-clinical and clinical candidates under investigation. This review summarises the key advancements over the past decade (2014-2024) in the development of steroidal enzyme inhibitors endowed with anticancer activity, illustrating their mechanisms of action, therapeutic potential, drug design approaches, and current clinical applications. Furthermore, we discuss challenges related to drug resistance, off-target effects, and future strategies to optimise their efficacy in oncology.
Collapse
Affiliation(s)
- Bruno Cerra
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Antimo Gioiello
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
2
|
Wu Z, Zhang J, Li L, Wang Z, Yang C. Biomarkers in metastatic castration-resistant prostate cancer for efficiency of immune checkpoint inhibitors. Ann Med 2025; 57:2426755. [PMID: 39895499 PMCID: PMC11792157 DOI: 10.1080/07853890.2024.2426755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/29/2024] [Accepted: 09/05/2024] [Indexed: 02/04/2025] Open
Abstract
Almost all patients with prostate cancer progress to metastatic castration-resistant prostate cancer (mCRPC) despite initial responses. In cases where traditional first-line treatments prove ineffective, the potential of immune checkpoint blockade (ICB) therapy emerges as a promising approach for managing mCRPC. However, while immune checkpoint inhibitor monotherapy or combination therapy targeting cytotoxic T lymphocyte antigen 4 (CTLA-4) and/or programmed cell death-1 (PD-1)/PD-1 ligand 1 (PD-L1) axis has been regarded as the standard therapy in many solid tumours, mCRPC as 'cold' tumours are considered to be relatively resistant to ICB treatment. Encouragingly, recent evidence suggests that ICB therapy may be particularly beneficial in specific subgroups of patients with high PD-L1 tumour expression, high tumour mutational burden or high tumour microsatellite instability/mismatch repair deficiency. Better understanding of these predictive biomarkers could screen which patients are most likely to benefit. This review article examines biomarkers for screening patients potentially effective in immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Zixi Wu
- Department of Urology, Tongji Hospital Affiliated Tongji Medical College of Huazhong University of Science and Technology (HUST), Wuhan, China
- Huangshi Hubei Medical Group of Maternal and Child Health Hospital, Hubei, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital Affiliated Tongji Medical College of Huazhong University of Science and Technology (HUST), Wuhan, China
- Huangshi Hubei Medical Group of Maternal and Child Health Hospital, Hubei, China
| | - Le Li
- Department of Urology, Tongji Hospital Affiliated Tongji Medical College of Huazhong University of Science and Technology (HUST), Wuhan, China
- Huangshi Hubei Medical Group of Maternal and Child Health Hospital, Hubei, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital Affiliated Tongji Medical College of Huazhong University of Science and Technology (HUST), Wuhan, China
- Huangshi Hubei Medical Group of Maternal and Child Health Hospital, Hubei, China
| | - Chunguang Yang
- Department of Urology, Tongji Hospital Affiliated Tongji Medical College of Huazhong University of Science and Technology (HUST), Wuhan, China
- Huangshi Hubei Medical Group of Maternal and Child Health Hospital, Hubei, China
| |
Collapse
|
3
|
Abida W, Beltran H, Raychaudhuri R. State of the Art: Personalizing Treatment for Patients With Metastatic Castration-Resistant Prostate Cancer. Am Soc Clin Oncol Educ Book 2025; 45:e473636. [PMID: 40112242 DOI: 10.1200/edbk-25-473636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Until recently, the treatment of metastatic castration-resistant prostate cancer (mCRPC) relied exclusively on hormonal therapies and taxane chemotherapy. The advent of modern molecular profiling methods applied in the clinic, namely, next-generation sequencing and advanced positron emission tomography (PET) imaging, has allowed for the development of biomarker-driven therapeutics including anti-PD-L1 therapy for microsatellite instability-high or tumor mutation burden-high disease, poly(ADP-ribose) polymerase (PARP) inhibitors for patients with DNA damage repair mutations, and lutetium 177 vipivotide tetraxetan (177Lu-PSMA-617) for patients with prostate-specific membrane antigen (PSMA) PET-avid disease. While these targeted therapies have improved outcomes, there is an opportunity to refine biomarkers to optimize patient selection, understand resistance, and develop novel combination strategies. In addition, studies in the laboratory and in patient-derived samples have shown that a subset of mCRPC tumors lose expression of common prostate cancer markers such as prostate-specific antigen and PSMA because of lineage plasticity and the development of non-androgen receptor (AR)-driven disease. Non-AR-driven prostate cancer has been associated with aggressive behavior and poor prognosis, including in some cases histologic transformation to a poorly differentiated neuroendocrine prostate cancer (NEPC). The clinical management of NEPC typically follows the treatment paradigm for small cell lung cancer and increasingly relies on genomic and phenotypic characterization of disease, including loss of tumor suppressors and expression of cell surface markers such as DLL3. Therefore, both genomic subtyping and phenotypic subtyping are important to consider and can guide the clinical management of patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruben Raychaudhuri
- University of Washington and the Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
4
|
Clements AN, Casillas AL, Flores CE, Liou H, Toth RK, Chauhan SS, Sutterby K, Deshmukh SK, Wu S, Xiu J, Farrell A, Radovich M, Nabhan C, Heath EI, McKay RR, Subah N, Centuori S, Weeler TJ, Cress AE, Rogers GC, Wilson JE, Recio-Boiles A, Warfel NA. Inhibition of PIM Kinase in Tumor-Associated Macrophages Suppresses Inflammasome Activation and Sensitizes Prostate Cancer to Immunotherapy. Cancer Immunol Res 2025; 13:633-645. [PMID: 39982419 DOI: 10.1158/2326-6066.cir-24-0591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/26/2024] [Accepted: 02/20/2025] [Indexed: 02/22/2025]
Abstract
Immune checkpoint inhibitors (ICI) have changed the treatment paradigm for many cancers but have not shown benefit in prostate cancer. Chronic inflammation contributes to the immunosuppressive prostate tumor microenvironment and is associated with poor response to ICIs. The primary source of inflammatory cytokine production is the inflammasome. In this study, we identify the proviral integration site for Moloney murine leukemia virus (PIM) kinases as regulators of inflammasome activation in tumor-associated macrophages (TAM). The analysis of clinical data from a cohort of patients with treatment-naïve, hormone-responsive prostate cancer revealed that tumors from patients with high PIM1/2/3 displayed an immunosuppressive tumor microenvironment characterized by high inflammation and a high density of repressive immune cells, most notably TAMs. Macrophage-specific knockout of PIM reduced tumor growth in syngeneic models of prostate cancer. Transcriptional analyses indicated that eliminating PIM from macrophages enhanced the adaptive immune response and increased cytotoxic immune cells. Combined treatment with PIM inhibitors and ICIs synergistically reduced tumor growth. Immune profiling revealed that PIM inhibitors sensitized prostate cancer tumors to ICIs by increasing tumor suppressive TAMs and increasing the activation of cytotoxic T cells. Our data implicate macrophage PIM as a driver of inflammation that limits ICI potency and provide preclinical evidence that PIM inhibitors are an effective strategy to improve the ICI efficacy in prostate cancer.
Collapse
Affiliation(s)
- Amber N Clements
- Cancer Biology Graduate Program, University of Arizona, Tucson, Arizona
| | - Andrea L Casillas
- Cancer Biology Graduate Program, University of Arizona, Tucson, Arizona
| | - Caitlyn E Flores
- Cancer Biology Graduate Program, University of Arizona, Tucson, Arizona
| | - Hope Liou
- Cancer Biology Graduate Program, University of Arizona, Tucson, Arizona
| | - Rachel K Toth
- University of Arizona Cancer Center, Tucson, Arizona
| | | | - Kai Sutterby
- University of Arizona Cancer Center, Tucson, Arizona
| | | | - Sharon Wu
- Caris Life Sciences, Phoenix, Arizona
| | | | | | | | | | - Elisabeth I Heath
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Rana R McKay
- University of California, San Diego, San Diego, California
| | - Noor Subah
- University of Arizona Health Sciences, Tucson, Arizona
| | - Sara Centuori
- University of Arizona Cancer Center, Tucson, Arizona
| | - Travis J Weeler
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Anne E Cress
- University of Arizona Cancer Center, Tucson, Arizona
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Gregory C Rogers
- University of Arizona Cancer Center, Tucson, Arizona
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Justin E Wilson
- University of Arizona Cancer Center, Tucson, Arizona
- Department of Immunobiology, University of Arizona, Tucson, Arizona
| | - Alejandro Recio-Boiles
- University of Arizona Cancer Center, Tucson, Arizona
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Noel A Warfel
- University of Arizona Cancer Center, Tucson, Arizona
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
5
|
Rifai S, Rifai A, Shi X, Khan MA, Guang W, Wang L, Tallon L, Hussain A. Genomic and transcriptomic sequencing in prostate cancer. Curr Opin Oncol 2025; 37:240-249. [PMID: 40071471 DOI: 10.1097/cco.0000000000001136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW Genomic and transcriptomic sequencing technologies have revolutionized our ability to characterize prostate cancer at the molecular level. The underlying premise of next-generation sequencing technologies and their current and evolving applications in prostate cancer management are provided in the review. RECENT FINDINGS Improved methodologies are allowing timely sequencing of the coding regions or both the coding and noncoding regions of the genome to help identify potential mutations and structural variations in the prostate cancer genome, some of which are currently also targetable therapeutically. DNA microarray- based differential gene expression has been supplanted by RNA sequencing (RNA-seq), which not only allows for more accurate quantitation but also nucleotide-level resolution to investigate the entire transcriptome, including alternative gene spliced transcripts and noncoding RNA transcripts, whose full clinical implications have yet to be fully understood and realized. Gene classifier platforms that predict risk of recurrence or metastasis are being incorporated into prostate cancer management algorithms. In the appropriate clinical context, not only somatic but also germline mutation testing is being recommended. SUMMARY Continued clinical integration of sequencing technologies and ongoing research will lead to improved understanding of prostate cancer biology and prostate cancer treatment.
Collapse
Affiliation(s)
- Safiullah Rifai
- University of Maryland Greenebaum Comprehensive Cancer Center
| | - Azimullah Rifai
- University of Maryland Greenebaum Comprehensive Cancer Center
| | - Xiaolei Shi
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
| | | | - Wei Guang
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
| | - Linbo Wang
- University of Maryland Greenebaum Comprehensive Cancer Center
| | | | - Arif Hussain
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
- Department of Pathology
- Depepartment of Biochemistry and Molecular Biology
- Baltimore VA Medical Center, Baltimore, Maryland USA
| |
Collapse
|
6
|
McNevin CS, Keogh A, Mohammed Nur M, McGovern B, McFadden J, Baird AM, Cadoo K, Mc Carron S, O'Brien C, Barr MP, Gray SG, Sheils O, Sutton LA, Flanagan S, Mucci LA, Stopsack KH, Finn SP. Prevalence of Mismatch Repair Deficiency in Primary Prostate Cancer in a Large Prospective Cohort. Clin Cancer Res 2025; 31:1746-1753. [PMID: 40105779 PMCID: PMC12045727 DOI: 10.1158/1078-0432.ccr-24-1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/10/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Mismatch repair (MMR) deficiency and microsatellite instability are predictive biomarkers for immunotherapy response. The best approach to identify patients with such tumors is unclear in prostate cancer. EXPERIMENTAL DESIGN This study included 1,016 men diagnosed with primary prostate cancer during prospective follow-up of the Health Professionals Follow-up Study and Physicians' Health Study. The highest-grade/index lesions from radical prostatectomy (95%) or transurethral resections of the prostate were mounted on tissue microarrays. Scoring of immunohistochemistry for the MMR proteins MLH1, MSH2, MSH6, and PMS2 required a nontumor internal positive control for designating deficiency. Validation was done on full sections and with PCR-based quantification of microsatellite repeats. RESULTS Tumor stage was predominantly pathologically localized with a full distribution of Gleason scores. MMR tumor scoring could be performed with available internal positive control tissue in 75% to 90% of cases, depending on the MMR protein. Of the 903 tumors evaluable for MSH2 protein loss, 4 tumors had loss of MSH2 (prevalence, 0.4%; 95% confidence interval, 0.2%-1.1%), and 3 of 708 evaluable tumors had concomitant loss of MSH6 (prevalence, 0.4%; 95% confidence interval, 0.1%-1.2%). No tumor had loss of MLH1 or PMS2. The four MMR-deficient cases had higher Gleason scores, and three had non-zero microsatellite repeats. CONCLUSIONS In this nationwide prospective study, MMR deficiency was rare in primary, surgically treated prostate cancer. The low prevalence and the need for an internal positive control for this assay are feasibility concerns for unselected routine immunohistochemistry-based screening for MMR deficiency on limited tissue specimens, such as prostate biopsies.
Collapse
Affiliation(s)
- Ciara S McNevin
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Anna Keogh
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | | | - Brianán McGovern
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - Julie McFadden
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Karen Cadoo
- Department of Medical Oncology, St. James's Hospital, Dublin, Ireland
| | - Sarah Mc Carron
- Cancer Molecular Diagnostics, St. James's Hospital, Dublin, Ireland
| | - Cathal O'Brien
- Cancer Molecular Diagnostics, St. James's Hospital, Dublin, Ireland
| | - Martin P Barr
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Steven G Gray
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Lesley A Sutton
- Cancer Molecular Diagnostics, St. James's Hospital, Dublin, Ireland
| | - Sinead Flanagan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Konrad H Stopsack
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology - BIPS and Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Ambrosini M, Manca P, Nasca V, Sciortino C, Ghelardi F, Seligmann JF, Taieb J, Pietrantonio F. Epidemiology, pathogenesis, biology and evolving management of MSI-H/dMMR cancers. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01015-z. [PMID: 40181086 DOI: 10.1038/s41571-025-01015-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
Deficiency in DNA mismatch repair (dMMR) is a common pathway of carcinogenesis across different tumour types and confers a characteristic microsatellite instability-high (MSI-H) molecular phenotype. The prevalence of the MSI-H/dMMR phenotype is highest in endometrial and colorectal cancers, and this phenotype is associated with a distinct tumour biology, prognosis and responsiveness to various anticancer treatments. In a minority of patients, MSI-H/dMMR cancers result from an inherited pathogenic variant in the context of Lynch syndrome, which has important implications for familial genetic screening. Whether these hereditary cancers have a different biology and clinical behaviour to their sporadic counterparts remains uncertain. Interest in this tumour molecular subtype has increased following the discovery of the high sensitivity of metastatic MSI-H/dMMR cancers to immune-checkpoint inhibitors (ICIs) in a histology-agnostic manner, which reflects the genomic hypermutation resulting from dMMR that renders these tumours highly immunogenic and immune infiltrated. This vulnerability is now also being exploited in early stage disease settings. Despite this common biological foundation, different MSI-H/dMMR cancers have histotype-specific features that correspond to their particular cell or tissue of origin, which might be associated with differences in prognosis and sensitivity to ICIs. In this Review, we provide an overview of the epidemiology, biology, pathogenesis, clinical diagnosis and treatment of MSI-H/dMMR tumours as a histology-agnostic cancer phenomenon. We also highlight peculiarities associated with specific pathogenetic alterations and histologies of MSI-H/dMMR tumours.
Collapse
Affiliation(s)
- Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Paolo Manca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vincenzo Nasca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carolina Sciortino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Ghelardi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Jenny F Seligmann
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Paris-Cité University, SIRIC CARPEM Comprehensive Cancer Center, Paris, France
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
8
|
Wang C, Han X, Kong S, Zhang S, Ning H, Wu F. Deciphering the mechanisms of PARP inhibitor resistance in prostate cancer: Implications for precision medicine. Biomed Pharmacother 2025; 185:117955. [PMID: 40086424 DOI: 10.1016/j.biopha.2025.117955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Prostate cancer is a leading malignancy among men. While early-stage prostate cancer can be effectively managed, metastatic prostate cancer remains incurable, with a median survival of 3-5 years. The primary treatment for advanced prostate cancer is androgen deprivation therapy (ADT), but resistance to ADT often leads to castrationresistant prostate cancer (CRPC), presenting a significant therapeutic challenge. The advent of precision medicine has introduced promising new treatments, including PARP inhibitors (PARPi), which target defects in DNA repair mechanisms in cancer cells. PARPi have shown efficacy in treating advanced prostate cancer, especially in patients with metastatic CRPC (mCRPC) harboring homologous recombination (HR)-associated gene mutations. Despite these advancements, resistance to PARPi remains a critical issue. Here, we explored the primary mechanisms of PARPi resistance in prostate cancer. Key resistance mechanisms include homologous recombination recovery through reverse mutations in BRCA genes, BRCA promoter demethylation, and non-degradation of mutated BRCA proteins. The tumor microenvironment and overactivation of the base excision repair pathway also play significant roles in bypassing PARPi-induced synthetic lethality. In addition, we explored the clinical implications and therapeutic strategies to overcome resistance,emphasizing the need for precision medicine approaches. Our findings highlight the need for comprehensive strategies to improve PARPi sensitivity and effectiveness,ultimately aiming to extend patient survival and improve the quality of life for those with advanced prostate cancer. As our understanding of PARPi resistance evolves, more diverse and effective individualized treatment regimens will emerge.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China
| | - Xiaoran Han
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shaoqiu Kong
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shanhua Zhang
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Hao Ning
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| | - Fei Wu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| |
Collapse
|
9
|
Kwon WA, Joung JY. Immunotherapy in Prostate Cancer: From a "Cold" Tumor to a "Hot" Prospect. Cancers (Basel) 2025; 17:1064. [PMID: 40227610 PMCID: PMC11987915 DOI: 10.3390/cancers17071064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Immunotherapy has shown limited efficacy in prostate cancer, largely due to low tumor immunogenicity, sparse tumor-infiltrating lymphocytes, and a suppressive microenvironment. Recent therapeutic strategies aim to boost immune responses and counteract immunosuppressive factors through interventions such as immune checkpoint inhibitors, immunogenic cell death-inducing therapies, and the targeted blockade of pathways like that of transforming growth factor-β. Vaccine-based approaches, potent immune adjuvants, and engineered chimeric antigen receptor (CAR) T cells are also being investigated to overcome local immune inhibitory signals. Advancements in imaging, multi-omic profiling, and liquid biopsies offer promising avenues for real-time monitoring, better patient selection, and precision treatment. This review provides an overview of the key immunosuppressive features of prostate cancer, current immunotherapeutic modalities, and emerging strategies to transform "cold" tumors into more responsive "hot" targets. By integrating these approaches, we may achieve more durable clinical benefits for patients with advanced or metastatic prostate cancer.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Jae Young Joung
- Department of Urology, Urological Cancer Center, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
10
|
Vis DJ, Palit SAL, Corradi M, Cuppen E, Mehra N, Lolkema MP, Wessels LFA, van der Heijden MS, Zwart W, Bergman AM. Whole genome sequencing of 378 prostate cancer metastases reveals tissue selectivity for mismatch deficiency with potential therapeutic implications. Genome Med 2025; 17:24. [PMID: 40114169 PMCID: PMC11927350 DOI: 10.1186/s13073-025-01445-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Survival of patients with metastatic castration-resistant prostate cancer (mCRPC) depends on the site of metastatic dissemination. METHODS Patients with mCRPC were prospectively included in the CPCT-02 metastatic site biopsy study. We evaluated whole genome sequencing (WGS) of 378 mCRPC metastases to understand the genetic traits that affect metastatic site distribution. RESULTS Our findings revealed that RB1, PIK3CA, JAK1, RNF43, and TP53 mutations are the most frequent genetic determinants associated with site selectivity for metastatic outgrowth. Furthermore, we explored mutations in the non-coding genome and found that androgen receptor (AR) chromatin binding sites implicated in metastatic prostate cancer differ in mutation frequencies between metastatic sites, converging on pathways that impact DNA repair. Notably, liver and visceral metastases have a higher tumor mutational load (TML) than bone and lymph node metastases, independent of genetic traits associated with neuroendocrine differentiation. We found that TML is strongly associated with DNA mismatch repair (MMR)-deficiency features in these organs. CONCLUSIONS Our results revealed gene mutations that are significantly associated with metastatic site selectivity and that frequencies of non-coding mutations at AR chromatin binding sites differ between metastatic sites. Immunotherapeutics are thus far unsuccessful in unselected mCRPC patients. We found a higher TML in liver and visceral metastases compared to bone and lymph node metastases. As immunotherapeutics response is associated with mutational burden, these findings may assist in selecting mCRPC patients for immunotherapy treatment based on organs affected by metastatic disease. TRIAL REGISTRATION NUMBER NCT01855477.
Collapse
Affiliation(s)
- Daniel J Vis
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Computational Cancer Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sander A L Palit
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marie Corradi
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Edwin Cuppen
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Hartwig Medical Foundation, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lodewyk F A Wessels
- Division of Computational Cancer Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Michiel S van der Heijden
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Wilbert Zwart
- Oncode Institute, Utrecht, The Netherlands.
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Andries M Bergman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Cheng PL, Wang WJ, Chuang CY, Lin CH, Huang CY, Hsiao TH, Hsu CP. Distinct genomic features and mutational signatures of nucleotide excision repair and mismatch repair in thymoma. Am J Cancer Res 2025; 15:1189-1200. [PMID: 40226447 PMCID: PMC11982733 DOI: 10.62347/zwsb8391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/05/2025] [Indexed: 04/15/2025] Open
Abstract
Thymoma is a rare malignancy with an unclear etiology of occurrence and development. We observed a higher incidence of thymoma in the Taiwanese population compared to other Western populations, suggesting the existence of different genomic features. Since most genomic studies are based on Western populations, we aimed to characterize the genomic profile of the Taiwanese population and compare it to the TCGA cohort in this study. We analyzed the genome of 47 thymoma patients using the Tumor Mutational Burden Panel to discover the genetic profile of the Taiwanese population. We also characterized the mutational signatures of these samples. Additionally, we leveraged RNA seq to estimate the gene expression profile and explored the featured pathways of thymoma in the Taiwanese population through gene set enrichment analysis.We identified several frequently mutated genes related to transcription, such as FAT1, KMT2D, and ZFHX3, as well as consensus mutational signatures associated with nucleotide excision repair (NER) and mismatch repair (MMR) deficiency. Our study also revealed increased activity of NER and MMR functions in our study cohort. Upon comparison with the TCGA cohort, we found dramatic differences in the most frequently mutated genes and mutational profiles between the Taiwanese and TCGA cohorts. Furthermore, we identified mismatch repair deficiency as a Taiwanese population-specific mutational signature with higher activity. These results highlight the distinct genomic background and molecular mechanisms of thymoma in the Taiwanese population, which may contribute to the development of new diagnostic and therapeutic strategies in the future.
Collapse
Affiliation(s)
- Po-Liang Cheng
- Department of Medical Research, Taichung Veterans General HospitalTaichung, Taiwan
| | - Wei-Jan Wang
- Department of Biological Science and Technology, China Medical UniversityTaichung, Taiwan
| | - Cheng-Yen Chuang
- Division of Thoracic Surgery, Department of Surgery, Taichung Veterans General HospitalTaichung, Taiwan
| | - Chih-Hung Lin
- Division of Thoracic Surgery, Department of Surgery, Taichung Veterans General HospitalTaichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi HospitalHualien, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and TechnologyHualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical UniversityTaichung, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General HospitalTaichung, Taiwan
- Department of Public Health, Fu Jen Catholic UniversityNew Taipei, Taiwan
- Institute of Genomics and Bioinformatics, National Chung Hsing UniversityTaichung, Taiwan
| | - Chung-Ping Hsu
- Division of Thoracic Surgery, Department of Surgery, Taichung Veterans General HospitalTaichung, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Buddhist Tzu Chi Medical Center, Hualien Tzu Chi HospitalHualien, Taiwan
| |
Collapse
|
12
|
Li S, Song K, Sun H, Tao Y, Huang A, Bhatia V, Hanratty B, Patel RA, Long HW, Morrissey C, Haffner MC, Nelson PS, Graeber TG, Lee JK. Defined cellular reprogramming of androgen receptor-active prostate cancer to neuroendocrine prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637904. [PMID: 40027790 PMCID: PMC11870442 DOI: 10.1101/2025.02.12.637904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Neuroendocrine prostate cancer (NEPC) arises primarily through neuroendocrine transdifferentiation (NEtD) as an adaptive mechanism of therapeutic resistance. Models to define the functional effects of putative drivers of this process on androgen receptor (AR) signaling and NE cancer lineage programs are lacking. We adapted a genetically defined strategy from the field of cellular reprogramming to directly convert AR-active prostate cancer (ARPC) to AR-independent NEPC using candidate factors. We delineated critical roles of the pioneer factors ASCL1 and NeuroD1 in NEtD and uncovered their abilities to silence AR expression and signaling by remodeling chromatin at the somatically acquired AR enhancer and global AR binding sites with enhancer activity. We also elucidated the dynamic temporal changes in the transcriptomic and epigenomic landscapes of cells undergoing acute lineage conversion from ARPC to NEPC which should inform future therapeutic development. Further, we distinguished the activities of ASCL1 and NeuroD1 from the inactivation of RE-1 silencing transcription factor (REST), a master suppressor of a major neuronal gene program, in establishing a NEPC lineage state and in modulating the expression of genes associated with major histocompatibility complex class I (MHC I) antigen processing and presentation. These findings provide important, clinically relevant insights into the biological processes driving NEtD of prostate cancer.
Collapse
Affiliation(s)
- Shan Li
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Kai Song
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Huiyun Sun
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Yong Tao
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Arthur Huang
- Department of Urology, University of Washington, Seattle, WA, 98195, USA
| | - Vipul Bhatia
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Brian Hanratty
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Radhika A Patel
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Center for Functional Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, 98195, USA
| | - Michael C Haffner
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Pathology and Laboratory Medicine, University of Washington, Seattle, WA. 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Pathology and Laboratory Medicine, University of Washington, Seattle, WA. 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, 98195, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Crump Institute for Molecular Imaging, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - John K Lee
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Institute of Urologic Oncology, Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
13
|
Obinata D, Yamada Y, Sumiyoshi T, Tanegashima T, Watanabe R, Kobayashi H, Ito D, Urabe F. Recent advances in basic research on prostate cancer: Where we are heading? Int J Urol 2025; 32:219-228. [PMID: 39474871 DOI: 10.1111/iju.15628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/17/2024] [Indexed: 03/21/2025]
Abstract
In the over 80 years since androgens were found to play a pivotal role in prostate cancer (PCa) progression, androgen deprivation therapy (ADT) has been a cornerstone in treating advanced PCa. Castration-resistant PCa persists, however, with some of these tumors evolving to androgen receptor (AR)-independent forms like neuroendocrine PCa. The development of novel diagnostic and therapeutic approaches to PCa is therefore crucial. This review provides an overview of recent basic research in PCa, focusing on two main areas: PCa cells and their tumor microenvironments. The first section describes current knowledge on the intricate mechanisms of AR signaling pathways, emphasizing the roles of coactivators and chromatin state alterations in gene regulation. Genomic analyses have revealed recurrent mutations and copy number alterations critical for precision medicine. Liquid biopsy has become a promising tool for real-time tumor monitoring, identifying genetic alterations in circulating-tumor DNA or extracellular vesicles. The second section describes the tumor microenvironment of PCa, highlighting its immunosuppressive landscape and the potential of combining ADT with immunotherapy. Advanced techniques, including single-cell RNA sequencing and spatial transcriptomics offer insights into cellular heterogeneity and interactions within the tumor microenvironment, paving the way for novel therapeutic strategies. Integration of these diverse research areas will provide a comprehensive understanding of the current state and future directions of PCa research, underscoring the importance of personalized medicine and the dynamic nature of cancer treatment strategies.
Collapse
Affiliation(s)
- Daisuke Obinata
- Department of Urology, Nihon University School of Medicine, Tokyo, Japan
| | - Yasutaka Yamada
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takayuki Sumiyoshi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tokiyoshi Tanegashima
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuta Watanabe
- Department of Urology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroaki Kobayashi
- Department of Urology, National Defense Medical College, Saitama, Japan
| | - Daisuke Ito
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Tanegashima T, Shiota M, Terada N, Saito T, Yokomizo A, Kohei N, Goto T, Kawamura S, Hashimoto Y, Takahashi A, Kimura T, Tabata KI, Tomida R, Hashimoto K, Sakurai T, Shimazui T, Sakamoto S, Kamiyama M, Tanaka N, Mitsuzuka K, Kato T, Narita S, Yasumoto H, Teraoka S, Kato M, Osawa T, Nagumo Y, Matsumoto H, Enokida H, Sugiyama T, Kuroiwa K, Kitamura H, Kamoto T, Eto M. Improved prognosis of de novo metastatic prostate cancer after an introduction of life-prolonging agents for castration-resistant prostate cancer. Int J Clin Oncol 2025; 30:551-558. [PMID: 39688742 DOI: 10.1007/s10147-024-02681-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND In Japan, since 2014, new treatments such as androgen receptor signaling inhibitors and cabazitaxel have become applicable for metastatic castration-resistant prostate cancer (mCRPC), leading to dramatic changes in treatment options. OBJECTIVE This study aims to evaluate the impact of recent advancements in treatment options on the overall survival (OS) of patients diagnosed with de novo metastatic castration-sensitive prostate cancer (mCSPC) in Japan. METHODS A retrospective analysis was conducted on 2450 Japanese men diagnosed with de novo mCSPC between 2008 and 2018. Patients were stratified into two groups based on the period of diagnosis: an earlier period (2008-2013) and a later period (2014-2018). OS was compared between earlier and later periods using Kaplan-Meier analysis in total and propensity score matched subpopulation as well as risk-stratified subgroups. RESULTS Patients diagnosed in the later period exhibited significantly improved OS compared to those diagnosed in the earlier period. The risk score, calculated based on ISUP grade group, LDH levels, and ALP levels, was a poor prognostic factor. In the later period, compared to the earlier period, there was no improvement in OS in the favorable-risk group, but a significant improvement was observed in the poor-risk group. CONCLUSION It was suggested that the introduction of novel androgen receptor signaling inhibitors and chemotherapy treatment regimens since 2014 has led to improved survival outcomes for patients with de novo mCSPC, particularly those with poor-risk profiles. The findings highlight the impact of recent advancements in treatment on the prognosis of patients with metastatic prostate cancer in Japan.
Collapse
Affiliation(s)
- Tokiyoshi Tanegashima
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Naoki Terada
- Department of Urology, Miyazaki University, Miyazaki, Japan
| | - Toshihiro Saito
- Department of Urology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Akira Yokomizo
- Department of Urology, Harasanshin Hospital, Fukuoka, Japan
| | - Naoki Kohei
- Department of Urology, Shizuoka General Hospital, Shizuoka, Japan
| | - Takayuki Goto
- Department of Urology, Kyoto University, Kyoto, Japan
| | | | | | - Atsushi Takahashi
- Department of Urology, Hakodate Goryoukaku Hospital, Hakodate, Japan
| | | | - Ken-Ichi Tabata
- Department of Urology, Kitasato University, Sagamihara, Japan
| | - Ryotaro Tomida
- Department of Urology, Shikoku Cancer Center, Matsuyama, Japan
| | - Kohei Hashimoto
- Department of Urology, Sapporo Medical University, Sapporo, Japan
| | | | - Toru Shimazui
- Department of Urology, Ibaraki Prefectural Central Hospital, Ibaraki Cancer Center, Kasama, Japan
| | | | - Manabu Kamiyama
- Department of Urology, University of Yamanashi Hospital, Chuo, Japan
| | - Nobumichi Tanaka
- Department of Urology, and Department of Prostate Brachytherapy, Nara Medical University, Kashihara, Japan
| | | | - Takuma Kato
- Department of Urology, Kagawa University, Kagawa, Japan
| | | | | | - Shogo Teraoka
- Department of Urology, Tottori University, Yonago, Japan
| | - Masashi Kato
- Department of Urology, Nagoya University, Nagoya, Japan
| | - Takahiro Osawa
- Department of Renal and Genitourinary Surgery, Hokkaido University, Sapporo, Japan
| | - Yoshiyuki Nagumo
- Department of Urology, University of Tsukuba Hospital, Tsukuba, Japan
| | | | - Hideki Enokida
- Department of Urology, Kagoshima University, Kagoshima, Japan
| | - Takayuki Sugiyama
- Department of Urology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kentaro Kuroiwa
- Department of Urology, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Hiroshi Kitamura
- Department of Urology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, Japan
| | | | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
15
|
Fanelli GN, Nuzzo PV, Pederzoli F, Loda M. Deciphering Complexity: The Molecular Landscape of Castration-Resistant Prostate Cancer. Surg Pathol Clin 2025; 18:25-39. [PMID: 39890307 PMCID: PMC11787547 DOI: 10.1016/j.path.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
Despite improvements in diagnosis and treatment approaches, prostate cancer (PC) remains a leading cause of cancer-related death in men. PC progresses through various stages, mostly driven by androgen receptor signaling. However, after androgen deprivation therapies, in a significant portion of patients, several different molecular mechanisms contribute to the development of castration resistance. Delving deeply into the molecular landscape of castration-resistant PC, grasping the selective pressures exerted by therapies, and understanding the drivers of lineage plasticity is pivotal to prevent progression. Targeting genetic and epigenetic alterations that drive this transition will guide clinical management strategies and possibly prevent and/or treat lethal disease.
Collapse
Affiliation(s)
- Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, Pisa 56125, Italy
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
16
|
Taniguchi H, Ikeda J, Masuo Y, Kinoshita H. The impact of the number of prior androgen receptor pathway inhibitors before cabazitaxel treatment in patients with metastatic castration-resistant prostate cancer. Jpn J Clin Oncol 2025:hyaf034. [PMID: 39987487 DOI: 10.1093/jjco/hyaf034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/09/2025] [Indexed: 02/25/2025] Open
Abstract
OBJECTIVE Cabazitaxel (CAZ) has been shown to prolong overall survival (OS) in patients with metastatic castration-resistant prostate cancer (mCRPC) following docetaxel treatment. However, the impact of the number of prior androgen receptor pathway inhibitors (ARPIs) on CAZ efficacy remains unclear. This study aimed to analyze the effectiveness of CAZ based on the number of prior ARPIs administered before CAZ treatment. METHODS A retrospective review was conducted on mCRPC patients who received CAZ. The differences in CAZ efficacy based on the number of prior ARPIs were evaluated and prognostic factors for prostate-specific antigen (PSA) progression-free survival (PFS) and OS were analyzed. RESULTS A total of 59 patients were categorized into three groups: 12 patients with no prior ARPI, 26 with one prior ARPI, and 21 with two or more prior ARPIs. The median number of CAZ cycles and relative dose intensity were 5% and 60%, respectively. No significant differences were observed in PSA response or PFS among the three groups. Although OS from the first CAZ administration did not significantly differ among the groups, the OS from mCRPC diagnosis was shorter in the group with no prior ARPI. Multivariate analysis identified a time to mCRPC diagnosis of less than 11 months and low serum hemoglobin levels before CAZ administration as significant prognostic factors of poor OS following CAZ treatment. CONCLUSIONS PSA response, PFS, and OS after CAZ administration did not significantly differ based on the number of prior ARPIs. CAZ efficacy appears consistent regardless of the number of prior ARPI sequences.
Collapse
Affiliation(s)
- Hisanori Taniguchi
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| | - Junichi Ikeda
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| | - Yuki Masuo
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shinmachi, Hirakata, Osaka 573-1191, Japan
| |
Collapse
|
17
|
Khorasanchi A, Hong F, Yang Y, Singer EA, Wang P, Li M, Zheng L, Monk P, Mortazavi A, Meng L. Overcoming drug resistance in castrate-resistant prostate cancer: current mechanisms and emerging therapeutic approaches. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:9. [PMID: 40051495 PMCID: PMC11883235 DOI: 10.20517/cdr.2024.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 03/09/2025]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is driven by a complex network of resistance mechanisms against standard-of-care therapies, resulting in poor long-term outcomes. This review offers a uniquely comprehensive and integrative perspective on these resistance pathways, systematically examining both androgen receptor (AR)-dependent factors (including AR overexpression, point mutations, glucocorticoid receptor signaling, splice variants, post-translational modifications, altered coregulators, and intratumoral hormone biosynthesis) and AR-independent pathways (such as neuroendocrine differentiation, lineage plasticity, and alternative growth factor signaling). We also highlight resistance mechanisms influencing immunotherapy, chemotherapy, radiopharmaceutical therapy and targeted therapy. By synthesizing emerging insights across these domains, this review not only clarifies the underlying biology of mCRPC resistance but also identifies key leverage points for more effective interventions. Building on this foundation, we propose a forward-looking framework for overcoming mCRPC drug resistance, emphasizing the importance of biomarker-guided patient selection, combination strategies that simultaneously target multiple resistance mechanisms, and novel therapies under investigation. These recommendations are intended to guide future clinical trial designs and research priorities that move beyond incremental improvements. Ultimately, this comprehensive synthesis aims to serve as a resource for clinicians and researchers to accelerate the development of durable, precision-based treatment strategies in mCRPC.
Collapse
Affiliation(s)
- Adam Khorasanchi
- Division of Hospital Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Feng Hong
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yuanquan Yang
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Eric A. Singer
- Division of Urologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Peng Wang
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Mingjia Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Linghua Zheng
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Paul Monk
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Stone BV, Dominas CA, Bhagavatula SK, Ahn SW, Tatarova Z, Jakubik J, Matthew D, Mossanen M, Furtado D, Tuncali K, Hata N, Tempany C, Jonas O, Kibel AS. Novel Intraprostatic Magnetic Resonance-Guided Implantation of Multidrug-Eluting Microdevice for Testing of Systemic Therapy Agents In Situ: Proof of Concept in Intermediate-Risk and High-Risk Prostate Cancer. J Urol 2025; 213:173-182. [PMID: 39348711 PMCID: PMC11717626 DOI: 10.1097/ju.0000000000004269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
PURPOSE The purpose of this study was to assess safety and feasibility of percutaneous magnetic resonance-guided placement of an implantable microdevice (IMD) to evaluate in situ intratumor response to multiple pharmacologic agents in men with intermediate-risk and high-risk localized prostate cancer. MATERIALS AND METHODS Biocompatible IMDs measuring 750 µm in diameter and 5 mm in length were prepared with 20 reservoirs containing candidate drug and drug combinations including second-generation androgen inhibitors, PARP inhibitors, PD-1 inhibitors, and conventional chemotherapy. Men with intermediate-risk or high-risk localized prostate cancer and MRI-visible lesions were enrolled. Up to 4 IMDs were placed using a transperineal approach into MRI-visible tumors 2 days before planned radical prostatectomy. After radical prostatectomy, the IMDs and a small segment of surrounding tumor tissue were removed and sectioned, stained, and analyzed for tissue drug response by a variety of pharmacodynamic markers. RESULTS Fourteen patients were enrolled: 7 (50%) with intermediate-risk and 7 (50%) with high-risk localized prostate cancer. A total of 53 IMDs were implanted (mean 3.8 per patient), and 49 IMDs (92%) were successfully retrieved. All men underwent uncomplicated robotic-assisted radical prostatectomy and bilateral pelvic lymph node dissection 2 days after IMD placement. There were no severe adverse events. Pathological examination of the tissues adjacent to the IMDs demonstrated differential drug response within patients and between patients. Limitations include small sample size. CONCLUSIONS A multidrug IMD can be safely placed percutaneously into MRI-visible lesions before radical prostatectomy, enabling assessment of tumor-specific local response to multiple agents simultaneously within the tumor's normal stromal environment to guide targeted systemic therapy. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT04399876.
Collapse
Affiliation(s)
- Benjamin V. Stone
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Urology, Medical University of South Carolina, Charleston, SC
| | - Christine A. Dominas
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sharath K. Bhagavatula
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sebastian W. Ahn
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Zuzana Tatarova
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Juraj Jakubik
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Destiny Matthew
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Matthew Mossanen
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Daniella Furtado
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Kemal Tuncali
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Nobuhiko Hata
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Clare Tempany
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Oliver Jonas
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Adam S. Kibel
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
19
|
Ohaegbulam KC, Post CM, Farris PE, Garzotto M, Beer TM, Hung A, Williamson CW. Safety and Efficacy of Neoadjuvant Docetaxel and Radiotherapy in Localized High-Risk Prostate Cancer: Results From a Prospective Pilot Study. Am J Clin Oncol 2025; 48:75-82. [PMID: 39473059 DOI: 10.1097/coc.0000000000001151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
OBJECTIVES Approximately 15% of patients with localized prostate cancer are at high risk for disease recurrence. Many clinical trials have evaluated the impact of neoadjuvant therapy before radical prostatectomy with mixed results (NCT00321698). METHODS This phase I/II clinical trial evaluated the tolerability and preliminary efficacy of neoadjuvant radiation therapy and docetaxel before prostatectomy in 25 men with high-risk prostate cancer. The treatment regimen included 45 Gy radiotherapy in 25 fractions to the prostate and seminal vesicles over 5 weeks, along with weekly dose-escalated docetaxel up to 30 mg/m², followed by prostatectomy and bilateral lymph node dissection. The primary endpoint was the rate of pathologic complete response (pCR). Secondary endpoints included adverse events, symptom and quality of life measures, and prostate-specific antigen metrics. RESULTS All 25 patients completed the planned treatment. The primary endpoint of pCR was not achieved. Lymphopenia was the most common grade 3 or higher toxicity, with no grade 3 or higher genitourinary or gastrointestinal toxicities observed. With a median follow-up of 11.6 years, the 10-year biochemical recurrence-free survival was 60%, and distant metastasis-free survival was 80%. Prostate cancer-specific survival and overall survival at 10 years were 84% and 60%, respectively. CONCLUSIONS Although pCR was not met, the treatment demonstrated a modest toxicity profile and reasonable long-term outcomes, suggesting feasibility and safety. Further studies are needed to optimize endpoints and assess the efficacy of neoadjuvant treatments compared with standard approaches in high-risk prostate cancer patients.
Collapse
Affiliation(s)
| | | | - Paige E Farris
- Oregon Health & Science University Knight Cancer Institute, School of Medicine, Oregon Health & Science University
| | - Mark Garzotto
- Urology, School of Medicine, Oregon Health & Science University
- Department of Urology, Portland Veterans Affairs Medical Center, Portland, OR
| | - Tomasz M Beer
- Oregon Health & Science University Knight Cancer Institute, School of Medicine, Oregon Health & Science University
| | | | | |
Collapse
|
20
|
Ding T, He L, Lin G, Xu L, Zhu Y, Wang X, Liu X, Guo J, Lei F, Zuo Z, Zheng J. Integrated analysis of single-cell and bulk transcriptomes uncovers clinically relevant molecular subtypes in human prostate cancer. Chin J Cancer Res 2025; 37:90-114. [PMID: 40078560 PMCID: PMC11893346 DOI: 10.21147/j.issn.1000-9604.2025.01.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/26/2024] [Indexed: 03/14/2025] Open
Abstract
Objective Prostate cancer (PCa) is a complex disease characterized by diverse cellular ecosystems within the tumor microenvironment (TME) and high tumor heterogeneity, which challenges clinically stratified management and reinforces the need for novel strategies to fight against castration-resistant PCa (CRPC). Methods We performed single-cell RNA sequencing (scRNA-seq) on 10 untreated primary PCa tissues and integrated public scRNA-seq resources from three normal prostate tissues, two untreated primary PCa tissues, and six CRPC tumors to portray a comprehensive cellular and molecular interaction atlas of PCa. We further integrated the single-cell and bulk transcriptomes of PCa to establish a molecular classification system. Results scRNA-seq profiles revealed substantial inter- and intra-tumoral heterogeneity across different cell subpopulations in untreated PCa and CRPC tumors. In the malignant epithelial reservoir, cells evolved along decoupled paths in treatment-naive PCa and CRPC tumors, and distinct transcriptional reprogramming processes were activated, highlighting anti-androgen therapy-induced lineage plasticity. Based on the specifically expressed markers of the epithelial subpopulations, we conducted unsupervised clustering analysis in The Cancer Genome Atlas prostate adenocarcinoma (TCGA-PRAD) cohort and identified three molecularly and clinically distinct subtypes. The C1 subtype, characterized by high enrichment of CRPC-enriched epithelial cells, had a high risk of rapid development of anti-androgen resistance and might require active surveillance and additional promising intervention treatments, such as integrin A3 (ITGA3) + integrin B1 (ITGB1) inhibition. The C2 subtype resembled the immune-modulated subtype that was most likely to benefit from anti-LAG3 immunotherapy. The C3 subtype had a favorable prognosis. Conclusions Our study provides a comprehensive and high-resolution landscape of the intricate architecture of the PCa TME, and our trichotomic molecular taxonomy could help facilitate precision oncology.
Collapse
Affiliation(s)
- Tao Ding
- Department of Urology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan 512025, China
- Department of Urology, Southern Medical University Affifiliated Fengxian Hospital, Shanghai 201499, China
| | - Lina He
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Guowen Lin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lei Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yanjun Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xinan Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xuefei Liu
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Fanghong Lei
- Department of Pathology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan 512025, China
| | - Zhixiang Zuo
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Jianghua Zheng
- Central Laboratory, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Department of Laboratory Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| |
Collapse
|
21
|
Palacios PA, Flores I, Cereceda L, Otero FF, Müller M, Brebi P, Contreras HR, Carreño LJ. Patient-Derived Organoid Models for NKT Cell-Based Cancer Immunotherapy. Cancers (Basel) 2025; 17:406. [PMID: 39941775 PMCID: PMC11815936 DOI: 10.3390/cancers17030406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Invariant Natural Killer T (iNKT) cells are a unique subset of T cells that bridge innate and adaptive immunity, displaying potent anti-tumor properties through cytokine secretion, direct cytotoxicity, and recruitment of immune effector cells such as CD8+ T cells and NK cells. Despite their therapeutic potential, the immunosuppressive tumor microenvironment (TME), characterized by regulatory T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), limits iNKT cell efficacy. Patient-derived organoid (PDO) platforms provide an innovative model for dissecting these complex interactions and evaluating strategies to reinvigorate iNKT cell functionality within the TME. PDOs closely mimic the genetic, phenotypic, and structural characteristics of primary tumors, enabling the study of tumor-immune dynamics. Integrating iNKT cells into PDOs offers a robust platform for investigating CD1d-mediated interactions, Th1-biased immune responses driven by glycolipid analogs like α-GalCer, and combination therapies such as immune checkpoint inhibitors. Additionally, PDO systems can assess the effects of metabolic modulation, including reducing lactic acid accumulation or targeting glutamine pathways, on enhancing iNKT cell activity. Emerging innovations, such as organoid-on-a-chip systems, CRISPR-Cas9 gene editing, and multi-omics approaches, further expand the potential of PDO-iNKT platforms for personalized immunotherapy research. Although the application of iNKT cells in PDOs is still undeveloped, these systems hold immense promise for bridging preclinical studies and clinical translation. By addressing the challenges of the TME and optimizing therapeutic strategies, PDO-iNKT platforms offer a transformative avenue for advancing cancer immunotherapy and personalized medicine.
Collapse
Affiliation(s)
- Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Iván Flores
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8350499, Chile
| | - Lucas Cereceda
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Francisco F. Otero
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Priscilla Brebi
- Millennium Institute on Immunology and Immunotherapy, Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile
- Biomedical Research Consortium (BMRC), Santiago 8331150, Chile
| | - Héctor R. Contreras
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8350499, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8350499, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Biomedical Research Consortium (BMRC), Santiago 8331150, Chile
| |
Collapse
|
22
|
Li J, Chong T, Wang L, Li Z, Jin Y, Chen Y, Liu D, Jiang L, Chen D, Li Z. Prostate ductal adenocarcinoma with MLH1 copy number loss, microsatellite instability high and BRCA2 mutation. Int Cancer Conf J 2025; 14:46-49. [PMID: 39758786 PMCID: PMC11695545 DOI: 10.1007/s13691-024-00734-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/14/2024] [Indexed: 01/07/2025] Open
Abstract
Mismatch repair deficiency (MMRd) or microsatellite instability high (MSI-H) is rare in prostate cancer and more frequently observed in cases with ductal histology. MLH1 copy number loss is extremely rare in MMRd tumors. Herein, we describe a case of prostate ductal adenocarcinoma with MLH1 copy number loss, microsatellite instability high and BRCA2 mutation could derive benefit from immunotherapy plus ADT. A 72-year-old Chinese patient was diagnosed with poorly differentiated prostate ductal adenocarcinoma mixed with acinar adenocarcinoma (Gleason 5 + 4). Next-generation sequencing (NGS) showed a hypermutated tumor with a mutational burden of 34.71 mutations per Mb and microsatellite instability high (MSI-H). Suspected biallelic MLH1 loss (copy number 0.16) and a pathogenic somatic BRCA2 variant (E2981Kfs*7) were detected. After surgery, the patient received androgen-deprivation therapy (ADT) with goserelin (10.8 mg every 3 months) and tislelizumab (200 mg every 3 weeks). At the 1-year follow-up, the PSA level was lower than 0.01 ng/ml and a pelvic MRI revealed no abnormalities. Our case highlights the intricate molecular mechanisms of MMRd prostate cancer.
Collapse
Affiliation(s)
- Jianping Li
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 China
| | - Li Wang
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 China
| | - Zihao Li
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 China
| | - Yaofeng Jin
- Department of Pathology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yan Chen
- Xiamen Spacegen Co. Ltd, Xiamen, China
| | - Dujuan Liu
- Suzhou Spaceseq MedLab Co. Ltd, Suzhou, China
| | | | - Deyi Chen
- Suzhou Spaceseq MedLab Co. Ltd, Suzhou, China
| | - Zhaolun Li
- Department of Urology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 China
| |
Collapse
|
23
|
Klümper N, Eckstein M. Reply to Feng Qi's Letter to the Editor re: Niklas Klümper, Viktor Grünwald, Arndt Hartmann, et al. The Role of Microsatellite Instability/DNA Mismatch Repair Deficiency and Tumor Mutational Burden as Biomarkers in Predicting Response to Immunotherapy in Castration-resistant Prostate Cancer. Eur Urol. 2024;86:388-390. Eur Urol 2025; 87:e17-e18. [PMID: 39317629 DOI: 10.1016/j.eururo.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Affiliation(s)
- Niklas Klümper
- Department of Urology, University Hospital Bonn, Bonn, Germany; Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Bonn, Germany; Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; EMN Comprehensive Cancer Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Bavarian Center for Cancer Research, Munich, Germany.
| |
Collapse
|
24
|
Lyu A, Fan Z, Clark M, Lea A, Luong D, Setayesh A, Starzinski A, Wolters R, Arias-Badia M, Allaire K, Wu K, Gurunathan V, Valderrábano L, Wei XX, Miller RA, Van Allen EM, Fong L. Evolution of myeloid-mediated immunotherapy resistance in prostate cancer. Nature 2025; 637:1207-1217. [PMID: 39633050 PMCID: PMC11779626 DOI: 10.1038/s41586-024-08290-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024]
Abstract
Patients with advanced metastatic castration-resistant prostate cancer (mCRPC) are refractory to immune checkpoint inhibitors (ICIs)1,2, partly because there are immunosuppressive myeloid cells in tumours3,4. However, the heterogeneity of myeloid cells has made them difficult to target, making blockade of the colony stimulating factor-1 receptor (CSF1R) clinically ineffective. Here we use single-cell profiling on patient biopsies across the disease continuum and find that a distinct population of tumour-associated macrophages with elevated levels of SPP1 transcripts (SPP1hi-TAMs) becomes enriched with the progression of prostate cancer to mCRPC. In syngeneic mouse modelling, an analogous macrophage population suppresses CD8+ T cell activity in vitro and promotes ICI resistance in vivo. Furthermore, Spp1hi-TAMs are not responsive to anti-CSF1R antibody treatment. Pathway analysis identifies adenosine signalling as a potential mechanism for SPP1hi-TAM-mediated immunotherapeutic resistance. Indeed, pharmacological inhibition of adenosine A2A receptors (A2ARs) significantly reverses Spp1hi-TAM-mediated immunosuppression in CD8+ T cells in vitro and enhances CRPC responsiveness to programmed cell death protein 1 (PD-1) blockade in vivo. Consistent with preclinical results, inhibition of A2ARs using ciforadenant in combination with programmed death 1 ligand 1 (PD-L1) blockade using atezolizumab induces clinical responses in patients with mCRPC. Moreover, inhibiting A2ARs results in a significant decrease in SPP1hi-TAM abundance in CRPC, indicating that this pathway is involved in both induction and downstream immunosuppression. Collectively, these findings establish SPP1hi-TAMs as key mediators of ICI resistance in mCRPC through adenosine signalling, emphasizing their importance as both a therapeutic target and a potential biomarker for predicting treatment efficacy.
Collapse
Affiliation(s)
- Aram Lyu
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Immunotherapy Integrated Research Center, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Zenghua Fan
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew Clark
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Averey Lea
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Diamond Luong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ali Setayesh
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Alec Starzinski
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Rachel Wolters
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Marcel Arias-Badia
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kate Allaire
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kai Wu
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Vibha Gurunathan
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Laura Valderrábano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xiao X Wei
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Eliezer M Van Allen
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- Immunotherapy Integrated Research Center, Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
25
|
Bryce AH, Agarwal N, Beltran H, Hussain MH, Sartor O, Shore N, Antonarakis ES, Armstrong AJ, Calais J, Carducci MA, Dorff TB, Efstathiou JA, Gleave M, Gomella LG, Higano C, Hope TA, Iagaru A, Morgans AK, Morris DS, Morris MJ, Petrylak DP, Reiter RE, Rettig MB, Ryan CJ, Sellinger SB, Spratt DE, Srinivas S, Tagawa ST, Taplin M, Yu EY, Zhang T, McKay RR, Koo PJ, Crawford ED. Implementing evidence-based strategies for men with biochemically recurrent and advanced prostate cancer: Consensus recommendations from the US Prostate Cancer Conference 2024. Cancer 2025; 131:e35612. [PMID: 39616467 PMCID: PMC11694557 DOI: 10.1002/cncr.35612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 01/03/2025]
Abstract
Current US clinical practice guidelines for advanced prostate cancer management contain recommendations based on high-level evidence from randomized controlled trials; however, these guidelines do not address the nuanced clinical questions that are unanswered by prospective trials but nonetheless encountered in day-to-day practice. To address these practical questions, the 2024 US Prostate Cancer Conference (USPCC 2024) was created to generate US-focused expert clinical decision-making guidance for circumstances in which level 1 evidence is lacking. At the second annual USPCC meeting (USPCC 2024), a multidisciplinary panel of experts convened to discuss ongoing clinical challenges related to 5 topic areas: biochemical recurrence; metastatic, castration-sensitive prostate cancer; poly [ADP-ribose] polymerase inhibitors; prostate-specific membrane antigen radioligand therapy; and metastatic, castration-resistant prostate cancer. Through a modified Delphi process, 34 consensus recommendations were developed and are intended to provide clinicians who manage prostate cancer with guidance related to the implementation of novel treatments and technologies. In this report, the authors review the areas of consensus identified by the USPCC 2024 experts and evaluate ongoing unmet needs regarding translational application of the current clinical evidence.
Collapse
Affiliation(s)
- Alan H. Bryce
- Department of Medical Oncology and Therapeutics ResearchCity of Hope PhoenixPhoenixArizonaUSA
| | - Neeraj Agarwal
- Huntsman Cancer InstituteUniversity of UtahSalt Lake CityUtahUSA
| | - Himisha Beltran
- Department of Medical OncologyDana‐Farber Cancer Institute and Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Maha H. Hussain
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityEvanstonIllinoisUSA
| | - Oliver Sartor
- Department of Medical OncologyMayo ClinicRochesterMinnesotaUSA
| | - Neal Shore
- Carolina Urologic Research CenterMyrtle BeachSouth CarolinaUSA
| | | | - Andrew J. Armstrong
- Duke Cancer Institute Center for Prostate and Urologic CancersDurhamNorth CarolinaUSA
| | - Jeremie Calais
- Ahmanson Translational Theranostics DivisionDepartment of Molecular and Medical PharmacologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | | | | | - Jason A. Efstathiou
- Department of Radiation OncologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Martin Gleave
- Urological SciencesVancouver Prostate CenterUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Leonard G. Gomella
- Department of UrologySidney Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Thomas A. Hope
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular ImagingDepartment of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Alicia K. Morgans
- Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Michael J. Morris
- Genitourinary Oncology ServiceDepartment of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | | | - Robert E. Reiter
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Matthew B. Rettig
- Division of Hematology‐OncologyDepartment of MedicineVA Greater Los AngelesLos AngelesCaliforniaUSA
- Department of MedicineDavid Geffen School of Medicine at the University of California Los AngelesLos AngelesCaliforniaUSA
- Department of UrologyDavid Geffen School of Medicine at the University of California Los AngelesLos AngelesCaliforniaUSA
| | - Charles J. Ryan
- Department of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | | | - Sandy Srinivas
- Division of Medical OncologyStanford University Medical CenterStanfordCaliforniaUSA
| | - Scott T. Tagawa
- Division of Hematology and Medical OncologyWeill Cornell MedicineNew YorkNew YorkUSA
| | - Mary‐Ellen Taplin
- Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
| | - Evan Y. Yu
- Division of Hematology and OncologyDepartment of MedicineUniversity of Washington and Fred Hutchinson Cancer CenterSeattleWashingtonUSA
| | - Tian Zhang
- Division of Hematology and OncologyDepartment of Internal MedicineSimmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Rana R. McKay
- Department of UrologyUniversity of California San DiegoLa JollaCaliforniaUSA
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | | | - E. David Crawford
- Department of UrologyUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
26
|
Qi F. Re: Niklas Klümper, Viktor Grünwald, Arndt Hartmann, et al. The Role of Microsatellite Instability/DNA Mismatch Repair Deficiency and Tumor Mutational Burden as Biomarkers in Predicting Response to Immunotherapy in Castration-resistant Prostate Cancer. Eur Urol. 2025;87:388-390. Eur Urol 2025; 87:e15-e16. [PMID: 39327116 DOI: 10.1016/j.eururo.2024.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/07/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Feng Qi
- Department of Urologic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
27
|
Francini E, Agarwal N, Castro E, Cheng HH, Chi KN, Clarke N, Mateo J, Rathkopf D, Saad F, Tombal B. Intensification Approaches and Treatment Sequencing in Metastatic Castration-resistant Prostate Cancer: A Systematic Review. Eur Urol 2025; 87:29-46. [PMID: 39306478 DOI: 10.1016/j.eururo.2024.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND OBJECTIVE Recently, research on treatment intensification has gathered momentum, and three novel therapy combinations were approved for metastatic castration-resistant prostate cancer (mCRPC). This systematic review summarizes the current and emerging evidence around intensified strategies for mCRPC and provides guidance for an ideal therapeutic sequencing. METHODS Preferred Reporting Items for Systematic Review and Meta-analysis Protocols (PRISMA-P) guidelines were followed to perform this review. PubMed, EMBASE, Web of Science, Cochrane Library, ClinicalTrials.gov, and major international societies' online proceedings were searched comprehensively until May 15, 2024, for terms related to treatment intensification and sequencing for mCRPC. KEY FINDINGS AND LIMITATIONS Overall, 28 clinical trials and 24 ongoing studies of intensification treatments were included in this review. Algorithms of optimal sequencing of approved treatments for mCRPC were outlined according to the use of androgen receptor pathway inhibitors (ARPIs) with or without docetaxel for earlier disease states. In first line, poly(ADP-ribose) polymerase inhibitor + ARPI combinations improve radiographical progression-free survival (rPFS), particularly for those with BRCA1/2 alterations. The AKT inhibitor combination of ipatasertib + abiraterone extends rPFS in those with PTEN loss or PIK3CA/AKT1/PTEN alterations. In those with two or more risk factors for early progression on enzalutamide, radionuclide 177-Lu-PSMA-617 + enzalutamide prolongs progression-free survival. Ongoing research of intensified approaches for mCRPC, and available and potential predictive and prognostic biomarkers are discussed. CONCLUSIONS AND CLINICAL IMPLICATIONS Recent approvals and ongoing investigations of single agents and intensification approaches will keep transforming the mCRPC treatment landscape. Improvement of patient profiling applying recognized genomic, molecular, and clinical predictive and prognostic indicators is fundamental to optimize sequential use of available therapies.
Collapse
Affiliation(s)
- Edoardo Francini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Neeraj Agarwal
- Huntsman Cancer Institute (NCI-CCC), University of Utah, Salt Lake City, UT, USA
| | - Elena Castro
- Hospital Universitario 12 de octubre, Madrid, Spain
| | - Heather H Cheng
- University of Washington and the Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kim N Chi
- BC Cancer - Vancouver Center, University of British Columbia, Vancouver, BC, Canada
| | - Noel Clarke
- The Christie and Salford Royal Hospital NHS Foundation Trusts and University of Manchester, Manchester, UK
| | - Joaquin Mateo
- Vall d'Hebron Institute of Oncology and Vall d'Hebron University Hospital, Barcelona, Spain
| | - Dana Rathkopf
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Fred Saad
- Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Bertrand Tombal
- Division of Urology, Institut de Recherche Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
28
|
VanderWeele DJ, Hussain M. Management Decisions for Metastatic Castration-resistant Prostate Cancer in 2024. Eur Urol 2025; 87:47-48. [PMID: 39428329 DOI: 10.1016/j.eururo.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Affiliation(s)
- David J VanderWeele
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
29
|
Yang RK, Alvarez H, Lucas AS, Roy-Chowdhuri S, Rashid A, Chen H, Ballester LY, Sweeney K, Routbort MJ, Patel KP, Luthra R, Medeiros LJ, Toruner GA. Microsatellite instability and high tumor mutational burden detected by next generation sequencing are concordant with loss of mismatch repair proteins by immunohistochemistry. Cancer Genet 2025; 290-291:44-50. [PMID: 39700818 DOI: 10.1016/j.cancergen.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Impairment of DNA mismatch repair function in neoplasms can be assessed by DNA-based methods to assess for high microsatellite instability (MSI-High) or immunohistochemical (IHC) analysis to assess for deficiency of mismatch repair proteins (dMMR). Neoplasms with mismatch repair deficiency often have high tumor mutational burden (TMB-High). MSI-High, dMMR, and TMB-High are all histology agnostic biomarkers for potential therapy using immune checkpoint inhibitors (ICI). In this single center, retrospective study, our primary aim was to assess if NGS-based positive TMB/MSI findings are concordant with patient matched concurrent MMR IHC studies. In addition, we determined if positive TMB/MSI findings are attributable to genetic/epigenetic alterations of MMR genes. Finally, we explored potential associations between IHC, TMB and MSI findings and specific tumor types We screened 4,258 patients in our database who had tumor-normal-testing with our institutional high-throughput NGS-based CLIA assay between Apr 1, 2021-August 31, 2022 for TMB and MSI. We identified 65 patients who had neoplasms with documented TMB-High/MSI-High (n = 59) or TMB-High/MSI-Undetermined (n = 6) results as well as concurrent IHC results for MMR proteins [colorectal (n = 25), endometrial (n = 28), prostatic (n = 7), urothelial (n = 3), other (n = 5)]. The concordance between positive NGS TMB/MSI and MMR results was 98 %. Genetic/epigenetic alterations of MMR genes were documented in 78 % of the neoplasms. IHC studies for dMMR proteins revealed loss of MLH1/PMS2 (n = 33), MSH2/MSH6 (n = 14), MLH1/MSH2/PMS2 (n = 1), MLH1 (n = 1), MSH2 (n = 2), MSH6 (n = 6) and PMS2 (n = 6). All six prostatic neoplasms with dMMR had loss of MSH2/MSH6 (p < 0.0001). We conclude that neoplasms with positive results for TMB/MSI are highly concordant with positive dMMR results. Genetic/epigenetic alterations in the MMR genes are an underlying reason for most positive findings. The association of MSH2/MSH6 loss with prostatic neoplasms is of in-terest, but sample size is limited, and further studies are warranted to address this association.
Collapse
Affiliation(s)
- Richard K Yang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hector Alvarez
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Antony San Lucas
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Asif Rashid
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hui Chen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Leomar Y Ballester
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Keith Sweeney
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mark J Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gokce A Toruner
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
30
|
Isaacsson Velho P, Bastos DA, Saint'ana PT, Rigatti B, da Costa ET, Muniz DQB, Andreis F, Ferreira RDP, Giongo Pedrotti L, Maistro S, Katayama MLH, Folgueira MAAK, Morelle A, Leal A, de Castro G. Nivolumab in Patients with Metastatic Castration-Resistant Prostate Cancer with and without DNA Repair Defects. Clin Cancer Res 2024; 30:5342-5352. [PMID: 39330991 DOI: 10.1158/1078-0432.ccr-24-1595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/03/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE Despite the success of immune checkpoint inhibitors (ICI) across various cancers, their efficacy in metastatic castration-resistant prostate cancer (mCRPC) is modest, except for a subset of patients who experience significant, yet unpredictable, benefits. DNA repair defects (DRD) are associated with higher neoantigen load, which may predict response. Our study explored the potential of DRD for enhanced responsiveness to the ICI nivolumab. PATIENTS AND METHODS We conducted a phase II, multicenter, single-arm trial evaluating nivolumab in patients with mCRPC with prior docetaxel therapy. The DRD were assessed using ctDNA. The primary endpoint was PSA50 response. Secondary endpoints included the objective response rate, radiographic progression-free survival (rPFS), and overall survival. Also, exploratory comprehensive genomic profiling was performed via whole-exome sequencing of tumor samples and matched normal tissues, alongside PD-L1 expression evaluation. RESULTS Among the 38 enrolled patients, DRD was identifiable in 30.5% (11/36) through ctDNA and/or whole-exome sequencing analyses. The overall PSA50 response rate was 10.5% (4/38). The PSA50 and objective response rates did not significantly differ between patients with and without DRD (18.2% vs. 8%; P = 0.57 and 50% vs. 17.6%; P = 0.27, respectively). The median PSA-PFS (1.9 vs. 2.8 months; P = 0.52) and rPFS (3.4 vs. 5.5 months; P = 0.7) were not statistically different between patients with and without DRD. Grade ≥ 3 adverse events were reported in 47.3% of participants. CONCLUSIONS Nivolumab has clinical activity in a subset of patients with mCRPC; however, DRD does not predict response. These results highlight the necessity of identifying new biomarkers to more accurately determine patients with mCRPC who might respond to ICIs.
Collapse
Affiliation(s)
- Pedro Isaacsson Velho
- Hospital Moinhos de Vento, Porto Alegre, Brazil
- Johns Hopkins University, Baltimore, Maryland
| | | | | | | | | | - David Q B Muniz
- Hospital Sirio Libanes, São Paulo, Brazil
- Instituto do Cancer do Estado de Sao Paulo, São Paulo, Brazil
| | | | | | | | - Simone Maistro
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Centro de Investigação Translacional em Oncologia (CTO), Instituto do Cancer do Estado de Sao Paulo, Universidade de Sao Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology-C2PO, São Paulo, Brazil
| | - Maria Lucia Hirata Katayama
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Centro de Investigação Translacional em Oncologia (CTO), Instituto do Cancer do Estado de Sao Paulo, Universidade de Sao Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology-C2PO, São Paulo, Brazil
| | - Maria Aparecida Azevedo Koike Folgueira
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Centro de Investigação Translacional em Oncologia (CTO), Instituto do Cancer do Estado de Sao Paulo, Universidade de Sao Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology-C2PO, São Paulo, Brazil
| | | | | | - Gilberto de Castro
- Hospital Sirio Libanes, São Paulo, Brazil
- Instituto do Cancer do Estado de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Barnett RM, Jang A, Lanka S, Fu PI, Bucheit LA, Babiker H, Bryce A, Meyer HM, Choi Y, Moore C, Garje R, Gao X, Kim DW, Chang RY, Gulhati P, Ramaker R, Bansal R, Zhang T, Oliver Sartor A, Armstrong AJ, Bilen MA, Barata P. Blood-based tumor mutational burden impacts clinical outcomes of immune checkpoint inhibitor treated breast and prostate cancers. COMMUNICATIONS MEDICINE 2024; 4:256. [PMID: 39623081 PMCID: PMC11612475 DOI: 10.1038/s43856-024-00687-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Breast and prostate tumors are known to be less responsive to immune checkpoint inhibitors (ICIs). Tissue-based tumor mutation burden (tTMB) has emerged as a predictive biomarker of response to ICIs, including in these "cold tumors". In clinical practice, when tTMB is not available, blood-based TMB score (bTMB) can be used to consider treatment with ICIs. METHODS This retrospective, real-world study included a final cohort of metastatic breast and prostate cancer patients treated with an ICI following a liquid biopsy test. Multiple bTMB-High cut-offs were assessed. Clinical, genomic, and outcomes data were collected. We hypothesized that a cut-off of bTMB ≥10 mut/Mb is not a strong predictor of response to ICIs in this setting. The Guardant Health genomic database (GHGD) was then queried (N = 13,992) for associations of bTMB with genomic alterations. RESULTS In the clinical cohort (N = 48), ICI treatment is offered after a median of 3 (1-9) lines of treatment. The median bTMB is 16.4 (10-186) mut/Mb. The median time on ICI and PFS is 2.1 (0-1.7) and 3.1 months (95%CI, 1.6-4.6) respectively; no difference by MSI/MMR status (p = 0.152). Response rate among eligible patients (n = 36) is 16.7%; only N = 1/6 in bTMB <16 mut/Mb. High bMSI is associated with higher bTMB (correlation test, r = 0.66, p = 0.000). In the GHGD, patients with bTMB high have significantly more alterations than bTMB low and TP53, PIK3CA, ATM, ESR1, NF1, BRCA2, ARID1A, and APC were the most frequently altered genes. CONCLUSIONS In this study, the practice of offering an ICIs based on bTMB was uncommon and did not independently predict ICI benefits in patients with refractory, advanced breast and prostate cancers.
Collapse
Affiliation(s)
| | - Albert Jang
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Tulane Cancer Center, New Orleans, LA, USA
| | - Sree Lanka
- Tulane Cancer Center, New Orleans, LA, USA
| | - PIngfu Fu
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | | | | | | | | | | | | | - Rohan Garje
- Miami Cancer Institute Baptist Health, Miami, FL, USA
| | - Xin Gao
- Massachuesetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Richard Y Chang
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | | | | | | | - Tian Zhang
- UT Southwestern University, Dallas, TX, USA
| | | | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC, USA
| | | | - Pedro Barata
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA.
- Tulane Cancer Center, New Orleans, LA, USA.
| |
Collapse
|
32
|
Ou Y, Xia C, Ye C, Liu M, Jiang H, Zhu Y, Yang D. Comprehensive scRNA-seq analysis to identify new markers of M2 macrophages for predicting the prognosis of prostate cancer. Ann Med 2024; 56:2398195. [PMID: 39221762 PMCID: PMC11370685 DOI: 10.1080/07853890.2024.2398195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) has become the highest incidence of malignant tumor among men in the world. Tumor microenvironment (TME) is necessary for tumor growth. M2 macrophages play an important role in many solid tumors. This research aimed at the role of M2 macrophages' prognosis value in PCa. METHODS Single-cell RNA-seq (scRNA-seq) data and mRNA expression data were obtained from the Gene Expression Omnibus database (GEO) and The Cancer Genome Atlas (TCGA). Quality control, normalization, reduction, clustering, and cell annotation of scRNA-seq data were preformed using the Seruat package. The sub-populations of the tumor-associated macrophages (TAMs) were analysis and the marker genes of M2 macrophage were selected. Differentially expressed genes (DEGs) in PCa were identified using limma and the immune infiltration was detected using CIBERSORTx. Then, a weighted correlation network analysis (WGCNA) was constructed to identify the M2 macrophage-related modules and genes. Integration of the marker genes of M2 macrophage from scRNA-seq data analysis and hub genes from WGCNA to select the prognostic gene signature based on Univariate and LASSO regression analysis. The risk score was calculated, and the DEGs, biological function, immune characteristics related to risk score were explored. And a predictive nomogram was constructed. CCK8, Transwell, and wound healing were used to verify cell phenotype changes after co-cultured. RESULTS A total of 2431 marker genes of M2 macrophage and 650 hub M2 macrophage-related genes were selected based on scRNA-seq data and WGCNA. Then, 113 M2 macrophage-related genes were obtained by overlapping the scRNA-seq data and WGCNA results. Nine M2 macrophage-related genes (SMOC2, PLPP1, HES1, STMN1, GPR160, ABCG1, MAZ, MYC, and EPCAM) were screened as prognostic gene signatures. M2 risk score was calculated, the DEGs, Immune score, stromal score, ESTIMATE score, tumor purity, and immune cell infiltration, immune checkpoint expression, and responses of immunotherapy and chemotherapy were identified. And a predictive nomogram was constructed. CCK8, Transwell invasion, and wound healing further verified that M2 macrophages promoted the proliferation, invasion, and migration of PCa (p < 0.05). CONCLUSIONS We uncovered that M2 macrophages and relevant genes played key roles in promoting the occurrence, development, and metastases of PCa and played as convincing predictors in PCa.
Collapse
Affiliation(s)
- Yitian Ou
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Chengxing Xia
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Chunwei Ye
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Mingming Liu
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Haiyang Jiang
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Yong Zhu
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Delin Yang
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| |
Collapse
|
33
|
Hamid AA, Sweeney CJ, Hovens C, Corcoran N, Azad AA. Precision medicine for prostate cancer: An international perspective. Urol Oncol 2024; 42:392-401. [PMID: 38614920 DOI: 10.1016/j.urolonc.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 04/15/2024]
Abstract
Greater personalization of cancer medicine continues to shape therapy development and patient selection accordingly. The treatment of prostate cancer has evolved considerably since the discovery of androgen deprivation therapy. The comprehensive profiling of the prostate cancer genome has mapped the targetable molecular landscape of the disease and identified opportunities for the implementation of novel and combination therapies. In this review, we provide an overview of the molecular biology of prostate cancer and tools developed to aid prognostication and prediction of therapy benefit. Modern treatment of advanced prostate cancer is reviewed as a paradigm of increasing precision-informed approach to patient care, and must be considered on a global scale with respect to the state of science and care delivery.
Collapse
Affiliation(s)
- Anis A Hamid
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Surgery, University of Melbourne, Melbourne, Australia.
| | | | | | - Niall Corcoran
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Arun A Azad
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
34
|
van Wilpe S, Kloots ISH, Slootbeek PHJ, den Brok M, Westdorp H, Franken MD, Coskunturk M, Osinga T, Bloemendal H, Adema G, Smeenk RJ, Nagarajah J, van Ipenburg J, Kroeze LI, Ligtenberg MJL, Schalken J, Gerritsen WR, Mehra N. Ipilimumab with nivolumab in molecularly selected patients with castration-resistant prostate cancer: primary analysis of the phase II INSPIRE trial. Ann Oncol 2024; 35:1126-1137. [PMID: 39293514 DOI: 10.1016/j.annonc.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) typically exhibits resistance to immune checkpoint inhibitors (ICIs). However, a subset of mCRPC patients displays a more immunogenic profile. This study examines efficacy and safety of dual ICI therapy in molecularly selected mCRPC. PATIENTS AND METHODS This single-arm, phase II trial included 69 molecularly selected mCRPC patients with mismatch repair deficiency (dMMR), non-synonymous tumour mutational burden ≥7.1 muts/Mb (hTMB), a BRCA2 mutation (BRCAm), or biallelic CDK12 inactivation (CDK12i). Efficacy was assessed in ICI-naïve patients (cohort A) with RECIST 1.1 (A1) and Prostate Cancer Working Group 3 (A2) measurable disease. Safety was evaluated in cohorts A and B (prior ICI monotherapy). Treatment included nivolumab 3 mg/kg and ipilimumab 1 mg/kg every 3 weeks for four cycles, followed by nivolumab 480 mg every 4 weeks for up to 1 year. The primary endpoint was disease control rate beyond 6 months (DCR > 6), aiming to surpass a DCR > 6 of 22%. RESULTS Patients initiated treatment between January 2021 and February 2024. Cohort A included 65 patients. Of these, 21 had dMMR (32%), 8 had hTMB (12%), 20 had BRCAm (31%), and 16 had CDK12i (25%). DCR > 6 was achieved in 38% of patients [95% confidence interval (CI) 27% to 51%], and was highest in dMMR (81%), followed by hTMB (25%), CDK12i (19%), and BRCAm (15%). Objective response rate in cohort A was 38% (95% CI 22% to 55%) and 47% (95% CI 34% to 60%) exhibited a 50% decline in prostate-specific antigen levels. Median progression-free survival (PFS) was 4.0 months (95% CI 3.5-12.0 months) in cohort A, and 32.7 months (95% CI 21.8 months-not reached) in dMMR patients. Treatment-related adverse events (TRAEs) led to permanent discontinuation in 14 of 69 patients (20%). Grade ≥3 TRAEs occurred in 48% of patients, with diarrhoea and elevated transaminases each in 10%. There was one treatment-related death due to a bowel perforation and a second following euthanasia after grade 4 toxicity. CONCLUSIONS This trial of dual ICIs in molecularly selected mCRPC met its primary endpoint, showing DCR > 6 in 38% of patients. Dual ICIs exhibited modest responses in the hTMB, BRCAm, and CDK12i subgroups, but demonstrated exceptional efficacy in dMMR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - G Adema
- Department of Radiation Oncology
| | | | | | | | | | | | - J Schalken
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - N Mehra
- Department of Medical Oncology.
| |
Collapse
|
35
|
San-Jose Manso L, Alfranca A, Moreno-Pérez I, Ruiz-Vico M, Velasco C, Toquero P, Pacheco M, Zapatero A, Aldave D, Celada G, Albers E, Fenor de la Maza MD, García J, Castro E, Olmos D, Colomer R, Romero-Laorden N. Immunome profiling in prostate cancer: a guide for clinicians. Front Immunol 2024; 15:1398109. [PMID: 39635522 PMCID: PMC11614818 DOI: 10.3389/fimmu.2024.1398109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Tumor immune microenvironment (TIME) plays a key role to understand how tumors respond to prostate cancer (PC) therapies and potential mechanisms of resistance. Previous research has suggested that specific genomic aberrations, such as microsatellite instability (MSI) or CDK12 bi-allelic loss can allow PC patients more likely to respond to immune checkpoint inhibitors (ICI) or other immune therapies. However, responses to these treatments remain highly variable even in selected patients. Thus, it is essential to obtain more information about tumor immune cells that infiltrate these tumors, and on their plasticity and interactions, in order to better understand the underlying biology to allow development of new therapeutic strategies. This review analyzes: 1) How interactions among immune cell populations and other cells infiltrating the tumor stroma can modulate the progression of PC, 2) How the standard therapies to treat PC (such as androgen deprivation therapy, new androgen-directed hormone therapy or chemotherapy) may influence the dynamic changes of the immunome and 3) What are the limitations in characterizing the immune landscape of the host´s response to tumors.
Collapse
Affiliation(s)
| | - Arantzazu Alfranca
- Immunology Department, Hospital Universitario La Princesa, Madrid, Spain
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ignacio Moreno-Pérez
- Medical Oncology Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - María Ruiz-Vico
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Clara Velasco
- Urology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Patricia Toquero
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - María Pacheco
- GU Translational Research Unit, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain
| | - Almudena Zapatero
- Radiation Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Diego Aldave
- Radiation Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Guillermo Celada
- Urology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Eduardo Albers
- Urology Department, Hospital Universitario La Princesa, Madrid, Spain
| | | | - Jorge García
- Biocomputing Unit, Hospital Niño Jesús, Instituto de Investigación Sanitaria de la Princesa, Madrid, Spain
| | - Elena Castro
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - David Olmos
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ramón Colomer
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| | - Nuria Romero-Laorden
- Personalized Precision Medicine Chair, Universidad Autónoma de Madrid, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
| |
Collapse
|
36
|
Yan H, Mao AW, Li D, Pérez-Baena MJ, Jiménez-Navas A, Wang D, Hong R, Cai W, Pérez-Losada J, Jen KY, Wang S, Peng S, Barcellos-Hoff MH, Mao JH, Fu Y, Iczkowski KA, Gulati S, Chang H. AI-Powered cellular morphometric biomarkers discovered in needle biopsy of prostatic cancer predict neoadjuvant androgen deprivation therapy response and prognosis: an international multicenter retrospective study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.17.24317411. [PMID: 39606414 PMCID: PMC11601692 DOI: 10.1101/2024.11.17.24317411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
It is imperative to identify patients with prostate cancer (PCa) who will benefit from androgen receptor signaling inhibitors that can impact quality of life upon prolonged use. Using our extensively-validated artificial-intelligence technique: cellular morphometric biomarker via machine learning (CMB-ML), we identified 13 CMBs from whole slide images of needle biopsies from the trial specimens ( NCT02430480 , n=37) that accurately predicted response to neoadjuvant androgen deprivation therapy (NADT) (AUC: 0.980). Notably, 13-CMB model stratified PCa patients into responder and non-responder groups after NADT treatment in an independent hospital cohort (n=122) that significantly associated with pathologic complete response (p=0.0005), biochemical-recurrence-free survival (p=0.024) and mTOR signaling pathway (p=0.03), suggesting potentially more clinical benefit from mTOR inhibitors in non-responder group. Additionally, genetic and genomic analysis revealed interplay between genetic variants and CMBs on NADT resistance, and provided molecular annotations for CMBs. Overall, prospective clinical implementation of 13-CMB model could assist precision care of PCa patients. Significance We describe a highly accurate CMB model to predict the therapeutic benefit in prostate cancer patients and uncover the complex interplay between genetic variants and CMBs on NADT resistance. Our model relies only on widely available needle biopsy specimens and provides a robust and cost-effective solution for clinical implementation.
Collapse
|
37
|
Song H, Lu T, Han D, Zhang J, Gan L, Xu C, Liu S, Li P, Zhang K, Hu Z, Li H, Li Y, Zhao X, Zhang J, Xing N, Shi C, Wen W, Yang F, Qin W. YAP1 Inhibition Induces Phenotype Switching of Cancer-Associated Fibroblasts to Tumor Suppressive in Prostate Cancer. Cancer Res 2024; 84:3728-3742. [PMID: 39137404 PMCID: PMC11565174 DOI: 10.1158/0008-5472.can-24-0932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/29/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
Prostate cancer rarely responds to immune-checkpoint blockade (ICB) therapies. Cancer-associated fibroblasts (CAF) are critical components of the immunologically "cold" tumor microenvironment and are considered a promising target to enhance the immunotherapy response. In this study, we aimed to reveal the mechanisms regulating CAF plasticity to identify potential strategies to switch CAFs from protumorigenic to antitumor phenotypes and to enhance ICB efficacy in prostate cancer. Integration of four prostate cancer single-cell RNA sequencing datasets defined protumorigenic and antitumor CAFs, and RNA-seq, flow cytometry, and a prostate cancer organoid model demonstrated the functions of two CAF subtypes. Extracellular matrix-associated CAFs (ECM-CAF) promoted collagen deposition and cancer cell progression, and lymphocyte-associated CAFs (Lym-CAF) exhibited an antitumor phenotype and induced the infiltration and activation of CD8+ T cells. YAP1 activity regulated the ECM-CAF phenotype, and YAP1 silencing promoted switching to Lym-CAFs. NF-κB p65 was the core transcription factor in the Lym-CAF subset, and YAP1 inhibited nuclear translocation of p65. Selective depletion of YAP1 in ECM-CAFs in vivo promoted CD8+ T-cell infiltration and activation and enhanced the therapeutic effects of anti-PD-1 treatment on prostate cancer. Overall, this study revealed a mechanism regulating CAF identity in prostate cancer and highlighted a therapeutic strategy for altering the CAF subtype to suppress tumor growth and increase sensitivity to ICB. Significance: YAP1 regulates cancer-associated fibroblast phenotypes and can be targeted to switch cancer-associated fibroblasts from a protumorigenic subtype that promotes extracellular matrix deposition to a tumor-suppressive subtype that stimulates antitumor immunity and immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongtao Song
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Tong Lu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jiayu Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lunbiao Gan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Peng Li
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi’an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhihao Hu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hongji Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaolong Zhao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jingliang Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi’an, China
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
38
|
Aksoy A, Vicdanli S, Artas G. The evaluation of cyclin D1 expression in prostate carcinoma cases. North Clin Istanb 2024; 11:534-540. [PMID: 39650327 PMCID: PMC11622750 DOI: 10.14744/nci.2023.79735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/02/2023] [Accepted: 11/02/2023] [Indexed: 12/11/2024] Open
Abstract
OBJECTIVE Cyclin D1 (CDDN1) is an important protein for mitotic cell cycle advancement through the G1 phase and contributes to the control of the cyclin-dependent kinases CDK4 and CDK6. We evaluated the relationship between CDDN1 expression and clinicopathological features in prostate cancer (PCa) cases and whether CDDN1 could be used as a prognostic biomarker for PCa cases in this study. METHODS This study comprised ninety cases; seventy-five had PCa and fifteen had benign prostatic hypertrophy (BPH) diagnoses (as the control group). The pathological specimens were stained immunohistochemically and categorized as a 'low' (L) or a 'high' (H) group for CDDN1 expression. The cases' clinicopathological features and survival rates were evaluated statistically, within a 95% confidence interval, p<0.05, retrospectively. RESULTS The median follow-up time was 75 (17-96) months, and the median overall survival (OS) was 87 months (CI 95%: 74.74-99.25). While the OS was 66 months (CI 95%: 49.61-82.38) in the H-CDDN1 group, the OS of the L-CDDN1 group was not yet reached. The OS of the L-CDDN1 group was longer in statistical significance (p=0.011). A Cox regression analysis revealed that the levels of CDDN1 expression, the values of lactate dehydrogenase, and post-treatment prostate specific antigen were found to be prognostic factors for OS in PCa cases (p<0.05). CONCLUSION Our results suggest the overexpression of CDDN1 is a potentially useful but poor prognostic biomarker for PCa cases.
Collapse
Affiliation(s)
- Asude Aksoy
- Department of Medical Oncology, University of Health Sciences, Fethi Sekin City Hospital, Elazig, Turkiye
| | - Selcen Vicdanli
- Department of Internal Medicine, Fethi Sekin City Hospital, Elazig, Turkiye
| | - Gokhan Artas
- Department of Pathology, Firat University Faculty of Medicine, Elazig, Turkiye
| |
Collapse
|
39
|
Klümper N, Grünwald V, Hartmann A, Hölzel M, Eckstein M. The Role of Microsatellite Instability/DNA Mismatch Repair Deficiency and Tumor Mutational Burden as Biomarkers in Predicting Response to Immunotherapy in Castration-resistant Prostate Cancer. Eur Urol 2024; 86:388-390. [PMID: 38744632 DOI: 10.1016/j.eururo.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
Large trials of immune checkpoint inhibitors (ICIs) in castration-resistant prostate cancer (CRPC) have mostly failed. Biomarker-selected CRPC patients, especially those with high microsatellite instability (MSI-H), mismatch repair deficiency (dMMR), or elevated tumor mutational burden (TMB), may benefit from single-agent ICIs. Despite their rarity in CRPC (∼2-5%), identification of MSI-H, dMMR, or TMB-H could improve patient selection for immunotherapy.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology, University Hospital Bonn, Bonn, Germany; Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Bonn, Germany; Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany.
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumor Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; EMN Comprehensive Cancer Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Bavarian Center for Cancer Research, Munich, Germany
| | - Michael Hölzel
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Bonn, Germany; Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; EMN Comprehensive Cancer Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Bavarian Center for Cancer Research, Munich, Germany
| |
Collapse
|
40
|
Ploussard G, Baboudjian M, Barret E, Brureau L, Fiard G, Fromont G, Olivier J, Dariane C, Mathieu R, Rozet F, Peyrottes A, Roubaud G, Renard-Penna R, Sargos P, Supiot S, Turpin L, Rouprêt M. French AFU Cancer Committee Guidelines - Update 2024-2026: Prostate cancer - Diagnosis and management of localised disease. THE FRENCH JOURNAL OF UROLOGY 2024; 34:102717. [PMID: 39581668 DOI: 10.1016/j.fjurol.2024.102717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/22/2024] [Accepted: 08/02/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE The aim of the Oncology Committee of the French Urology Association is to propose updated recommendations for the diagnosis and management of localized prostate cancer (PCa). METHODS A systematic review of the literature from 2022 to 2024 was conducted by the CCAFU on the elements of diagnosis and therapeutic management of localized PCa, evaluating references with their level of evidence. RESULTS The recommendations set out the genetics, epidemiology and diagnostic methods of PCa, as well as the concepts of screening and early detection. MRI, the reference imaging test for localized cancer, is recommended before prostate biopsies are performed. Molecular imaging is an option for disease staging. Performing biopsies via the transperineal route reduces the risk of infection. Active surveillance is the standard treatment for tumours with a low risk of progression. Therapeutic methods are described in detail, and recommended according to the clinical situation. CONCLUSION This update of French recommendations should help to improve the management of localized PCa.
Collapse
Affiliation(s)
- Guillaume Ploussard
- Department of Urology, La Croix du Sud Hospital, Quint-Fonsegrives, France; Department of Radiotherapy, Institut Curie, Paris, France.
| | | | - Eric Barret
- Department of Urology, Institut Mutualiste Montsouris, Paris, France
| | - Laurent Brureau
- Department of Urology, CHU de Pointe-à-Pitre, University of Antilles, University of Rennes, Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail (Irset), UMR_S 1085, 97110 Pointe-à-Pitre, Guadeloupe
| | - Gaëlle Fiard
- Department of Urology, Grenoble Alpes University Hospital, Université Grenoble Alpes, CNRS, Grenoble INP, TIMC-IMAG, Grenoble, France
| | | | | | - Charles Dariane
- Department of Urology, Hôpital européen Georges-Pompidou, AP-HP, Paris, France; Paris University, U1151 Inserm, INEM, Necker, Paris, France
| | | | - François Rozet
- Department of Urology, Institut Mutualiste Montsouris, Paris, France
| | | | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, 33000 Bordeaux, France
| | - Raphaële Renard-Penna
- Sorbonne University, AP-HP, Radiology, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonié, 33000 Bordeaux, France
| | - Stéphane Supiot
- Radiotherapy Department, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Léa Turpin
- Nuclear Medicine Department, Hôpital Foch, Suresnes, France
| | - Morgan Rouprêt
- Sorbonne University, GRC 5 Predictive Onco-Uro, AP-HP, Urology, Pitié-Salpêtrière Hospital, 75013 Paris, France
| |
Collapse
|
41
|
Clements AN, Casillas AL, Flores CE, Liou H, Toth RK, Chauhan SS, Sutterby K, Deshmukh SK, Wu S, Xiu J, Farrell A, Radovich M, Nabhan C, Heath EI, McKay RR, Subah N, Centuori S, Wheeler TJ, Cress AE, Rogers GC, Wilson JE, Recio-Boiles A, Warfel NA. Inhibition of PIM kinase in tumor associated macrophages suppresses inflammasome activation and sensitizes prostate cancer to immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.618756. [PMID: 39484473 PMCID: PMC11526960 DOI: 10.1101/2024.10.21.618756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Immunotherapy has changed the treatment paradigm for many types of cancer, but immune checkpoint inhibitors (ICIs) have not shown benefit in prostate cancer (PCa). Chronic inflammation contributes to the immunosuppressive prostate tumor microenvironment (TME) and is associated with poor response to ICIs. The primary source of inflammatory cytokine production is the inflammasome. Here, we identify PIM kinases as important regulators of inflammasome activation in tumor associated macrophages (TAMs). Analysis of clinical data from a cohort of treatment naïve, hormone responsive PCa patients revealed that tumors from patients with high PIM1/2/3 display an immunosuppressive TME characterized by high inflammation (IL-1β and TNFα) and a high density of repressive immune cells, most notably TAMs. Strikingly, macrophage-specific knockout of PIM reduced tumor growth in syngeneic models of prostate cancer. Transcriptional analyses indicate that eliminating PIM from macrophages enhanced the adaptive immune response and increased cytotoxic immune cells. Combined treatment with PIM inhibitors and ICIs synergistically reduced tumor growth. Immune profiling revealed that PIM inhibitors sensitized PCa tumors to ICIs by increasing tumor suppressive TAMs and increasing the activation of cytotoxic T cells. Collectively, our data implicate macrophage PIM as a driver of inflammation that limits the potency of ICIs and provides preclinical evidence that PIM inhibitors are an effective strategy to improve the efficacy of immunotherapy in prostate cancer.
Collapse
|
42
|
Hansen SB, Unal B, Kuzu OF, Saatcioglu F. Immunological facets of prostate cancer and the potential of immune checkpoint inhibition in disease management. Theranostics 2024; 14:6913-6934. [PMID: 39629128 PMCID: PMC11610136 DOI: 10.7150/thno.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/27/2024] [Indexed: 12/06/2024] Open
Abstract
Prostate cancer (PCa) is the most common non-cutaneous cancer in men and a major cause of cancer-related deaths. Whereas localized PCa can be cured by surgery and radiotherapy, metastatic disease can be treated, but is not curable. Inhibition of androgen signaling remains the main therapeutic intervention for treatment of metastatic PCa, in addition to chemotherapy, radionuclide therapy and emerging targeted therapies. Although initial responses are favorable, resistance to these therapies invariably arise with development of castration resistant PCa (CRPC) and lethal phenotypes. Recent findings have implicated the crosstalk between PCa cells and the tumor microenvironment (TME) as a key factor for disease progression and metastasis, and the immune system is becoming an increasingly attractive target for therapy. Given the striking success of immune checkpoint inhibitors (ICIs) in various cancer types, preclinical and clinical studies have begun to explore their potential in PCa. It has become clear that the PCa TME is largely immunosuppressive, and ICI therapy does not have efficacy for PCa. Intense effort is therefore being made in the field to understand the mechanisms of suppression and to turn the immunosuppressive TME into an immune active one that would enable ICI efficacy. Herein we examine this recent body of knowledge and how the mutational landscape of PCa integrates with an immunosuppressive TME to circumvent ICI-mediated T-cell activity and tumor killing. We then review the emerging potential success of combinatorial ICI approaches, utility of careful patient selection, and potential novel strategies to improve the efficacy of ICI for PCa therapy.
Collapse
Affiliation(s)
| | - Bilal Unal
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Omer Faruk Kuzu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
43
|
McManus HD, Dorff T, Morgans AK, Sartor O, Shore N, Armstrong AJ. Navigating therapeutic sequencing in the metastatic castration-resistant prostate cancer patient journey. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00906-z. [PMID: 39420184 PMCID: PMC12003708 DOI: 10.1038/s41391-024-00906-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Novel therapies for metastatic castration-resistant prostate cancer (mCRPC) have improved patient outcomes. However, there is uncertainty on the optimal selection of therapeutic agents for subsequent lines of therapy. METHODS We conducted a comprehensive review of published evidence from pivotal clinical trials and recent guidelines for the treatment of mCRPC. We further identify gaps in knowledge and areas for future research. RESULTS Key considerations to help guide treatment selection for patients with mCRPC include personal treatment history, individual clinical characteristics, symptoms, prognosis, availability of clinical trials, and other patient-specific factors. Genetic testing and prostate-specific membrane antigen-targeted imaging are important tools to evaluate candidacy for newer therapeutic options such as poly (ADP-ribose) polymerase inhibitors, alone or in combination with androgen receptor pathway inhibitors, and [177Lu]Lu-PSMA-617. CONCLUSION This article provides an overview of the evolving treatment landscape of mCRPC, discussing guideline-recommended treatment options and data from key clinical trials, while highlighting ongoing trials that may impact the future treatment landscape. Recommendations for optimal treatment sequencing based on individual patient factors are provided.
Collapse
Affiliation(s)
- Hannah D McManus
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Duke University, Durham, NC, USA
| | - Tanya Dorff
- City of Hope National Cancer Center, Duarte, CA, USA
| | | | | | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Duke University, Durham, NC, USA.
| |
Collapse
|
44
|
Yu J, Zhou K, Wang J, Mao L. Preliminary Efficacy, Tolerability, and Safety Analysis of Darolutamide for Metastatic Castration-Resistant Prostate Cancer: A Single-Center, Open-Label Study. Urol Int 2024; 109:167-174. [PMID: 39406197 DOI: 10.1159/000541929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION Darolutamide is a structurally unique second-generation androgen receptor antagonist that has been approved for indications in patients with non-metastatic castration-resistant prostate cancer (nmCRPC) and metastatic hormone-sensitive prostate cancer (mHSPC). The aim was to assess the efficacy and safety of Darolutamide for mCRPC. METHODS In this single-center, open-label study, patients with previously untreated mCRPC were enrolled and received androgen deprivation therapy (goserelin acetate 3.6 mg every 28 days) and docetaxel (75 mg per square meter of body surface area every 21 days) with Denosumab (120 mg every 28 days) for bone metastases, Darolutamide (300 mg orally twice daily) in the experimental group, and the control group received the corresponding of placebo. Serum PSA changes were detected and recorded, and imaging changes and adverse events (AEs) were evaluated. The primary endpoints were safety, tolerability, and antitumor efficacy, and the second endpoint was radiographic progression-free survival (rPFS). RESULTS Thirty-seven patients with mCRPC were enrolled. The median time to PSA50 in the Darolutamide group was 1.5 months (95% CI: 0.2619-0.9545), significantly lower than that in the placebo group (3.0 months [95% CI: 1.048-3.818], p = 0.0259). The median time to PSA90 in the experimental group was 4 months (95% CI: 0.3094-1.437), 2 months shorter than that in the placebo group (6.0 months [95% CI: 0.6961-3.232]). With the median follow-up of 6 months, the median decrease in serum PSA was -81.8% (range -60.4 to -99.9%) in the Darolutamide group and -69.4% (range -50.3 to -89.6%) in the placebo group. Tumor-related pain and AEs were not increased, and the median rPFS was not reached. CONCLUSIONS The combination of Darolutamide and docetaxel was well tolerated with more clinically beneficial than docetaxel alone in previously untreated mCRPC. Darolutamide rapidly reduced PSA levels and prolonged rPFS and did not increase the incidence of AEs.
Collapse
Affiliation(s)
- JunJie Yu
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,
| | - KaiChen Zhou
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - JunQi Wang
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - LiJun Mao
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
45
|
Chen L, Xu YX, Wang YS, Ren YY, Dong XM, Wu P, Xie T, Zhang Q, Zhou JL. Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer 2024; 23:229. [PMID: 39395984 PMCID: PMC11470719 DOI: 10.1186/s12943-024-02137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent malignancies in males worldwide. Increasing research attention has focused on the PCa microenvironment, which plays a crucial role in tumor progression and therapy resistance. This review aims to provide a comprehensive overview of the key components of the PCa microenvironment, including immune cells, vascular systems, stromal cells, and microbiota, and explore their implications for diagnosis and treatment. METHODS Keywords such as "prostate cancer", "tumor microenvironment", "immune cells", "vascular system", "stromal cells", and "microbiota" were used for literature retrieval through online databases including PubMed and Web of Science. Studies related to the PCa microenvironment were selected, with a particular focus on those discussing the roles of immune cells, vascular systems, stromal cells, and microbiota in the development, progression, and treatment of PCa. The selection criteria prioritized peer-reviewed articles published in the last five years, aiming to summarize and analyze the latest research advancements and clinical relevance regarding the PCa microenvironment. RESULTS The PCa microenvironment is highly complex and dynamic, with immune cells contributing to immunosuppressive conditions, stromal cells promoting tumor growth, and microbiota potentially affecting androgen metabolism. Vascular systems support angiogenesis, which fosters tumor expansion. Understanding these components offers insight into the mechanisms driving PCa progression and opens avenues for novel therapeutic strategies targeting the tumor microenvironment. CONCLUSIONS A deeper understanding of the PCa microenvironment is crucial for advancing diagnostic techniques and developing precision therapies. This review highlights the potential of targeting the microenvironment to improve patient outcomes, emphasizing its significance in the broader context of PCa research and treatment innovation.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying-Ying Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xue-Man Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Pu Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Qi Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China.
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
46
|
Saini KS, Somara S, Ko HC, Thatai P, Quintana A, Wallen ZD, Green MF, Mehrotra R, McGuigan S, Pang L, Das S, Yadav K, Neric D, Cantini L, Joshi C, Iwamoto K, Dubbewar S, Vidal L, Chico I, Severson E, Lorini L, Badve S, Bossi P. Biomarkers in head and neck squamous cell carcinoma: unraveling the path to precision immunotherapy. Front Oncol 2024; 14:1473706. [PMID: 39439946 PMCID: PMC11493772 DOI: 10.3389/fonc.2024.1473706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Recent strides in understanding the molecular underpinnings of head and neck cancers have sparked considerable interest in identifying precise biomarkers that can enhance prognostication and enable personalized treatment strategies. Immunotherapy has particularly revolutionized the therapeutic landscape for head and neck squamous cell carcinoma, offering new avenues for treatment. This review comprehensively examines the application and limitations of the established and emerging/novel biomarkers for head and neck squamous cell carcinoma. Established biomarkers, including well-characterized genetic mutations, protein expressions, and clinical factors, have been extensively studied and validated in clinical practice. Novel biomarkers identified through molecular analyses, including novel genetic alterations, immune-related markers, and molecular signatures, are currently being investigated and validated in preclinical and clinical settings. Biomarkers hold the potential to deepen our understanding of head and neck squamous cell carcinoma biology and guide therapeutic strategies. The evolving paradigm of predictive biomarkers facilitates the study of individual responses to specific treatments, including targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Kamal S. Saini
- Fortrea Inc., Durham, NC, United States
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | | - Soma Das
- Fortrea Inc., Durham, NC, United States
| | - Kavita Yadav
- George Institute for Global Health, New Delhi, India
| | | | | | | | | | | | | | | | | | - Luigi Lorini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Cancer Centre, Milan, Italy
| | - Sunil Badve
- Emory University, Atlanta, GA, United States
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Cancer Centre, Milan, Italy
- Università degli Studi di Brescia, Brescia, Italy
| |
Collapse
|
47
|
Tang H, Li YX, Lian JJ, Ng HY, Wang SSY. Personalized treatment using predictive biomarkers in solid organ malignancies: A review. TUMORI JOURNAL 2024; 110:386-404. [PMID: 39091157 DOI: 10.1177/03008916241261484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In recent years, the influence of specific biomarkers in the diagnosis and prognosis of solid organ malignancies has been increasingly prominent. The relevance of the use of predictive biomarkers, which predict cancer response to specific forms of treatment provided, is playing a more significant role than ever before, as it affects diagnosis and initiation of treatment, monitoring for efficacy and side effects of treatment, and adjustment in treatment regimen in the long term. In the current review, we explored the use of predictive biomarkers in the treatment of solid organ malignancies, including common cancers such as colorectal cancer, breast cancer, lung cancer, prostate cancer, and cancers associated with high mortalities, such as pancreatic cancer, liver cancer, kidney cancer and cancers of the central nervous system. We additionally analyzed the goals and types of personalized treatment using predictive biomarkers, and the management of various types of solid organ malignancies using predictive biomarkers and their relative efficacies so far in the clinical settings.
Collapse
|
48
|
Abu Hijlih R, Sharaf B, Salah S, Bani Hani H, Nielsen SM, Heald B, Esplin ED, Ghanem R, Alzibdeh A, Al-Batsh T, Al-Masri Y, Abdel-Razeq H. Patterns and Frequency of Pathogenic Germline Variants Among Prostate Cancer Patients Utilizing Multi-Gene Panel Genetic Testing. World J Oncol 2024; 15:801-808. [PMID: 39328335 PMCID: PMC11424115 DOI: 10.14740/wjon1896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/21/2024] [Indexed: 09/28/2024] Open
Abstract
Background Germline genetic testing (GGT) has significant implications in the management of patients with prostate cancer (PCa). Herein, we report on patterns and frequency of pathogenic/likely pathogenic germline variants (P/LPGVs) among newly diagnosed Arab patients with PCa. Methods Patients meeting the National Comprehensive Cancer Network (NCCN) eligibility criteria for GGT were offered a 19-gene PCa panel or an expanded 84-gene multi-cancer panel. Results During the study period, 231 patients were enrolled; 107 (46.3%) had metastatic disease at diagnosis. In total, 17 P/LPGVs were detected in 17 patients (7.4%). Among the 113 (48.9%) patients who underwent GGT with the 19-gene panel, eight (7.1%) had P/LPGVs, compared to nine (7.6%) of the 118 (51.1%) who did GGT through the expanded 84-gene panel (P = 0.88). Variant of uncertain significance (VUS) rate was higher (n = 73, 61.9%) among the group who underwent expanded 84-gene panel testing compared to those who underwent the 19-gene PCa panel (n = 35, 30.9%) (P = 0.001). P/LPGVs in DNA damage repair (DDR) genes, most frequently BRCA2, CHEK2 and TP53, were the most common P/LPGVs findings. Conclusion This study is the first to characterize the germline genetic profile of an Arab population with PCa. All detected P/LPGVs were potentially actionable, with most variants able to be detected with a PCa-specific panel.
Collapse
Affiliation(s)
- Ramiz Abu Hijlih
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Baha Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Samer Salah
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | | | | | | | - Rami Ghanem
- Department of Surgery, King Hussein Cancer Center, Amman, Jordan
| | - Abdulla Alzibdeh
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Tamer Al-Batsh
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Yosra Al-Masri
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
- School of Medicine, the University of Jordan, Amman, Jordan
| |
Collapse
|
49
|
Liu D, Wang L, Guo Y. Advances in and prospects of immunotherapy for prostate cancer. Cancer Lett 2024; 601:217155. [PMID: 39127338 DOI: 10.1016/j.canlet.2024.217155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/07/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has shown promising therapeutic effects in hematological malignancies and certain solid tumors and has emerged as a critical and highly potential treatment modality for cancer. However, prostate cancer falls under the category of immune-resistant cold tumors, for which immunotherapy exhibits limited efficacy in patients with solid tumors. Thus, it is important to gain a deeper understanding of the tumor microenvironment in prostate cancer to facilitate immune system activation and overcome immune suppression to advance immunotherapy for prostate cancer. In this review, we discuss the immunosuppressive microenvironment of prostate cancer, which is characterized by the presence of few tumor-infiltrating lymphocytes, abundant immunosuppressive cells, low immunogenicity, and a noninflammatory phenotype, which significantly influences the efficacy of immunotherapy for prostate cancer. Immunotherapy is mainly achieved by activating the host immune system and overcoming immunosuppression. In this regard, we summarize the therapeutic advances in immune checkpoint blockade, immunogenic cell death, reversal of the immunosuppressive tumor microenvironment, tumor vaccines, immune adjuvants, chimeric antigen receptor T-cell therapy, and overcoming penetration barriers in prostate cancer, with the aim of providing novel research insights and approaches to enhance the effectiveness of immunotherapy for prostate cancer.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
50
|
Porter LH, Harrison SG, Risbridger GP, Lister N, Taylor RA. Left out in the cold: Moving beyond hormonal therapy for the treatment of immunologically cold prostate cancer with CAR T cell immunotherapies. J Steroid Biochem Mol Biol 2024; 243:106571. [PMID: 38909866 DOI: 10.1016/j.jsbmb.2024.106571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
Prostate cancer is primarily hormone-dependent, and medical treatments have focused on inhibiting androgen biosynthesis or signaling through various approaches. Despite significant advances with the introduction of androgen receptor signalling inhibitors (ARSIs), patients continue to progress to castration-resistant prostate cancer (CRPC), highlighting the need for targeted therapies that extend beyond hormonal blockade. Chimeric Antigen Receptor (CAR) T cells and other engineered immune cells represent a new generation of adoptive cellular therapies. While these therapies have significantly enhanced outcomes for patients with hematological malignancies, ongoing research is exploring the broader use of CAR T therapy in solid tumors, including advanced prostate cancer. In general, CAR T cell therapies are less effective against solid cancers with the immunosuppressive tumor microenvironment hindering T cell infiltration, activation and cytotoxicity following antigen recognition. In addition, inherent tumor heterogeneity exists in patients with advanced prostate cancer that may prevent durable therapeutic responses using single-target agents. These barriers must be overcome to inform clinical trial design and improve treatment efficacy. In this review, we discuss the innovative and rationally designed strategies under investigation to improve the clinical translation of cellular immunotherapy in prostate cancer and maximise therapeutic outcomes for these patients.
Collapse
Affiliation(s)
- L H Porter
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - S G Harrison
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - G P Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cabrini Institute, Cabrini Health, Malvern, VIC 3144, Australia
| | - Natalie Lister
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - R A Taylor
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cabrini Institute, Cabrini Health, Malvern, VIC 3144, Australia; Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|