101
|
Serritella AV, Taylor A, Haffner MC, Abida W, Bryce A, Karsh LI, Tagawa ST, Twardowski P, Armstrong AJ, Lang JM. Therapeutic implications of homologous repair deficiency testing in patients with prostate cancer (Part 2 of 2). Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00887-z. [PMID: 39333696 DOI: 10.1038/s41391-024-00887-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND/OBJECTIVES Unfortunately, not all metastatic castration-resistant prostate cancer (mCRPC) patients receive available life-prolonging systemic therapies, emphasizing the need to optimize mCRPC treatment selections. Better guidelines are necessary to determine genetic testing for prostate cancer. SUBJECTS/METHODS In this two-part expert opinion-based guide, we provide an expert consensus opinion on the utilization of germline and somatic testing to detect HRR alterations in patients with mCRPC. This guide was developed by a multidisciplinary expert panel that convened in 2023-2024, including representatives from medical oncology, urology, radiation oncology, pathology, medical genomics, and basic science. RESULTS/CONCLUSIONS In this second part, we highlight how genetic testing can lead to improved, life-prolonging mCRPC therapeutic strategies based on a review of the recent phase III trials and subsequent regulatory approvals for PARP inhibitors in mCRPC.
Collapse
Affiliation(s)
| | - Amy Taylor
- University of Wisconsin, Madison, WI, USA
| | | | - Wassim Abida
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
102
|
Sabater A, Sanchis P, Seniuk R, Pascual G, Anselmino N, Alonso D, Cayol F, Vazquez E, Marti M, Cotignola J, Toro A, Labanca E, Bizzotto J, Gueron G. Unmasking Neuroendocrine Prostate Cancer with a Machine Learning-Driven 7-Gene Stemness Signature that Predicts Progression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.24.24314303. [PMID: 39399052 PMCID: PMC11469473 DOI: 10.1101/2024.09.24.24314303] [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: 10/15/2024]
Abstract
Prostate cancer (PCa) poses a significant global health challenge, particularly due to its progression into aggressive forms like neuroendocrine prostate cancer (NEPC). This study developed and validated a stemness-associated gene signature using advanced machine learning techniques, including Random Forest and Lasso regression, applied to large-scale transcriptomic datasets. The resulting 7-gene signature (KMT5C, MEN1, TYMS, IRF5, DNMT3B, CDC25B and DPP4) was validated across independent cohorts and patient-derived xenograft (PDX) models. The signature demonstrated strong prognostic value for progression-free, disease-free, relapse-free, metastasis-free, and overall survival. Importantly, the signature not only identified specific NEPC subtypes, such as large-cell neuroendocrine carcinoma, which is associated with very poor outcomes, but also predicted a poor prognosis for PCa cases that exhibit this molecular signature, even when they were not histopathologically classified as NEPC. This dual prognostic and classifier capability makes the 7-gene signature a robust tool for personalized medicine, providing a valuable resource for predicting disease progression and guiding treatment strategies in PCa management.
Collapse
Affiliation(s)
- Agustina Sabater
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Pablo Sanchis
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Rocio Seniuk
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Gaston Pascual
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Nicolas Anselmino
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daniel Alonso
- Centro de Oncología Molecular y Traslacional y Plataforma de Servicios Biotecnológicos, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal B1876BXD, Argentina
| | - Federico Cayol
- Sector de Oncología Clínica, Hospital Italiano de Buenos Aires, Buenos Aires, C1199ABB, Argentina
| | - Elba Vazquez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Marcelo Marti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Javier Cotignola
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Ayelen Toro
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juan Bizzotto
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Geraldine Gueron
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| |
Collapse
|
103
|
Hachem S, Yehya A, El Masri J, Mavingire N, Johnson JR, Dwead AM, Kattour N, Bouchi Y, Kobeissy F, Rais-Bahrami S, Mechref Y, Abou-Kheir W, Woods-Burnham L. Contemporary Update on Clinical and Experimental Prostate Cancer Biomarkers: A Multi-Omics-Focused Approach to Detection and Risk Stratification. BIOLOGY 2024; 13:762. [PMID: 39452071 PMCID: PMC11504278 DOI: 10.3390/biology13100762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024]
Abstract
Prostate cancer remains a significant health challenge, being the most prevalent non-cutaneous cancer in men worldwide. This review discusses the critical advancements in biomarker discovery using single-omics and multi-omics approaches. Multi-omics, integrating genomic, transcriptomic, proteomic, metabolomic, and epigenomic data, offers a comprehensive understanding of the molecular heterogeneity of prostate cancer, leading to the identification of novel biomarkers and therapeutic targets. This holistic approach not only enhances the specificity and sensitivity of prostate cancer detection but also supports the development of personalized treatment strategies. Key studies highlighted include the identification of novel genes, genetic mutations, peptides, metabolites, and potential biomarkers through multi-omics analyses, which have shown promise in improving prostate cancer management. The integration of multi-omics in clinical practice can potentially revolutionize prostate cancer prognosis and treatment, paving the way for precision medicine. This review underscores the importance of continued research and the application of multi-omics to overcome current challenges in prostate cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Sana Hachem
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Amani Yehya
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Jad El Masri
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Nicole Mavingire
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.)
| | - Jabril R. Johnson
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Abdulrahman M. Dwead
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.)
| | - Naim Kattour
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Yazan Bouchi
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Firas Kobeissy
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Soroush Rais-Bahrami
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Radiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Leanne Woods-Burnham
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.)
| |
Collapse
|
104
|
Hall R, Bancroft E, Pashayan N, Kote-Jarai Z, Eeles RA. Genetics of prostate cancer: a review of latest evidence. J Med Genet 2024; 61:915-926. [PMID: 39137963 DOI: 10.1136/jmg-2024-109845] [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: 01/02/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024]
Abstract
Prostate cancer (PrCa) is a largely heritable and polygenic disease. It is the most common cancer in people with prostates (PwPs) in Europe and the USA, including in PwPs of African descent. In the UK in 2020, 52% of all cancers were diagnosed at stage I or II. The National Health Service (NHS) long-term plan is to increase this to 75% by 2028, to reduce absolute incidence of late-stage disease. In the absence of a UK PrCa screening programme, we should explore how to identify those at increased risk of clinically significant PrCa.Incorporating genomics into the PrCa screening, diagnostic and treatment pathway has huge potential for transforming patient care. Genomics can increase efficiency of PrCa screening by focusing on those with genetic predisposition to cancer-which when combined with risk factors such as age and ethnicity, can be used for risk stratification in risk-based screening (RBS) programmes. The goal of RBS is to facilitate early diagnosis of clinically significant PrCa and reduce overdiagnosis/overtreatment in those unlikely to experience PrCa-related symptoms in their lifetime. Genetic testing can guide PrCa management, by identifying those at risk of lethal PrCa and enabling access to novel targeted therapies.PrCa is curable if diagnosed below stage III when most people do not experience symptoms. RBS using genetic profiling could be key here if we could show better survival outcomes (or reduction in cancer-specific mortality accounting for lead-time bias), in addition to more cost efficiency than age-based screening alone. Furthermore, PrCa outcomes in underserved communities could be optimised if genetic testing was accessible, minimising health disparities.
Collapse
Affiliation(s)
- Rose Hall
- The Royal Marsden NHS Foundation Trust, London, UK
- Institute for Cancer Research, London, UK
| | | | | | | | - Rosalind A Eeles
- The Royal Marsden NHS Foundation Trust, London, UK
- Institute for Cancer Research, London, UK
| |
Collapse
|
105
|
Woo BJ, Moussavi-Baygi R, Karner H, Karimzadeh M, Yousefi H, Lee S, Garcia K, Joshi T, Yin K, Navickas A, Gilbert LA, Wang B, Asgharian H, Feng FY, Goodarzi H. Integrative identification of non-coding regulatory regions driving metastatic prostate cancer. Cell Rep 2024; 43:114764. [PMID: 39276353 PMCID: PMC11466230 DOI: 10.1016/j.celrep.2024.114764] [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: 06/13/2023] [Revised: 07/08/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024] Open
Abstract
Large-scale sequencing efforts have been undertaken to understand the mutational landscape of the coding genome. However, the vast majority of variants occur within non-coding genomic regions. We designed an integrative computational and experimental framework to identify recurrently mutated non-coding regulatory regions that drive tumor progression. Applying this framework to sequencing data from a large prostate cancer patient cohort revealed a large set of candidate drivers. We used (1) in silico analyses, (2) massively parallel reporter assays, and (3) in vivo CRISPR interference screens to systematically validate metastatic castration-resistant prostate cancer (mCRPC) drivers. One identified enhancer region, GH22I030351, acts on a bidirectional promoter to simultaneously modulate expression of the U2-associated splicing factor SF3A1 and chromosomal protein CCDC157. SF3A1 and CCDC157 promote tumor growth in vivo. We nominated a number of transcription factors, notably SOX6, to regulate expression of SF3A1 and CCDC157. Our integrative approach enables the systematic detection of non-coding regulatory regions that drive human cancers.
Collapse
Affiliation(s)
- Brian J Woo
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Arc Institute, Palo Alto, CA 94305, USA
| | - Ruhollah Moussavi-Baygi
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Heather Karner
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Arc Institute, Palo Alto, CA 94305, USA
| | - Mehran Karimzadeh
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Vector Institute, Toronto, ON, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Arc Institute, Palo Alto, CA 94305, USA
| | - Hassan Yousefi
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Arc Institute, Palo Alto, CA 94305, USA
| | - Sean Lee
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Arc Institute, Palo Alto, CA 94305, USA
| | - Kristle Garcia
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Tanvi Joshi
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Keyi Yin
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Albertas Navickas
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Luke A Gilbert
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Arc Institute, Palo Alto, CA 94305, USA
| | - Bo Wang
- Vector Institute, Toronto, ON, Canada; Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Hosseinali Asgharian
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Felix Y Feng
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA.
| | - Hani Goodarzi
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Arc Institute, Palo Alto, CA 94305, USA; Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
106
|
Yu EM, Patel I, Hwang MW, Polani F, Aragon-Ching JB. The Rapidly Evolving Treatment Landscape of Metastatic Hormone-Sensitive Prostate Cancer. Clin Med Insights Oncol 2024; 18:11795549241277181. [PMID: 39323979 PMCID: PMC11423369 DOI: 10.1177/11795549241277181] [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: 02/14/2024] [Accepted: 08/04/2024] [Indexed: 09/27/2024] Open
Abstract
The management of metastatic hormone-sensitive prostate cancer (mHSPC) or castration-sensitive prostate cancer (mCSPC) has become increasingly complex with the tremendous progress that has been made in this space within the past few decades. In the early days of androgen deprivation therapy (ADT), ADT monotherapy was the mainstay for treatment of advanced prostate cancer. However, novel hormone therapies in the form of androgen receptor pathway inhibitors (ARPI) have emerged; vaccine therapy, chemotherapy with docetaxel and cabazitaxel, and radioactive ligands have shaped the treatment of metastatic prostate cancer in the last decade. Following the initial approval of several drugs for use in metastatic castration-resistant prostate cancer (mCRPC) in combination with primary ADT, these agents were studied and subsequently approved for use in mCSPC. Therefore, ADT monotherapy no longer constitutes an optimal therapeutic option for otherwise fit patients who present with mCSPC. We focus on the treatment of first-line de novo mHSPC or mCSPC and explore frontline doublet and triplet therapy and the pivotal trials that led to their United States Food and Drug Administration approval.
Collapse
Affiliation(s)
- Eun-Mi Yu
- GU Medical Oncology, Inova Schar Cancer Institute, Fairfax, VA, USA
| | - Ishan Patel
- Division of Hematology and Oncology, Department of Medicine, Inova Schar Cancer Institute, Fairfax, VA, USA
| | - Min Woo Hwang
- Department of Internal Medicine, Inova Fairfax Hospital, Fairfax, VA, USA
| | - Faran Polani
- Division of Hematology and Oncology, Department of Medicine, Inova Schar Cancer Institute, Fairfax, VA, USA
| | - Jeanny B Aragon-Ching
- GU Medical Oncology, Inova Schar Cancer Institute, Fairfax, VA, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
107
|
Antolini E, Filosa A, Santoni M, Antaldi E, Bartoli E, Sierchio L, Giantomassi F, Mandolesi A, Goteri G. Internal Overview of Prostatic Cancer Cases and Quality of BRCA1 and BRCA2 NGS Data from the FFPE Tissue. Diagnostics (Basel) 2024; 14:2067. [PMID: 39335746 PMCID: PMC11431729 DOI: 10.3390/diagnostics14182067] [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: 07/26/2024] [Revised: 08/16/2024] [Accepted: 09/01/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Comprehensive genomic profiling (CGP) has gained an important role in patients with advanced prostate cancer following the introduction of PARP inhibitors in daily clinical practice. Here, we report an overview of CGP results, specifically of BRCA1 and BRCA2 HRD-repair system genes, from patients with prostate cancer analyzed in our institution, and we compare our results with those available from more recent scientific literature. Methods: The study cohort consisted of 70 patients. Somatic DNA was extracted from Formalin-Fixed Paraffin-Embedded (FFPE) tissue using a MagCore Genomic DNA FFPE One-Step Kit for MagCore System. The DNA was quantified by EasyPGX® Real-Time qPCR and EasyPGX® Analysis Software (version 4.0.13). Tissue somatic DNA libraries were prepared with Myriapod® NGS BRCA1-2 panel-NG035 and sequenced in a Mi-Seq® System. The sequence alignment in hg19 and the variant calling were performed using Myriapod® NGS Data Analysis Software version 5.0.8 NG900-SW 5.0.8 with a software detection limit (LoD) of 95%. Variants with a coverage of 500 and VAF% ≥ 5 were evaluated. Results: Tumor tissue NGS was unsuccessful in 46/70 patients (66%). Mutations of the BRCA2 gene were detected in 4 of the samples: (1) BRCA2 ex10 c.1244A>G p.His415Arg VAF = 51.03%; (2) BRCA2 ex11 c.5946delT p.Ser1982fs VAF = 72.1%; (3) BRCA2 ex11 c.3302A>G p.His1101Arg VAF = 52.9%; and (4) BRCA2 ex11 c.3195_3198delTAAT p.Asn1066fs VAF = 51.1%. Conclusions: The results from our internal overview seem to support the data and to confirm the performance of the technical issues reported in the literature. Considering the advanced age of our patients, with 84% of men over the age of 65, the application of alternative and less invasive procedures such as liquid biopsy, could be a more suitable solution for some cases.
Collapse
Affiliation(s)
- Enrica Antolini
- Department of Biomedical Science and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Alessandra Filosa
- Department of Biomedical Science and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy
- Anatomic Pathology Unit, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Elena Antaldi
- Anatomic Pathology Unit, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Elisa Bartoli
- Anatomic Pathology Unit, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Lidia Sierchio
- Anatomic Pathology Unit, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Federica Giantomassi
- Department of Biomedical Science and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Alessandra Mandolesi
- Anatomic Pathology Unit, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Gaia Goteri
- Department of Biomedical Science and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy
- Anatomic Pathology Unit, Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| |
Collapse
|
108
|
Thapa B, De Sarkar N, Giri S, Sharma K, Kim M, Kilari D. Integrating PARP Inhibitors in mCRPC Therapy: Current Strategies and Emerging Trends. Cancer Manag Res 2024; 16:1267-1283. [PMID: 39308935 PMCID: PMC11416116 DOI: 10.2147/cmar.s411023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Metastatic castrate-resistant prostate cancer (mCRPC) is associated with poor prognosis. DNA damage response (DDR) genes are commonly altered in mCRPC rendering them as promising therapeutic targets. Poly (ADP ribose) polymerase inhibitors (PARPi) demonstrated antitumor activity in mCRPC patients with DDR gene mutations through synthetic lethality. Multiple clinical trials with PARPi monotherapy exhibited encouraging clinical outcomes in selected patients with mCRPC. More recently, three Phase III randomized clinical trials (RCTs) combining PARPi with androgen receptor signaling inhibitors (ARSIs) demonstrated improved antitumor activity compared to ARSI monotherapy in mCRPC patients as the first-line therapy. Clinical benefit was more pronounced in patients harboring DDR alterations, specifically BRCA1/2. Interestingly, antitumor activity was also observed irrespective of DDR gene mutations, highlighting BRCAness phenotype with androgen receptor blockade resulting in synergistic activity between ARSIs and PARPi. In this review, we discuss the clinical efficacy and safety data of the combination of PARPi plus ARSI in all Phase 3 randomized controlled trials (RCTs), emphasizing strategies for patient selection and highlighting emerging trends based on clinical trial data.
Collapse
Affiliation(s)
- Bicky Thapa
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Navonil De Sarkar
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
- Data Science Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Subhajit Giri
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Komal Sharma
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
- Data Science Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mingee Kim
- School of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Kilari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
109
|
Shrestha R, Chesner LN, Zhang M, Zhou S, Foye A, Lundberg A, Weinstein AS, Sjöström M, Zhu X, Moreno-Rodriguez T, Li H, Alumkal JJ, Aggarwal R, Small EJ, Lupien M, Quigley DA, Feng FY. An Atlas of Accessible Chromatin in Advanced Prostate Cancer Reveals the Epigenetic Evolution during Tumor Progression. Cancer Res 2024; 84:3086-3100. [PMID: 38990734 DOI: 10.1158/0008-5472.can-24-0890] [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: 03/15/2024] [Revised: 04/16/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is a lethal disease that resists therapy targeting androgen signaling, the primary driver of prostate cancer. mCRPC resists androgen receptor (AR) inhibitors by amplifying AR signaling or by evolving into therapy-resistant subtypes that do not depend on AR. Elucidation of the epigenetic underpinnings of these subtypes could provide important insights into the drivers of therapy resistance. In this study, we produced chromatin accessibility maps linked to the binding of lineage-specific transcription factors (TF) by performing assay for transposase-accessible chromatin sequencing on 70 mCRPC tissue biopsies integrated with transcriptome and whole-genome sequencing. mCRPC had a distinct global chromatin accessibility profile linked to AR function. Analysis of TF occupancy across accessible chromatin revealed 203 TFs associated with mCRPC subtypes. Notably, ZNF263 was identified as a putative prostate cancer TF with a significant impact on gene activity in the double-negative subtype (AR- neuroendocrine-), potentially activating MYC targets. Overall, this analysis of chromatin accessibility in mCRPC provides valuable insights into epigenetic changes that occur during progression to mCRPC. Significance: Integration of a large cohort of transcriptome, whole-genome, and ATAC sequencing characterizes the chromatin accessibility changes in advanced prostate cancer and identifies therapy-resistant prostate cancer subtype-specific transcription factors that modulate oncogenic programs.
Collapse
Affiliation(s)
- Raunak Shrestha
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Lisa N Chesner
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Meng Zhang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Stanley Zhou
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Adam Foye
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Arian Lundberg
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
- The Institute of Cancer Research and The Royal Marsden Hospital, London, United Kingdom
| | - Alana S Weinstein
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Martin Sjöström
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Xiaolin Zhu
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Thaidy Moreno-Rodriguez
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
| | - Haolong Li
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Joshi J Alumkal
- Division of Hematology and Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, California
| | - Felix Y Feng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
110
|
Li Q, Wang Y, Chen J, Zeng K, Wang C, Guo X, Hu Z, Hu J, Liu B, Xiao J, Zhou P. Machine learning based androgen receptor regulatory gene-related random forest survival model for precise treatment decision in prostate cancer. Heliyon 2024; 10:e37256. [PMID: 39296076 PMCID: PMC11407950 DOI: 10.1016/j.heliyon.2024.e37256] [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: 06/19/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Background It has been demonstrated that aberrant androgen receptor (AR) signaling contributes to the pathogenesis of prostate cancer (PCa). To date, the most efficacious strategy for the treatment of PCa remains to target the AR signaling axis. However, numerous PCa patients still face the issue of overtreatment or undertreatment. The establishment of a precise risk prediction model is urgently needed to distinguish patients with high-risk and select appropriate treatment modalities. Methods In this study, a consensus AR regulatory gene-related signature (ARS) was developed by integrating a total of 101 algorithm combinations of 10 machine learning algorithms. We evaluated the value of ARS in predicting patient prognosis and the therapeutic effects of the various treatments. Additionally, we conducted a screening of therapeutic targets and agents for high-risk patients, followed by the verification in vitro and in vivo. Results ARS was an independent risk factor for biochemical recurrence and distant metastasis in PCa patients. The enhanced and consistent prognostic predictive capability of ARS across various platforms was confirmed when compared with 44 previously published signatures. More importantly, PCa patients in the ARShigh group benefit more from PARP inhibitors and immunotherapy, while chemotherapy, radiotherapy, and AR-targeted therapy are more effective for ARSlow patients. The results of in silico screening suggest that AURKB could potentially serve as a promising therapeutic target for ARShigh patients. Conclusions Collectively, this prediction model based on AR regulatory genes holds great clinical translational potential to solve the dilemma of treatment choice and identify potential novel therapeutic targets in PCa.
Collapse
Affiliation(s)
- Qinyu Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Junjie Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Kai Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chengwei Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhiquan Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jia Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jun Xiao
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Peng Zhou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
111
|
Fernández Calvo O, Muñoz Iglesias J, Abou Jokh Casas E, Molina-Díaz A, Anido Herranz U, Casas Nebra J, García-Bernardo L, Martínez-Breijo S, Lázaro-Quintela M, Muñiz-García G, Vázquez-Estevez S. Recommendations from the Galician Oncological Society and the Galician Society of Nuclear Medicine for the use of 177Lu-PSMA-617 radioligand-therapy in prostate cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03662-7. [PMID: 39266875 DOI: 10.1007/s12094-024-03662-7] [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: 05/02/2024] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
Theragnostic is a type of precision medicine that uses molecules linked to radioactive isotopes for the diagnosis and treatment of diseases. In recent years, it has gained significant importance to treat neuroendocrine tumors and is currently being used in prostate cancer. Various radiopharmaceuticals have emerged for diagnosing and detecting lesions showing prostate-specific membrane antigen (PSMA) positivity on the Positron emission tomography/computed tomography scan, being the most widely used labeled with [68Ga] and [18F]. Its use as therapy in prostate cancer (PC) has been assessed in the VISION, TheraP, and PSMAfore clinical trials conducted with the radioligand [177Lu]Lu-PSMA-617, demonstrating significant antitumor activity. The aim of this article is to present practical recommendations, based on current available scientific evidence and on a multidisciplinary consensus, for the diagnosis and treatment with [177Lu]Lu-PSMA-617 in patients with PC.
Collapse
Affiliation(s)
- Ovidio Fernández Calvo
- Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain.
| | - José Muñoz Iglesias
- Department of Nuclear Medicine (SERGAS), University Hospital of Vigo, Meixoeiro Hospital, Vigo, Spain
| | | | - Aura Molina-Díaz
- Department of Medical Oncology, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Urbano Anido Herranz
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Javier Casas Nebra
- Uro-Oncology Unit, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Lucía García-Bernardo
- Department of Nuclear Medicine, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Sara Martínez-Breijo
- Department of Urology, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Martín Lázaro-Quintela
- Department of Medical Oncology, University Hospital of Vigo, Meixoeiro Hospital, Vigo, Spain
| | - Gloria Muñiz-García
- Department of Nuclear Medicine, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Sergio Vázquez-Estevez
- Department of Medical Oncology, Hospital Universitario Lucus Augusti de Lugo, Lugo, Spain
| |
Collapse
|
112
|
Chrenková E, Študentová H, Holá K, Kahounová Z, Hendrychová R, Souček K, Bouchal J. Castration-resistant prostate cancer monitoring by cell-free circulating biomarkers. Front Oncol 2024; 14:1394292. [PMID: 39319053 PMCID: PMC11420116 DOI: 10.3389/fonc.2024.1394292] [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: 03/01/2024] [Accepted: 08/23/2024] [Indexed: 09/26/2024] Open
Abstract
Background Prostate cancer is the second leading cause of male cancer-related deaths in Western countries, which is predominantly attributed to the metastatic castration-resistant stage of the disease (CRPC). There is an urgent need for better prognostic and predictive biomarkers, particularly for androgen receptor targeted agents and taxanes. Methods We have searched the PubMed database for original articles and meta-analyses providing information on blood-based markers for castration-resistant prostate cancer monitoring, risk group stratification and prediction of therapy response. Results The molecular markers are discussed along with the standard clinical parameters, such as prostate specific antigen, lactate dehydrogenase or C-reactive protein. Androgen receptor (AR) alterations are commonly associated with progression to CRPC. These include amplification of AR and its enhancer, point mutations and splice variants. Among DNA methylations, a novel 5-hydroxymethylcytosine activation marker of TOP2A and EZH2 has been identified for the aggressive disease. miR-375 is currently the most promising candidate among non-coding RNAs and sphingolipid analysis has recently emerged as a novel approach. Conclusions The promising biomarkers have the potential to improve the care of metastatic prostate cancer patients, however, they need further validation for routine implementation.
Collapse
Affiliation(s)
- Eva Chrenková
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Hana Študentová
- Department of Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Kateřina Holá
- Department of Oncology, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Romana Hendrychová
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, Olomouc, Czechia
| |
Collapse
|
113
|
Ruiz-Vico M, Wetterskog D, Orlando F, Thakali S, Wingate A, Jayaram A, Cremaschi P, Vainauskas O, Brighi N, Castellano-Gauna D, Åström L, Matveev VB, Bracarda S, Esen A, Feyerabend S, Senkus E, López-Brea Piqueras M, Gupta S, Wenstrup R, Boysen G, Martins K, Iwata K, Chowdhury S, Gourgioti G, Serikoff A, Gonzalez-Billalabeitia E, Merseburger AS, Demichelis F, Attard G. Liquid Biopsy in Progressing Prostate Cancer Patients Starting Docetaxel with or Without Enzalutamide: A Biomarker Study of the PRESIDE Phase 3b Trial. Eur Urol Oncol 2024:S2588-9311(24)00188-3. [PMID: 39261236 DOI: 10.1016/j.euo.2024.08.006] [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: 07/08/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND OBJECTIVE The PRESIDE (NCT02288247) randomized trial demonstrated prolonged progression-free survival (PFS) with continuing enzalutamide beyond progression in metastatic castration-resistant prostate cancer (mCRPC) patients starting docetaxel. This study aims to test the associations of PFS and circulating tumor DNA (ctDNA) prior to and after one cycle (cycle 2 day 1 [C2D1]) of docetaxel and with a liquid biopsy resistance biomarker (LBRB; plasma androgen receptor [AR] gain and/or circulating tumor cells [CTCs] expressing AR splice variant 7 [CTC-AR-V7]) prior to continuation of enzalutamide/placebo. METHODS Patients consenting to the biomarker substudy and donating blood before starting docetaxel with enzalutamide/placebo (N = 157) were included. Sequential plasma DNA samples were characterized with a prostate-cancer bespoke next-generation-sequencing capture panel (PCF_SELECT), and CTCs were assessed for AR-V7 (Epic Sciences, San Diego, CA, USA). Cox models, Kaplan-Meier, and restricted mean survival time (RMST) at 18 mo were calculated. KEY FINDINGS AND LIMITATIONS There was a significant association of worse PFS with pre-docetaxel ctDNA detection (N = 86 (55%), 8.1 vs 10.8 mo hazard ratio [HR] = 1.78, p = 0.004) or persistence/rise of ctDNA at C2D1 (N = 35/134, 5.5 vs 10.9 mo, HR = 1.95, 95% confidence interval [CI] = 1.15-3.30, p = 0.019). LBRB-positive patients (N = 62) had no benefit from continuing enzalutamide with docetaxel (HR = 0.78, 95% CI = 0.41-1.48, p = 0.44; RMST: 7.9 vs 7.1 mo, p = 0.50). Conversely, resistance biomarker-negative patients (N = 87) had significantly prolonged PFS (HR = 0.49, 95% CI = 0.29-0.82, p = 0.006; RMST: 11.5 vs 8.9 mo, p = 0.005). Eight patients were unevaluable. An exploratory analysis identified increased copy-number gains (CDK6/CDK4) at progression on docetaxel. Limitations included relatively low detection of CTC-AR-V7. Validation of impact on overall survival is required. CONCLUSIONS AND CLINICAL IMPLICATIONS Liquid biopsy gives an early indication of docetaxel futility, could guide patient selection for continuing enzalutamide, and identifies cell cycle gene alterations as a potential cause of docetaxel resistance in mCRPC. PATIENT SUMMARY In the PRESIDE biomarker study, we found that detecting circulating tumor DNA in plasma after starting treatment with docetaxel (chemotherapy) for metastatic prostate cancer resistant to androgen deprivation therapy can predict early how long patients will take to respond to treatment. Patients negative for a liquid biopsy resistance biomarker (based on the status of androgen receptor (AR) gene and AR splice variant 7 in circulating tumor cells) benefit from continuing enzalutamide in combination with docetaxel, while patients positive for the resistance biomarker did not. Additionally, we identified alterations in the cell cycle genes CDK6 and CDK4 as a potential genetic cause of resistance to docetaxel, which may support testing of specific drugs targeting these alterations.
Collapse
Affiliation(s)
- Maria Ruiz-Vico
- Oncology Department, University College London Cancer Institute, London, UK; PhD Program in Biomedicine Research, Universidad Complutense de Madrid, Madrid, Spain; Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Daniel Wetterskog
- Oncology Department, University College London Cancer Institute, London, UK
| | - Francesco Orlando
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Suparna Thakali
- Oncology Department, University College London Cancer Institute, London, UK
| | - Anna Wingate
- Oncology Department, University College London Cancer Institute, London, UK
| | - Anuradha Jayaram
- Oncology Department, University College London Cancer Institute, London, UK
| | - Paolo Cremaschi
- Oncology Department, University College London Cancer Institute, London, UK
| | | | - Nicole Brighi
- Oncology Department, University College London Cancer Institute, London, UK
| | | | - Lennart Åström
- Department of Immunology, Genetics and Pathology, University of Uppsala, Uppsala, Sweden
| | | | - Sergio Bracarda
- Medical Oncology, Azienda Ospedaliera Santa Maria, Terni, Italy
| | - Adil Esen
- Department of Urology, Dokuz Eylul University, Konak, Turkey
| | - Susan Feyerabend
- Studienpraxis Urologie, Medius Klinik Nürtingen, Nürtingen, Germany
| | - Elżbieta Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
| | | | - Santosh Gupta
- Translational Research, Epic Sciences Inc, San Diego, CA, USA
| | - Rick Wenstrup
- Translational Research, Epic Sciences Inc, San Diego, CA, USA
| | | | | | | | - Simon Chowdhury
- Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, King's College London, London, UK
| | | | | | - Enrique Gonzalez-Billalabeitia
- PhD Program in Biomedicine Research, Universidad Complutense de Madrid, Madrid, Spain; Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Axel S Merseburger
- Department of Urology, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Gerhardt Attard
- Oncology Department, University College London Cancer Institute, London, UK.
| |
Collapse
|
114
|
Markowski MC, Sternberg CN, Wang H, Wang T, Linville L, Marshall CH, Sullivan R, King S, Lotan TL, Antonarakis ES. TRIUMPH: phase II trial of rucaparib monotherapy in patients with metastatic hormone-sensitive prostate cancer harboring germline homologous recombination repair gene mutations. Oncologist 2024; 29:794-800. [PMID: 38885246 PMCID: PMC11379654 DOI: 10.1093/oncolo/oyae120] [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: 03/22/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The activity of PARP inhibitors (PARPi) in patients with homologous recombination repair (HRR) mutations and metastatic castration-resistant prostate cancer has been established. We hypothesized that the benefit of PARPi can be maintained in the absence of androgen deprivation therapy (ADT) in an HRR-mutated population. We report the results of a phase II clinical trial of rucaparib monotherapy in patients with metastatic hormone-sensitive prostate cancer (mHSPC). METHODS This was a multi-center, single-arm phase II trial (NCT03413995) for patients with asymptomatic, mHSPC. Patients were required to have a pathogenic germline mutation in an HRR gene for eligibility. All patients received rucaparib 600 mg by mouth twice daily, without androgen deprivation. The primary endpoint was a confirmed PSA50 response rate. RESULTS Twelve patients were enrolled, 7 with a BRCA1/2 mutation and 5 with a CHEK2 mutation. The confirmed PSA50 response rate to rucaparib was 41.7% (N = 5/12, 95% CI: 15.2-72.3%, one-sided P = .81 against the 50% null), which did not meet the pre-specified efficacy boundary to enroll additional patients. In patients with measurable disease, the objective response rate was 60% (N = 3/5), all with a BRCA2 mutation. The median radiographic progression-free survival on rucaparib was estimated at 12.0 months (95% CI: 8.0-NR months). The majority of adverse events were grade ≤2, and expected. CONCLUSION Rucaparib can induce clinical responses in a biomarker-selected metastatic prostate cancer population without concurrent ADT. However, the pre-specified efficacy threshold was not met, and enrolment was truncated. Although durable responses were observed in a subset of patients, further study of PARPi treatment without ADT in mHSPC is unlikely to change clinical practice.
Collapse
Affiliation(s)
- Mark C Markowski
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Cora N Sternberg
- Division of Hematology/Oncology, Englander Institute for Precision Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell, New York, NY, United States
| | - Hao Wang
- Division of Quantitative Sciences, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Tingchang Wang
- Division of Quantitative Sciences, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Laura Linville
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Catherine H Marshall
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Rana Sullivan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Serina King
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Emmanuel S Antonarakis
- Department of Medicine, Masonic Cancer Center, University of Minnesota Medical Center, Minneapolis, MN, United States
| |
Collapse
|
115
|
Wasserman JS, Fowle H, Hashmi R, Atar D, Patel KR, Yarmahmoodi A, Macfarlane AW, Tan Y, Cukierman E, Gligorijevic B, Karami A, Whelan KA, Campbell KS, Graña X. Derivation of human primary prostate epithelial cell lines by differentially targeting the CDKN2A locus along with expression of hTERT. Sci Rep 2024; 14:20409. [PMID: 39223207 PMCID: PMC11369182 DOI: 10.1038/s41598-024-71306-5] [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: 04/19/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Prostate cancer (PCa) is the most common cancer diagnosed in men worldwide and was the second leading cause of cancer-related deaths in US males in 2022. Prostate cancer also represents the second highest cancer mortality disparity between non-Hispanic blacks and whites. However, there is a relatively small number of prostate normal and cancer cell lines compared to other cancers. To identify the molecular basis of PCa progression, it is important to have prostate epithelial cell (PrEC) lines as karyotypically normal as possible. Our lab recently developed a novel methodology for the rapid and efficient immortalization of normal human PrEC that combines simultaneous CRISPR-directed inactivation of CDKN2A exon 2 (which directs expression of p16INK4A and p14ARF) and ectopic expression of an hTERT transgene. To optimize this methodology to generate immortalized lines with minimal genetic alterations, we sought to target exon 1α of the CDKN2A locus so that p16INK4A expression is ablated while the exons encoding p14ARF remains unaltered. Here we describe the establishment of two cell lines: one with the above-mentioned p16INK4A only loss, and a second line targeting both products in the CDKN2A locus. We characterize the potential lineage origin of these new cell lines along with our previously obtained clones, revealing distinct gene expression signatures. Based on the analyses of protein markers and RNA expression signatures, these cell lines are most closely related to a subpopulation of basal prostatic cells. Given the simplicity of this one-step methodology and the fact that it uses only the minimal genetic alterations necessary for immortalization, it should also be suitable for the establishment of cell lines from primary prostate tumor samples, an urgent need given the limited number of available prostate cancer cell lines.
Collapse
Affiliation(s)
- Jason S Wasserman
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Holly Fowle
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Rumesa Hashmi
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Diba Atar
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Kishan R Patel
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Amir Yarmahmoodi
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Alexander W Macfarlane
- Institute for Cancer Research, Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, USA
| | - Yinfei Tan
- Institute for Cancer Research, Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, USA
| | - Edna Cukierman
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
- Institute for Cancer Research, Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, USA
| | - Bojana Gligorijevic
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Institute for Cancer Research, Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, USA
- Bioengineering Department, Temple University, Philadelphia, PA, USA
| | - Adam Karami
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Kelly A Whelan
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
- Institute for Cancer Research, Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, USA
| | - Kerry S Campbell
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
- Institute for Cancer Research, Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, USA
| | - Xavier Graña
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, AHP Bldg., Room 308, 3307 North Broad St., Philadelphia, PA, 19140, USA.
- Institute for Cancer Research, Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, USA.
| |
Collapse
|
116
|
Takahashi S. Editorial for "Association of Pathological Features and Multiparametric MRI-Based Radiomics with TP53-Mutated Prostate Cancer". J Magn Reson Imaging 2024; 60:1146-1147. [PMID: 38190345 DOI: 10.1002/jmri.29224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/10/2024] Open
Affiliation(s)
- Satoru Takahashi
- Imaging Research Center, Takatsuki General Hospital, Takatsuki, Osaka, Japan
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| |
Collapse
|
117
|
Yuan H, Cai R, Chen B, Wang Q, Wang M, An J, An W, Tao Y, Yu J, Jiang B, Zhang Y, Xu M. Acetylated KHSRP impairs DNA-damage-response-related mRNA decay and facilitates prostate cancer tumorigenesis. Mol Oncol 2024; 18:2314-2330. [PMID: 38501452 PMCID: PMC11467790 DOI: 10.1002/1878-0261.13634] [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: 10/22/2023] [Revised: 01/28/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Androgen-regulated DNA damage response (DDR) is one of the essential mechanisms in prostate cancer (PCa), a hormone-sensitive disease. The heterogeneous nuclear ribonucleoprotein K (hnRNPK)-homology splicing regulatory protein known as far upstream element-binding protein 2 (KHSRP) is an RNA-binding protein that can attach to AU-rich elements in the 3' untranslated region (3'-UTR) of messenger RNAs (mRNAs) to mediate mRNA decay and emerges as a critical regulator in the DDR to preserve genome integrity. Nevertheless, how KHSRP responds to androgen-regulated DDR in PCa development remains unclear. This study found that androgen can significantly induce acetylation of KHSRP, which intrinsically drives tumor growth in xenografted mice. Moreover, enhanced KHSRP acetylation upon androgen stimuli impedes KHSRP-regulated DDR gene expression, as seen by analyzing RNA sequencing (RNA-seq) and Gene Set Enrichment Analysis (GSEA) datasets. Additionally, NAD-dependent protein deacetylase sirtuin-7 (SIRT7) is a promising deacetylase of KHSRP, and androgen stimuli impairs its interaction with KHSRP to sustain the increased KHSRP acetylation level in PCa. We first report the acetylation of KHSRP induced by androgen, which interrupts the KHSRP-regulated mRNA decay of the DDR-related genes to promote the tumorigenesis of PCa. This study provides insight into KHSRP biology and potential therapeutic strategies for PCa treatment, particularly that of castration-resistant PCa.
Collapse
Affiliation(s)
- Haihua Yuan
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Renjie Cai
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Biying Chen
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Qian Wang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Mengting Wang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Junyi An
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Weishu An
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Ye Tao
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Bin Jiang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Yanjie Zhang
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| | - Ming Xu
- Department of Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineChina
| |
Collapse
|
118
|
Triner D, Graf RP, Madison RW, Gjoerup O, Tukachinsky H, Ross JS, Quintanilha JCF, Li G, Cheng HH, Pritchard CC, Zurita AJ, Qin Q, Zhang T, Agarwal N, Reichert ZR, Mateo J, Cieslik M, Morgan TM. Durable benefit from poly(ADP-ribose) polymerase inhibitors in metastatic prostate cancer in routine practice: biomarker associations and implications for optimal clinical next-generation sequencing testing. ESMO Open 2024; 9:103684. [PMID: 39255537 PMCID: PMC11415711 DOI: 10.1016/j.esmoop.2024.103684] [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: 05/16/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Controlled trials have consistently demonstrated the efficacy of poly(ADP-ribose) polymerase inhibitors (PARPis) in patients with metastatic castration-resistant prostate cancer (mCRPC) and BRCA1 or BRCA2 alterations (BRCAalt). However, the reported efficacy of PARPi for alterations in other homologous recombination repair (HRR) genes is less consistent. We sought to evaluate the routine practice effectiveness of PARPi between and within these groups. DESIGN Patient-level data from a deidentified nationwide (USA-based) cancer clinico-genomic database between January 2011 and September 2023 were extracted. Patients with mCRPC and comprehensive genomic profiling by liquid biopsy [circulating tumor DNA (ctDNA)] or tissue (tumor) biopsy and who received single-agent PARPi were included and grouped by BRCAalt, ATMalt, other HRR, or no HRR. We further subcategorized BRCAalt into homozygous loss (BRCAloss) and all other deleterious BRCAalt (otherBRCAalt). RESULTS A total of 445 patients met inclusion criteria: 214 with tumor and 231 with ctDNA. BRCAalt had more favorable outcomes to PARPi compared with ATM, other HRR, and no HRR groups. Within the BRCAalt subgroup, compared with other BRCAalt, BRCAloss had a more favorable time to next treatment (median 9 versus 19.4 months, P = 0.005), time to treatment discontinuation (median 8 versus 14 months, P = 0.006), and routine practice overall survival (median 14.7 versus 19.4 months, P = 0.016). Tumor BRCAloss prevalence (3.1%) was similar to ctDNA prevalence in liquid biopsy specimens with high tumor fraction (>20%). BRCAloss was not detected in orthogonal germline testing. CONCLUSIONS PARPi routine practice effectiveness between groups mirrors prospective trials. Within the BRCAalt group, BRCAloss had the best outcomes. Unless the ctDNA tumor fraction is very high, somatic tissue testing (archival or metastatic) should be prioritized to identify patients who may benefit most from PARPi. When tissue testing is not clinically feasible, sufficient ctDNA tumor fraction levels for detection are enriched at clinical timepoints associated with tumor progression.
Collapse
Affiliation(s)
- D Triner
- Department of Urology, Michigan Medicine, Ann Arbor, USA
| | - R P Graf
- Foundation Medicine, Cambridge, USA
| | | | | | | | - J S Ross
- Foundation Medicine, Cambridge, USA; Department of Pathology, Upstate Medical University, Syracuse, USA; Department of Urology, Upstate Medical University, Syracuse, USA; Department of Medicine (Oncology), Upstate Medical University, Syracuse, USA
| | | | - G Li
- Foundation Medicine, Cambridge, USA
| | - H H Cheng
- University of Washington, Fred Hutchinson Cancer Center, Seattle, USA
| | - C C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - A J Zurita
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Q Qin
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA
| | - T Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA
| | - N Agarwal
- Department of Medical Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, USA
| | - Z R Reichert
- Department of Hematology/Oncology, University of Michigan, Ann Arbor, USA
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Barcelona Hospital Campus, Barcelona, Spain
| | - M Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - T M Morgan
- Department of Urology, Michigan Medicine, Ann Arbor, USA.
| |
Collapse
|
119
|
Narang A, Hage Chehade C, Ozay ZI, Nordblad B, Swami U, Agarwal N. Talazoparib for the treatment of prostate cancer. Expert Opin Pharmacother 2024; 25:1717-1727. [PMID: 39210559 DOI: 10.1080/14656566.2024.2397002] [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: 05/29/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Around 25% of patients with advanced prostate cancer harbor alterations in the homologous recombination/DNA damage repair (HRR) pathway. Inhibiting poly (ADP-ribose) polymerase (PARP) in these patients leads to synthetic lethality, making PARP inhibitors (PARPi), including talazoparib, a promising treatment for metastatic castration-resistant prostate cancer (mCRPC) and potentially for metastatic hormone-sensitive prostate cancer (mHSPC). AREAS COVERED This article examines the mechanism of action, chemical properties, pharmacokinetics, pharmacodynamics, and clinical safety and efficacy data of different PARPis, including talazoparib in prostate cancer. It reviews the TALAPRO-1 and TALAPRO-2 clinical trials and the ongoing TALAPRO-3 trial. EXPERT OPINION Despite recent therapeutic advancements, mCRPC remains a lethal disease. Androgen receptor pathway inhibitors (ARPIs) are approved for patients with mCRPC and mHSPC, yet most patients first receive these agents in the castration-resistant setting. Real-world data indicate that around half of patients with mCRPC do not receive subsequent lines of therapy, underscoring the efficacy of upfront combination therapies. The combinations of ARPI plus PARPi are indicated for patients with mCRPC harboring HRR mutations, though identifying these patients is challenging due to limited genomic testing. Further research and improved access to genomic testing are essential to optimize treatment strategies.
Collapse
Affiliation(s)
- Arshit Narang
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chadi Hage Chehade
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Zeynep Irem Ozay
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Blake Nordblad
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
120
|
Cai X, Yu X, Tang T, Xu Y, Wu T. JMJD2A promotes the development of castration-resistant prostate cancer by activating androgen receptor enhancer and inhibiting the cGAS-STING pathway. Mol Carcinog 2024; 63:1682-1696. [PMID: 38818897 DOI: 10.1002/mc.23753] [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: 02/28/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024]
Abstract
Exploring targets for inhibiting androgen receptor (AR) activity is an effective strategy for suppressing the development of castration-resistant prostate cancer (CRPC). Upregulation of histone demethylase JMJD2A activity is an important factor in increasing AR expression in CRPC. Based on our research, we found that the binding affinity between JMJD2A and AR increases in CRPC, while the level of AR histone methylation decreases and the H3K27ac level increases in the AR enhancer region. Further investigations revealed that overexpression of the histone demethylase JMJD2A increased the binding affinity between JMJD2A and AR, decreased AR histone methylation levels, upregulated H3K27ac in the AR enhancer region, and increased AR activity. Conversely, knocking down JMJD2A effectively reversed these effects. Additionally, in CRPC, JMJD2A expression was upregulated, the tumor-intrinsic immune cGAS-STING signaling pathway was suppressed, the tumor microenvironment was altered, and AR expression was upregulated. However, both knocking down JMJD2A and inhibiting the cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS-STING) signaling pathway reversed these effects. In summary, our study indicates that in CRPC, JMJD2A can directly bind to AR and activate residual AR enhancers through its demethylation activity, thereby promoting AR expression. Furthermore, upregulation of JMJD2A expression inhibits the innate immune cGAS-STING signaling pathway of the tumor, leading to a decrease in antitumor immune function, and further promoting AR expression.
Collapse
Affiliation(s)
- Xiang Cai
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaodong Yu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tielong Tang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yi Xu
- Department of Pharmacy, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Wu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
121
|
Leuva H, Zhou M, Jamaleddine N, Meseha M, Faiena I, Anna Park YH, McWilliams G, Luhrs C, Maxwell KN, Von Hoff D, Bates SE, Fojo T. Assessing olaparib efficacy in U.S. Veterans with metastatic prostate cancer utilizing a time-indifferent g-rate method ideal for real-world analyses. EBioMedicine 2024; 107:105288. [PMID: 39180789 PMCID: PMC11388178 DOI: 10.1016/j.ebiom.2024.105288] [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: 06/27/2023] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND We aimed to assess real-world efficacy of the PARP inhibitor, olaparib, in US Veterans with metastatic prostate cancer (mPC) by leveraging the national data repository and evaluate a novel approach to assess treatment efficacy in tumors considered rare or harboring rare mutations. METHODS Included Veterans had 1) mPC with somatic or germline alterations/mutations in genes involved in homologous recombination repair (HRR), 2) received olaparib monotherapy as well as a novel hormonal therapy/androgen receptor pathway inhibitors (NHT/ARPI), and/or chemotherapy, and 3) estimable rates of tumor growth (g-rate) using PSA values obtained while receiving treatment. Previous work has shown an excellent inverse correlation of g-rate with survival. Using g-rate, we determined tumor doubling time (DT) and DT ratios (DT on olaparib/DT on prior medication). We postulated that a DT ratio≥ 1 was associated with benefit. FINDINGS We identified 139 Veterans, including 42 Black males with tumors harboring mutations/alterations in HRR genes who received olaparib: BRCA2 (50), ATM (32), BRCA1 (10), other mutations (47). 62/139 (45%) of all and 21/42 (50%) of Black Veterans had DT ratios ≥1, including 31, 10, 2, and 19 with BRCA2, ATM, BRCA1, and other mutations, respectively (p = 0.006). Median survival with DT ratios ≥1 was superior, being 24.5 vs. 11.4 months for DT ratio <1 (p = 0.01, HR 0.50, 95% CI 0.29-0.85). Benefit from olaparib, defined as DT ratio ≥1, was not observed for germline status, starting PSA value, number of prior therapies, or immediate prior therapy. Compared to matched cohorts, tumors in the olaparib cohort had shorter DTs with enzalutamide in first line (367 vs. 884 days; p = 0.0043). INTERPRETATION Using equations indifferent to timing of assessments ideal for real-world efficacy analyses, we showed DT ratio ≥1 representing slower tumor growth on olaparib relative to the prior therapy correlates with improved survival. Olaparib efficacy in Veterans with mPC harboring mutations/alterations in HRR genes emulates clinical trial results. Black men had comparable results. Compared to matched cohorts, in first line, enzalutamide was less efficacious in tumors harboring mutations/alterations in HRR genes. FUNDING American Society of Clinical Oncology Conquer Cancer Foundation (ASCO CCF), the Blavatnik Family Foundation and the Prostate Cancer Foundation (PCF).
Collapse
Affiliation(s)
- Harshraj Leuva
- University of Nebraska Medical Center, Omaha, NE, USA; James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY, USA.
| | - Mengxi Zhou
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Nader Jamaleddine
- SUNY Downstate Health Sciences University, Brooklyn, NY, USA; Veterans Affairs New York Harbor Healthcare System - Brooklyn Campus, NY, USA
| | - Mina Meseha
- SUNY Downstate Health Sciences University, Brooklyn, NY, USA; Veterans Affairs New York Harbor Healthcare System - Brooklyn Campus, NY, USA
| | - Izak Faiena
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY, USA; James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY, USA
| | | | - Glen McWilliams
- James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY, USA
| | - Carol Luhrs
- SUNY Downstate Health Sciences University, Brooklyn, NY, USA; Veterans Affairs New York Harbor Healthcare System - Brooklyn Campus, NY, USA
| | - Kara N Maxwell
- Medicine and Genetics, Penn Medicine, and Corporal Michael Crescenz VAMC, USA
| | - Daniel Von Hoff
- Translational Genomics Research Institute, and HonorHealth Clinical Research Institute, Phoenix, AZ, USA
| | - Susan E Bates
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY, USA; James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY, USA
| | - Tito Fojo
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY, USA; James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY, USA.
| |
Collapse
|
122
|
Huber AK, Kaczorowski A, Schneider F, Böning S, Görtz M, Langhoff D, Schwab C, Stenzinger A, Hohenfellner M, Duensing A, Duensing S. Digital spatial profiling identifies the tumor center as a topological niche in prostate cancer characterized by an upregulation of BAD. Sci Rep 2024; 14:20281. [PMID: 39217197 PMCID: PMC11366015 DOI: 10.1038/s41598-024-71070-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Prostate cancer is characterized by a high degree of intratumoral heterogeneity. However, little is known about the spatial distribution of cancer cells with respect to specific functional characteristics and the formation of spatial niches. Here, we used digital spatial profiling (DSP) to investigate differences in protein expression in the tumor center versus the tumor periphery. Thirty-seven regions of interest were analyzed for the expression of 47 proteins, which included components of the PI3K-AKT, MAPK, and cell death signaling pathways as well as immune cell markers. A total of 1739 data points were collected from five patients. DSP identified the BCL-2 associated agonist of cell death (BAD) protein as the most significantly upregulated protein in the tumor center. BAD upregulation was confirmed by conventional immunohistochemistry, which furthermore showed a phosphorylation of BAD at serine 112 indicating its inactivation. Knockdown of BAD in prostate cancer cells in vitro led to decreased cell viability and colony growth. Clinically, high BAD expression was associated with a shorter time to biochemical recurrence in 158 mostly high-risk prostate cancer patients. Collectively, our results suggest that the tumor center is a topological niche with high BAD expression that may drive prostate cancer progression.
Collapse
Affiliation(s)
- Ann-Kathrin Huber
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Adam Kaczorowski
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Felix Schneider
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Sarah Böning
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Magdalena Görtz
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - David Langhoff
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Anette Duensing
- Precision Oncology of Urological Malignancies, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany.
| |
Collapse
|
123
|
Montoya-Novoa I, Gardeazábal-Torbado JL, Alegre-Martí A, Fuentes-Prior P, Estébanez-Perpiñá E. Androgen receptor post-translational modifications and their implications for pathology. Biochem Soc Trans 2024; 52:1673-1694. [PMID: 38958586 DOI: 10.1042/bst20231082] [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: 03/27/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
A major mechanism to modulate the biological activities of the androgen receptor (AR) involves a growing number of post-translational modifications (PTMs). In this review we summarise the current knowledge on the structural and functional impact of PTMs that affect this major transcription factor. Next, we discuss the cross-talk between these different PTMs and the presence of clusters of modified residues in the AR protein. Finally, we discuss the implications of these covalent modifications for the aetiology of diseases such as spinal and bulbar muscular atrophy (Kennedy's disease) and prostate cancer, and the perspectives for pharmacological intervention.
Collapse
Affiliation(s)
- Inés Montoya-Novoa
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - José Luis Gardeazábal-Torbado
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Andrea Alegre-Martí
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Pablo Fuentes-Prior
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Eva Estébanez-Perpiñá
- Structural Biology of Nuclear Receptors, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona (UB), 08028 Barcelona, Spain
| |
Collapse
|
124
|
Hommerding M, Hommerding O, Bernhardt M, Kreft T, Sanders C, Tischler V, Basitta P, Pelusi N, Wulf AL, Ohlmann CH, Ellinger J, Ritter M, Kristiansen G. Real-world data on the prevalence of BRCA1/2 and HRR gene mutations in patients with primary and metastatic castration resistant prostate cancer. World J Urol 2024; 42:491. [PMID: 39172235 PMCID: PMC11341621 DOI: 10.1007/s00345-024-05188-7] [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: 05/16/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
PURPOSE This study seeks to contribute real-world data on the prevalence of BRCA1/2 and HRR gene mutations in prostate cancer. METHODS We compiled sequencing data of 197 cases of primary and metastatic prostate cancer, in which HRR mutation analysis was performed upon clinical request within the last 5 years. All cases were analyzed using a targeted NGS BRCAness multigene panel, including 8 HRR genes (ATM, BRCA1, BRCA2, CDK12, CHEK2, FANCA, HDAC2, PALB2). RESULTS Our findings reveal a prevalence of potentially targetable mutations based on FDA criteria of 20.8%, which is comparable to the literature. However, the frequency of targetable BRCA2 mutations within our cohort was lower than reported for mCRPC and ATM and CHEK2 mutations were more prevalent instead. Thus, while 20.8% (n = 38) of the cases meet the criteria for olaparib treatment per FDA approval, only 4.9% (n = 9) align with the eligibility criteria according to the EMA approval. CONCLUSION This study offers valuable real-world insights into the landscape of BRCA1/2 and HRR gene mutations and the practical clinical management of HRR gene testing in prostate cancer, contributing to a better understanding of patient eligibility for PARPi treatment.
Collapse
Affiliation(s)
- Moritz Hommerding
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Oliver Hommerding
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Marit Bernhardt
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Tobias Kreft
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Christine Sanders
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Verena Tischler
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Patrick Basitta
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Natalie Pelusi
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | - Anna-Lena Wulf
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany
| | | | - Jörg Ellinger
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Manuel Ritter
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn (UKB), Venusberg-Campus 1, Bonn, 53127, Germany.
| |
Collapse
|
125
|
Li W, Guo F, Zeng R, Liang H, Wang Y, Xiong W, Wu H, Yang C, Jin X. CDK4/6 Alters TBK1 Phosphorylation to Inhibit the STING Signaling Pathway in Prostate Cancer. Cancer Res 2024; 84:2588-2606. [PMID: 38861362 DOI: 10.1158/0008-5472.can-23-3704] [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: 11/27/2023] [Revised: 04/02/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
The efficacy of immunotherapy in patients with prostate cancer is limited due to the "cold" tumor microenvironment and the paucity of neoantigens. The STING-TBK1-IRF3 signaling axis is involved in innate immunity and has been increasingly recognized as a candidate target for cancer immunotherapy. Here, we found that treatment with CDK4/6 inhibitors stimulates the STING pathway and enhances the antitumor effect of STING agonists in prostate cancer. Mechanistically, CDK4/6 phosphorylated TBK1 at S527 to inactivate the STING signaling pathway independent of RB1 in prostate cancer cells. CDK4/6-mediated phosphorylation of RB1 at S249/T252 also induced the interaction of RB1 with TBK1 to diminish the phosphorylation of TBK1 at S172, which suppressed STING pathway activation. Overall, this study showed that CDK4/6 suppresses the STING pathway through RB1-dependent and RB1-independent pathways, indicating that CDK4/6 inhibition could be a potential strategy to overcome immunosuppression in prostate cancer. Significance: Inhibiting CDK4/6 activates STING-TBK1-IRF3 signaling in prostate cancer by regulating TBK1 phosphorylation, suggesting that the combination of CDK4/6 inhibitors and STING agonists could be an effective approach to stimulate innate immunity.
Collapse
Affiliation(s)
- Wei Li
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Feng Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruijiang Zeng
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Huaiyuan Liang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Wei Xiong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunguang Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Jin
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
- Uro-Oncology Institute of Central South University, Changsha, China
| |
Collapse
|
126
|
Toscano-Guerra E, Maggio V, García J, Semidey ME, Celma A, Morote J, de Torres I, Giralt M, Ferrer-Costa R, Paciucci R. Association of the rs1042522 SNP with prostate cancer risk: a study of cancer tissues, primary tumor cultures, and serum samples from a Spanish Caucasian population. Front Oncol 2024; 14:1398411. [PMID: 39193388 PMCID: PMC11347290 DOI: 10.3389/fonc.2024.1398411] [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: 03/09/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Background Prostate cancer (PCa) is a leading cause of cancer-related deaths in European men, emphasizing the urgent need for effective risk assessment strategies. The TP53 gene, a tumor suppressor gene frequently mutated in cancer, commonly harbors the rs1042522 single nucleotide polymorphism (SNP), known as the P72R SNP, which may influence PCa susceptibility. This study investigated the prevalence of the P72R SNP in European Caucasian PCa samples and its association with PCa risk. Methods Genotyping was conducted on 12 hormone-naïve aggressive PCa cultures (hnPCs) from untreated patients (Gleason ≥8), 11 radical prostatectomies (RP), and 94 serum samples using DNA Sanger sequencing and melting curve analysis. Comparative analysis utilized data from the GnomAD database's European Caucasian non-cancer population. Results Our results demonstrate a significantly higher frequency of the P72R SNP in PCa samples and serums compared to the general European non-cancer population. A robust and statistically significant association (p < 0.0001) between the SNP and prostate cancer risk was identified, with an odds ratio of 7.937 (95% CI 5.37-11.00). Notably, the G allele (R72) showed a pronounced prevalence in high Gleason score (≥8) patients, although statistical significance was not reached. These results highlight a potential association with undifferentiated and malignant PCa lesions. Conclusion The compelling association between the P72R SNP and prostate cancer risk underscores the potential utility of this marker for the early identification of patients at risk of aggressive metastatic prostate cancer. This insight could empower further research to intervene at an early stage by offering enhanced opportunities for timely and targeted interventions.
Collapse
Affiliation(s)
- Emily Toscano-Guerra
- Cell Signaling and Cancer Progression Laboratory, Vall d’Hebron Institute of Research (VHIR), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Clinical Biochemistry Department, Biochemistry Service, Vall d’Hebron Hospital, Barcelona, Spain
- Facultad Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Valentina Maggio
- Cell Signaling and Cancer Progression Laboratory, Vall d’Hebron Institute of Research (VHIR), Barcelona, Spain
| | - Javier García
- Cell Signaling and Cancer Progression Laboratory, Vall d’Hebron Institute of Research (VHIR), Barcelona, Spain
| | - Maria Eugenia Semidey
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Department of Pathology, Vall d’Hebron Hospital, Barcelona, Spain
| | - Ana Celma
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Department of Urology, Vall d’Hebron Hospital, Barcelona, Spain
| | - Juan Morote
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Department of Pathology, Vall d’Hebron Hospital, Barcelona, Spain
| | - Inés de Torres
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Department of Pathology, Vall d’Hebron Hospital, Barcelona, Spain
| | - Marina Giralt
- Clinical Biochemistry Department, Biochemistry Service, Vall d’Hebron Hospital, Barcelona, Spain
| | - Roser Ferrer-Costa
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Clinical Biochemistry Department, Biochemistry Service, Vall d’Hebron Hospital, Barcelona, Spain
| | - Rosanna Paciucci
- Cell Signaling and Cancer Progression Laboratory, Vall d’Hebron Institute of Research (VHIR), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Spain
- Clinical Biochemistry Department, Biochemistry Service, Vall d’Hebron Hospital, Barcelona, Spain
| |
Collapse
|
127
|
Panebianco M, Cereda V, D’Andrea MR. Combination of the PARPi and ARSi in advanced castration resistant prostate cancer: a review of the recent phase III trials. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:997-1010. [PMID: 39351435 PMCID: PMC11438558 DOI: 10.37349/etat.2024.00260] [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: 04/09/2024] [Accepted: 05/31/2024] [Indexed: 10/04/2024] Open
Abstract
Tumors with an impaired ability to repair DNA double-strand breaks by homologous recombination, including those with alterations in breast cancer 1 and 2 (BRCA1 and BRCA2) genes, are very sensitive to blocking DNA single-strand repair by inhibition of the poly (ADP-ribose) polymerase (PARP) enzyme. This provides the basis for a synthetic deadly strategy in the treatment of different types of cancer, such as prostate cancer (PCa). The phase 3 PROfound study was the first to lead to olaparib approval in patients with metastatic castration resistant PCa (mCRPC) and BRCA genes mutations. In recent years, the benefit of combination therapy consisted of a PARP inhibitor (PARPi) plus an androgen receptor signalling inhibitor (ARSi), was evaluated as first-line treatment of mCRPC, regardless of the mutational state of genes, participating in the homologous recombination repair (HRR). This review explores the role of PARPi in PCa and analyses the data of latest clinical trials exploring the PARPi-ARSi combinations, and how these results could change our clinical practice.
Collapse
Affiliation(s)
| | - Vittore Cereda
- Medical Oncology of ASL Roma 4 Hospital, 00053 Civitavecchia, Italy
| | | |
Collapse
|
128
|
Cheng S, Li L, Yeh Y, Shi Y, Franco O, Corey E, Yu X. Unveiling novel double-negative prostate cancer subtypes through single-cell RNA sequencing analysis. NPJ Precis Oncol 2024; 8:171. [PMID: 39095562 PMCID: PMC11297170 DOI: 10.1038/s41698-024-00667-x] [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: 05/04/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
Recent advancements in single-cell RNA sequencing (scRNAseq) have facilitated the discovery of previously unrecognized subtypes within prostate cancer (PCa), offering new insights into cancer heterogeneity and progression. In this study, we integrated scRNAseq data from multiple studies, comprising publicly available cohorts and data generated by our research team, and established the Human Prostate Single cell Atlas (HuPSA) and Mouse Prostate Single cell Atlas (MoPSA) datasets. Through comprehensive analysis, we identified two novel double-negative PCa populations: KRT7 cells characterized by elevated KRT7 expression and progenitor-like cells marked by SOX2 and FOXA2 expression, distinct from NEPCa, and displaying stem/progenitor features. Furthermore, HuPSA-based deconvolution re-classified human PCa specimens, validating the presence of these novel subtypes. We then developed a user-friendly web application, "HuPSA-MoPSA" ( https://pcatools.shinyapps.io/HuPSA-MoPSA/ ), for visualizing gene expression across all newly established datasets. Our study provides comprehensive tools for PCa research and uncovers novel cancer subtypes that can inform clinical diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Siyuan Cheng
- Department of Biochemistry and Molecular Biology, LSU Health Shreveport, Shreveport, LA, USA.
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA.
| | - Lin Li
- Department of Biochemistry and Molecular Biology, LSU Health Shreveport, Shreveport, LA, USA
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA
| | - Yunshin Yeh
- Pathology & Laboratory Medicine Service, Overton Brooks VA Medical Center, Shreveport, LA, USA
| | - Yingli Shi
- Department of Biochemistry and Molecular Biology, LSU Health Shreveport, Shreveport, LA, USA
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA
| | - Omar Franco
- Department of Biochemistry and Molecular Biology, LSU Health Shreveport, Shreveport, LA, USA
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Xiuping Yu
- Department of Biochemistry and Molecular Biology, LSU Health Shreveport, Shreveport, LA, USA.
- Feist-Weiller Cancer Center, LSU Health Shreveport, Shreveport, LA, USA.
- Department of Urology, LSU Health Shreveport, Shreveport, LA, USA.
| |
Collapse
|
129
|
Qureshi Z, Altaf F, Khanzada M, Zaheer Z, Fatima E, Bakhtiar M. Capivasertib in Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative advanced breast cancer. Curr Probl Cancer 2024; 51:101114. [PMID: 38959565 DOI: 10.1016/j.currproblcancer.2024.101114] [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: 03/26/2024] [Revised: 05/24/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024]
Abstract
PURPOSE This review discusses the role and efficacy of Capivasertib in managing Hormone Receptor-Positive (HR+) breast cancer. SUMMARY Breast cancer is the most prevalent type of cancer among women worldwide. This article is an in-depth analysis of advanced therapeutic options involving Capivasertib in treating HR+ Breast Cancer. It focuses on the mode of action, efficacy, clinical trials, and comparison with fulvestrant alone. This review also highlights the therapy's precision in targeting specific cancer cells. Its mechanism of action involves preventing cancer cells from growing and having a cytotoxic effect on them. It improves progression-free survival while maintaining the quality of life. The side effects can be easily managed by dose reduction or discontinuation of the drug. This article sheds light on the ongoing trials and FDA recognition. CONCLUSION In conclusion, Capivasertib-fulvestrant therapy shows potential as an innovative therapeutic option for HR+ breast cancer but warrants additional research, especially in randomized control trials (RCT). It resulted in longer progression-free survival compared to fulvestrant alone. Its side effect profile is minimal.
Collapse
Affiliation(s)
- Zaheer Qureshi
- Assistant Professor of Medicine, The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, Connecticut, USA
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, USA
| | - Mikail Khanzada
- Department of Medicine, Lahore Medical and Dental College, Lahore, Pakistan
| | - Zaofashan Zaheer
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan.
| | - Muhammad Bakhtiar
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| |
Collapse
|
130
|
Slootbeek PH, Luna-Velez MV, Privé BM, van der Doelen MJ, Kloots IS, Pamidimarri Naga S, Onstenk HE, Nagarajah J, Westdorp H, van Oort IM, Kroeze LI, Schalken JA, Bloemendal HJ, Mehra N. Impact of TP53 loss-of-function alterations on the response to PSMA radioligand therapy in metastatic castration-resistant prostate cancer patients. Theranostics 2024; 14:4555-4569. [PMID: 39239510 PMCID: PMC11373632 DOI: 10.7150/thno.96322] [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: 02/01/2024] [Accepted: 07/11/2024] [Indexed: 09/07/2024] Open
Abstract
Rationale: PSMA-targeting radioligand therapy (PSMA-RLT) has shown promise in metastatic castration-resistant prostate cancer (mCRPC), particularly in PSMA-avid tumours. However, predicting response remains challenging. Preclinical data suggests aberrant p53-signalling as a predictor of poor response. Methods: The patient population of this pre-planned retrospective cohort study consists of 96 patients with mCRPC who underwent treatment with PSMA-RLT and were molecularly profiled by whole-genome sequencing and or targeted next-generation sequencing. Response to PSMA-RLT was assessed per molecular subtype, including TP53-mutational status. Results: Patients with TP53 loss-of-function alterations had a shorter median progression-free survival (3.7 versus 6.2 months, P<0.001), a lower median PSA change (-55% vs. -75%, P=0.012) and shorter overall survival from initiation of PMSA-RLT (7.6 vs. 13.9 months, P=0.003) compared to TP53-wildtype patients. Pathogenic alterations in AR, MYC, BRCA1, or BRCA2 as well as in genes linked to the PI3K or MAPK pathways or genes involved in homologous recombination repair, were not associated with response. Only lactate dehydrogenase was, alongside TP53-status, significantly associated with response. Transcriptome analysis of 21 patients, identified six p53 signalling genes whose low expression was associated to a shorter progression-free survival (P<0.05). Conclusion: TP53 loss-of-function may serve as a prognostic factor for PSMA-RLT outcomes in patients with mCRPC.
Collapse
Affiliation(s)
- Peter H.J. Slootbeek
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Bastiaan M. Privé
- Department of Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Iris S.H. Kloots
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Hilde E. Onstenk
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - James Nagarajah
- Department of Nuclear Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Roentgeninstitut Duesseldorf, Duesseldorf, Germany
| | - Harm Westdorp
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Inge M. van Oort
- Department of Urology, Radboud university medical center, Nijmegen, The Netherlands
| | - Leonie I. Kroeze
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - Jack. A. Schalken
- Department of Urology, Radboud university medical center, Nijmegen, The Netherlands
| | - Haiko J. Bloemendal
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
131
|
Zhao SG, Bootsma M, Zhou S, Shrestha R, Moreno-Rodriguez T, Lundberg A, Pan C, Arlidge C, Hawley JR, Foye A, Weinstein AS, Sjöström M, Zhang M, Li H, Chesner LN, Rydzewski NR, Helzer KT, Shi Y, Lynch M, Dehm SM, Lang JM, Alumkal JJ, He HH, Wyatt AW, Aggarwal R, Zwart W, Small EJ, Quigley DA, Lupien M, Feng FY. Integrated analyses highlight interactions between the three-dimensional genome and DNA, RNA and epigenomic alterations in metastatic prostate cancer. Nat Genet 2024; 56:1689-1700. [PMID: 39020220 PMCID: PMC11319208 DOI: 10.1038/s41588-024-01826-3] [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: 05/16/2023] [Accepted: 06/10/2024] [Indexed: 07/19/2024]
Abstract
The impact of variations in the three-dimensional structure of the genome has been recognized, but solid cancer tissue studies are limited. Here, we performed integrated deep Hi-C sequencing with matched whole-genome sequencing, whole-genome bisulfite sequencing, 5-hydroxymethylcytosine (5hmC) sequencing and RNA sequencing across a cohort of 80 biopsy samples from patients with metastatic castration-resistant prostate cancer. Dramatic differences were present in gene expression, 5-methylcytosine/5hmC methylation and in structural variation versus mutation rate between A and B (open and closed) chromatin compartments. A subset of tumors exhibited depleted regional chromatin contacts at the AR locus, linked to extrachromosomal circular DNA (ecDNA) and worse response to AR signaling inhibitors. We also identified topological subtypes associated with stark differences in methylation structure, gene expression and prognosis. Our data suggested that DNA interactions may predispose to structural variant formation, exemplified by the recurrent TMPRSS2-ERG fusion. This comprehensive integrated sequencing effort represents a unique clinical tumor resource.
Collapse
Grants
- R01 CA270539 NCI NIH HHS
- R01 CA276269 NCI NIH HHS
- R01 CA174777 NCI NIH HHS
- P50 CA097186 NCI NIH HHS
- 1DP2CA271832-01, P30 CA014520 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- DP2 CA271832 NCI NIH HHS
- P50 CA186786 NCI NIH HHS
- R01 CA282005 NCI NIH HHS
- R01 CA251245, P50 CA097186, P50 CA186786, P50 CA186786-07S1, P30 CA046592, and W81XWH-20-1-0405 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- P30 CA046592 NCI NIH HHS
- R01 CA251245 NCI NIH HHS
- P30 CA014520 NCI NIH HHS
- W81XWH2010799 U.S. Department of Defense (United States Department of Defense)
- W81XWH-21-1-0046 U.S. Department of Defense (United States Department of Defense)
- SU2C-AACR-DT0812 EIF | Stand Up To Cancer (SU2C)
- Prostate Cancer Foundation (PCF)
- UCSF Benioff Initiative for Prostate Cancer Research
- U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- Canadian Institute of Health Research (CIHR) (FRN-153234 & 168933), the Canadian Epigenetics, Environment, and Health Research Consortium (CEEHRC) (FRN-158225), the Ontario Institute for Cancer Research (OICR) through funding provided by the Government of Ontario (IA 031), and the Princess Margaret Cancer Foundation.
Collapse
Affiliation(s)
- Shuang G Zhao
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Madison, WI, USA
| | - Matthew Bootsma
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Stanley Zhou
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Raunak Shrestha
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Thaidy Moreno-Rodriguez
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Arian Lundberg
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Chu Pan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Arlidge
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - James R Hawley
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Adam Foye
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Alana S Weinstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Martin Sjöström
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Meng Zhang
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Haolong Li
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Lisa N Chesner
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Nicholas R Rydzewski
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kyle T Helzer
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Yue Shi
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Molly Lynch
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Scott M Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshi J Alumkal
- Department of Internal Medicine, Division of Hematology-Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Hansen H He
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Alexander W Wyatt
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Wilbert Zwart
- Netherlands Cancer Institute, Oncode Institute, Amsterdam, the Netherlands
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Department of Urology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
132
|
Paller CJ, Zahurak ML, Mandl A, Metri NA, Lalji A, Heath E, Kelly WK, Hoimes C, Barata P, Taksey J, Garrison DA, Patra K, Milne GL, Anders NM, Nauroth JM, Durham JN, Marshall CH, Markowski MC, Eisenberger MA, Antonarakis ES, Carducci MA, Denmeade SR, Levine M. High-Dose Intravenous Vitamin C Combined with Docetaxel in Men with Metastatic Castration-Resistant Prostate Cancer: A Randomized Placebo-Controlled Phase II Trial. CANCER RESEARCH COMMUNICATIONS 2024; 4:2174-2182. [PMID: 39076107 PMCID: PMC11333993 DOI: 10.1158/2767-9764.crc-24-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/21/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
High-dose intravenous vitamin C (HDIVC) administered to produce pharmacologic concentrations shows promise in preclinical models and small clinical trials, but larger prospective randomized trials are lacking. We evaluated the clinical benefit of combining HDIVC with docetaxel in patients with progressive metastatic castration-resistant prostate cancer (mCRPC). In this double-blind, placebo-controlled phase II trial, 47 patients were randomized 2:1 to receive docetaxel (75 mg/m2 i.v.) with either HDIVC (1 g/kg) or placebo. Coprimary endpoints were PSA50 response and adverse event rates. Secondary endpoints included overall survival, radiographic progression-free survival, and quality of life measured using the Functional Assessment of Cancer Therapy-Prostate instrument. Correlative analyses included pharmacokinetics and oxidative stress markers. Eighty-nine percent of patients previously had three or more lines of therapy. The PSA50 response rate was 41% in the HDIVC group and 33% in the placebo group (P = 0.44), with comparable adverse event rates in both groups. There were no significant differences in Functional Assessment of Cancer Therapy-Prostate scores. The median radiographic progression-free survival was not significantly different between the HDIVC and placebo groups, with durations of 10.1 and 10.0 months (HR, 1.35; 95% confidence interval, 0.66-2.75; P = 0.40), respectively. The median overall survival was 15.2 months in the HDIVC group and 29.5 months in the placebo group (HR, 1.98; 95% confidence interval, 0.85-4.58; P = 0.11). HDIVC did not decrease F2-isoprostanes, indicators of oxidative stress. The study was suspended after prespecified interim analysis indicated futility in achieving primary endpoints. In this patient population, combining HDIVC with docetaxel did not improve PSA response, toxicity, or other clinical outcomes compared with docetaxel alone. Findings do not support the routine use of HDIVC in mCRPC treatment outside of clinical trials. SIGNIFICANCE This is the first randomized, placebo-controlled, double-blind trial to evaluate HDIVC in cancer treatment. The addition of HDIVC to docetaxel in patients with mCRPC does not improve PSA response, toxicity, or other clinical outcomes compared with docetaxel alone. The routine use of HDIVC in mCRPC treatment is not supported outside of clinical trials.
Collapse
Affiliation(s)
- Channing J. Paller
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Marianna L. Zahurak
- Division of Biostatistics and Bioinformatics, Johns Hopkins University, Baltimore, Maryland.
| | - Adel Mandl
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Nicole A. Metri
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Aliya Lalji
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | | | | | | | - Pedro Barata
- Case Western Reserve University/University Hospitals, Cleveland, Ohio.
| | - Jason Taksey
- Maryland Oncology Hematology, US Oncology, Annapolis, Maryland.
| | - Dominique A. Garrison
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Kartick Patra
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland.
| | - Ginger L. Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Nicole M. Anders
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Julie M. Nauroth
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Jennifer N. Durham
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Catherine H. Marshall
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Mark C. Markowski
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Mario A. Eisenberger
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | | | - Michael A. Carducci
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Samuel R. Denmeade
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
133
|
Orme JJ, Taza F, De Sarkar N, Tewari AK, Arsalan Naqvi S, Riaz IB, Childs DS, Omar N, Adra N, Ashkar R, Cheng HH, Schweizer MT, Sokolova AO, Agarwal N, Barata P, Sartor O, Bastos D, Smaletz O, Berchuck JE, McClure H, Taplin ME, Aggarwal R, Sternberg CN, Vlachostergios PJ, Alva AS, Mehra N, Nelson PS, Hwang J, Dehm SM, Shi Q, Fleischmann Z, Sokol ES, Elliott A, Huang H, Bryce A, Marshall CH, Antonarakis ES. Co-occurring BRCA2/SPOP Mutations Predict Exceptional Poly (ADP-ribose) Polymerase Inhibitor Sensitivity in Metastatic Castration-Resistant Prostate Cancer. Eur Urol Oncol 2024; 7:877-887. [PMID: 38072760 PMCID: PMC11162506 DOI: 10.1016/j.euo.2023.11.014] [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: 08/31/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 06/10/2024]
Abstract
BACKGROUND AND OBJECTIVE BRCA2 mutations in metastatic castration-resistant prostate cancer (mCRPC) confer sensitivity to poly (ADP-ribose) polymerase (PARP) inhibitors. However, additional factors predicting PARP inhibitor efficacy in mCRPC are needed. Preclinical studies support a relationship between speckle-type POZ protein (SPOP) inactivation and PARP inhibitor sensitivity. We hypothesized that SPOP mutations may predict enhanced PARP inhibitor response in BRCA2-altered mCRPC. METHODS We conducted a multicenter retrospective study involving 13 sites. We identified 131 patients with BRCA2-altered mCRPC treated with PARP inhibitors, 14 of which also carried concurrent SPOP mutations. The primary efficacy endpoint was prostate-specific antigen (PSA) response rate (≥50% PSA decline). The secondary endpoints were biochemical progression-free survival (PSA-PFS), clinical/radiographic progression-free survival (PFS), and overall survival (OS). These were compared by multivariable Cox proportional hazard models adjusting for age, tumor stage, baseline PSA level, Gleason sum, prior therapies, BRCA2 alteration types, and co-occurring mutations. KEY FINDINGS AND LIMITATIONS Baseline characteristics were similar between groups. PSA responses were observed in 60% (70/117) of patients with BRCA2mut/SPOPwt disease and in 86% (12/14) of patients with BRCA2mut/SPOPmut disease (p = 0.06). The median time on PARP inhibitor treatment was 24.0 mo (95% confidence interval [CI] 19.2 mo to not reached) in this group versus 8.0 mo (95% CI 6.1-10.9 mo) in patients with BRCA2 mutation alone (p = 0.05). In an unadjusted analysis, patients with BRCA2mut/SPOPmut disease experienced longer PSA-PFS (hazard ratio [HR] 0.33 [95% CI 0.15-0.72], p = 0.005) and clinical/radiographic PFS (HR 0.4 [95% CI 0.18-0.86], p = 0.02), and numerically longer OS (HR 0.4 [95% CI 0.15-1.12], p = 0.08). In a multivariable analysis including histology, Gleason sum, prior taxane, prior androgen receptor pathway inhibitor, stage, PSA, BRCA2 alteration characteristics, and other co-mutations, patients with BRCA2mut/SPOPmut disease experienced longer PSA-PFS (HR 0.16 [95% CI 0.05-0.47], adjusted p = 0.001), clinical/radiographic PFS (HR 0.28 [95% CI 0.1-0.81], adjusted p = 0.019), and OS (HR 0.19 [95% CI 0.05-0.69], adjusted p = 0.012). In a separate cohort of patients not treated with a PARP inhibitor, there was no difference in OS between patients with BRCA2mut/SPOPmut versus BRCA2mut/SPOPwt disease (HR 0.97 [95% CI 0.40-2.4], p = 0.94). In a genomic signature analysis, Catalog of Somatic Mutations in Cancer (COSMIC) SBS3 scores predictive of homologous recombination repair (HRR) defects were higher for BRCA2mut/SPOPmut than for BRCA2mut/SPOPwt disease (p = 0.04). This was a retrospective study, and additional prospective validation cohorts are needed. CONCLUSIONS AND CLINICAL IMPLICATIONS In this retrospective analysis, PARP inhibitors appeared more effective in patients with BRCA2mut/SPOPmut than in patients with BRCA2mut/SPOPwt mCRPC. This may be related to an increase in HRR defects in coaltered disease. PATIENT SUMMARY In this study, we demonstrate that co-alteration of both BRCA2 and SPOP predicts superior clinical outcomes to treatment with poly (ADP-ribose) polymerase (PARP) inhibitors than BRCA2 alteration without SPOP mutation.
Collapse
Affiliation(s)
- Jacob J Orme
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Fadi Taza
- Division of Hematology & Medical Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Navonil De Sarkar
- Department of Pathology and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alok K Tewari
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Irbaz B Riaz
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Daniel S Childs
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Noha Omar
- Ascension St Agnes Hospital, Baltimore, MD, USA
| | - Nabil Adra
- Division of Hematology & Medical Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ryan Ashkar
- Division of Hematology & Medical Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heather H Cheng
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael T Schweizer
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Oliver Sartor
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Diogo Bastos
- Oncology Center, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Oren Smaletz
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jacob E Berchuck
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Heather McClure
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rahul Aggarwal
- University of California San Francisco, San Francisco, CA, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Niven Mehra
- Radboud University, Nijmegen, The Netherlands
| | - Peter S Nelson
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Justin Hwang
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Scott M Dehm
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA; Masonic Cancer Center, Minneapolis, MN, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA; Department of Urology, University of Minnesota, Minneapolis, MN, USA
| | - Qian Shi
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Alan Bryce
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Emmanuel S Antonarakis
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA; Masonic Cancer Center, Minneapolis, MN, USA.
| |
Collapse
|
134
|
Ridouani F, Alberto Vargas H, Holzwanger DJ, Schöder H, Waters E, Petre EN, Martin A, Satagopan J, Gonen M, Autio KA, Chen Y, Slovin SF, Danila DC, Morris MJ, Scher HI, Arcila ME, Solomon SB, Durack JC. Clinical, Imaging, and Technical Factors Associated with Successful Genomic Profiling of Bone Biopsy Tissue in Prostate Cancer. Eur Urol Oncol 2024:S2588-9311(24)00180-9. [PMID: 39095299 DOI: 10.1016/j.euo.2024.07.007] [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: 04/17/2024] [Revised: 06/14/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND AND OBJECTIVE The source of tissue for genomic profiling of metastatic castration-resistant prostate cancer (mCRPC) is often limited to osseous metastases. To guide patient management, metastatic site selection and the technique for targeted bone biopsies are critical for identifying deleterious gene mutations. Our objective was to identify key parameters associated with successful large-panel DNA sequencing. METHODS We analyzed parameters for 243 men with progressing mCRPC who underwent 269 bone biopsies for genomic profiling between 2014 and 2018. Univariate and multivariate analyses were performed for clinical, imaging (bone scan; fluorodeoxyglucose [FDG] positron emission tomography [PET]; computed tomography [CT]; magnetic resonance imaging), and technical (biopsy site, number of samples, needle gauge) features associated with successful genomic profiling. KEY FINDINGS AND LIMITATIONS Overall, 159 of 269 biopsies (59%) generated sufficient tumor material for a genomic profile. Seventy (26%) of the failures were histopathologically negative for mCRPC and 40 (15%) had insufficient tumor for genomic profiling. Of 199 mCRPC samples submitted for molecular testing, 159 (80%) yielded a genomic profile. On univariate analysis, PSA, serum acid phosphatase, number of biopsy samples, FDG PET positivity, CT attenuation, and CT morphology were significantly associated with genomic profiling success. On multivariate analysis, higher FDG maximum standardized uptake value (odds ratio [OR] 7.51, 95% confidence interval [CI] 3.01-18.78; p < 0.001), higher number of biopsy samples (OR 4.73, 95% CI 1.49-15.02; p = 0.008), and lower mean CT attenuation (OR 0.4, 95% CI 0.18-0.89; p = 0.025) were significantly associated with sequencing success. CONCLUSIONS AND CLINICAL IMPLICATIONS In patients with mCRPC, bone biopsies from sites with metabolic activity and lower CT attenuation are associated with higher success rates for genomic profiling via a large-panel DNA sequencing platform. PATIENT SUMMARY We identified factors associated with successful genetic testing of bone tissue for patients with metastatic prostate cancer. Our findings may help in guiding the right scan technique and biopsy site for personalized treatment planning.
Collapse
Affiliation(s)
- Fourat Ridouani
- Department of Radiology, Interventional Radiology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - H Alberto Vargas
- Department of Radiology, Oncologic Imaging Division, NYU Langone, New York, NY, USA
| | - Daniel J Holzwanger
- Department of Radiology, Interventional Radiology Division, Weill Cornell Medicine, New York, NY, USA
| | - Heiko Schöder
- Department of Radiology, Molecular Imaging and Therapy Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Emily Waters
- Department of Medicine, Genitourinary Oncology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Elena N Petre
- Department of Radiology, Interventional Radiology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Axel Martin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Jaya Satagopan
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, New Brunswick, NJ, USA
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Karen A Autio
- Department of Medicine, Genitourinary Oncology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Yu Chen
- Department of Medicine, Genitourinary Oncology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Susan F Slovin
- Department of Medicine, Genitourinary Oncology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Daniel C Danila
- Department of Medicine, Genitourinary Oncology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Michael J Morris
- Department of Medicine, Genitourinary Oncology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Howard I Scher
- Department of Medicine, Genitourinary Oncology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Stephen B Solomon
- Department of Radiology, Interventional Radiology Division, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Jeremy C Durack
- Department of Radiology, Interventional Radiology Division, Memorial Sloan Kettering Cancer, New York, NY, USA; Department of Radiology, Interventional Radiology Division, Veterans Administration Hospital, Palo Alto, CA, USA.
| |
Collapse
|
135
|
Yin J, Daryanani A, Lu F, Ku AT, Bright JR, Alilin ANS, Bowman J, Lake R, Li C, Truong TM, Twohig JD, Mostaghel EA, Ishikawa M, Simpson M, Trostel SY, Corey E, Sowalsky AG, Kelly K. Reproducible preclinical models of androgen receptor driven human prostate cancer bone metastasis. Prostate 2024; 84:1033-1046. [PMID: 38708958 PMCID: PMC11216894 DOI: 10.1002/pros.24718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Preclinical models recapitulating the metastatic phenotypes are essential for developing the next-generation therapies for metastatic prostate cancer (mPC). We aimed to establish a cohort of clinically relevant mPC models, particularly androgen receptor positive (AR+) bone metastasis models, from LuCaP patient-derived xenografts (PDX) that reflect the heterogeneity and complexity of mPC. METHODS PDX tumors were dissociated into single cells, modified to express luciferase, and were inoculated into NSG mice via intracardiac injection. The progression of metastases was monitored by bioluminescent imaging. Histological phenotypes of metastases were characterized by immunohistochemistry and immunofluorescence staining. Castration responses were further investigated in two AR-positive models. RESULTS Our PDX-derived metastasis (PDM) model collection comprises three AR+ adenocarcinomas (ARPC) and one AR- neuroendocrine carcinoma (NEPC). All ARPC models developed bone metastases with either an osteoblastic, osteolytic, or mixed phenotype, while the NEPC model mainly developed brain metastasis. Different mechanisms of castration resistance were observed in two AR+ PDM models with distinct genotypes, such as combined loss of TP53 and RB1 in one model and expression of AR splice variant 7 (AR-V7) expression in another model. Intriguingly, the castration-resistant tumors displayed inter- and intra-tumor as well as organ-specific heterogeneity in lineage specification. CONCLUSION Genetically diverse PDM models provide a clinically relevant system for biomarker identification and personalized medicine in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- JuanJuan Yin
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Asha Daryanani
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Fan Lu
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Anson T. Ku
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - John R. Bright
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Aian Neil S. Alilin
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Joel Bowman
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Ross Lake
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Chennan Li
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Tri M. Truong
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Joseph D. Twohig
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Elahe A. Mostaghel
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Masaki Ishikawa
- Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mark Simpson
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Shana Y. Trostel
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Adam G. Sowalsky
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
136
|
De Santis M, Breijo SM, Robinson P, Capone C, Pascoe K, Van Sanden S, Hashim M, Trevisan M, Daly C, Reitsma F, van Beekhuizen S, Ruan H, Heeg B, Verzoni E. Feasibility of Indirect Treatment Comparisons Between Niraparib Plus Abiraterone Acetate and Other First-Line Poly ADP-Ribose Polymerase Inhibitor Treatment Regimens for Patients with BRCA1/2 Mutation-Positive Metastatic Castration-Resistant Prostate Cancer. Adv Ther 2024; 41:3039-3058. [PMID: 38958846 PMCID: PMC11263413 DOI: 10.1007/s12325-024-02918-6] [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: 04/11/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Poly(ADP-ribose) polymerase inhibitors (PARPi) are a novel option to treat patients with metastatic castration-resistant prostate cancer (mCRPC). Niraparib plus abiraterone acetate and prednisone (AAP) is indicated for BRCA1/2 mutation-positive mCRPC. Niraparib plus AAP demonstrated safety and efficacy in the phase 3 MAGNITUDE trial (NCT03748641). In the absence of head-to-head studies comparing PARPi regimens, the feasibility of conducting indirect treatment comparisons (ITC) to inform decisions for patients with first-line BRCA1/2 mutation-positive mCRPC has been explored. METHODS A systematic literature review was conducted to identify evidence from randomized controlled trials on relevant comparators to inform the feasibility of conducting ITCs via network meta-analysis (NMA) or population-adjusted indirect comparisons (PAIC). Feasibility was assessed based on network connectivity, data availability in the BRCA1/2 mutation-positive population, and degree of within- and between-study heterogeneity or bias. RESULTS NMAs between niraparib plus AAP and other PARPi regimens (olaparib monotherapy, olaparib plus AAP, and talazoparib plus enzalutamide) were inappropriate due to the disconnected network, differences in trial populations related to effect modifiers, or imbalances within BRCA1/2 mutation-positive subgroups. The latter issue, coupled with the lack of a common comparator (except for olaparib plus AAP), also rendered anchored PAICs infeasible. Unanchored PAICs were either inappropriate due to lack of population overlap (vs. olaparib monotherapy) or were restricted by unmeasured confounders and small sample size (vs. olaparib plus AAP). PAIC versus talazoparib plus enzalutamide was not possible due to lack of published arm-level baseline characteristics and sufficient efficacy outcome data in the relevant population. CONCLUSION The current randomized controlled trial evidence network does not permit robust comparisons between niraparib plus AAP and other PARPi regimens for patients with 1L BRCA-positive mCRPC. Decision-makers should scrutinize any ITC results in light of their limitations. Real-world evidence combined with clinical experience should inform treatment recommendations in this indication.
Collapse
Affiliation(s)
- Maria De Santis
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Urology, Medical University Vienna, Vienna, Austria
| | - Sara Martínez Breijo
- A Coruña University Hospital, C/As Xubias de Arriba nº 86, 15006, A Coruña, Spain
- Department of Urology, Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidad de A Coruña (UDC), 15006, A Coruña, Spain
| | - Paul Robinson
- Janssen-Cilag Limited, 50-100 Holmers Farm Way, High Wycombe, HP12 4EG, UK
| | - Camille Capone
- Janssen-Cilag, 1 Rue Camille Desmoulins, 92130, Issy Les Moulineaux, France.
| | - Katie Pascoe
- Janssen-Cilag Limited, 50-100 Holmers Farm Way, High Wycombe, HP12 4EG, UK
| | | | - Mahmoud Hashim
- Janssen Vaccines & Prevention BV, Archimedesweg 4-6, 2333 CN, Leiden, The Netherlands
| | - Marco Trevisan
- Janssen-Cilag AG, Gubelstrasse 34, 6300, Zug, Switzerland
| | - Caitlin Daly
- Cytel Inc., 1 University Avenue, 3rd Floor, Toronto, ON, M5J 2P1, Canada
| | - Friso Reitsma
- Cytel Inc., Weena 316-318, 3012 NJ, Rotterdam, The Netherlands
| | | | - Haoyao Ruan
- Cytel Inc., 1 University Avenue, 3rd Floor, Toronto, ON, M5J 2P1, Canada
| | - Bart Heeg
- Cytel Inc., Weena 316-318, 3012 NJ, Rotterdam, The Netherlands
| | - Elena Verzoni
- SSD Genitourinary Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
137
|
Chiong E, Murphy DG, Buchan N, Chen K, Chen SS, Chua MLK, Hamid AR, Kanesvaran R, Khochikar M, Letran J, Lojanapiwat B, Mallik I, Ng CF, Ong TA, Poon DMC, Pu YS, Saad M, Schubach K, Takahara K, Tey J, Thang SP, Toh PC, Türkeri L, Vinh NT, Williams S, Ye D, Davis ID. Management of advanced prostate cancer in the Asia-Pacific region: Summary of the Asia-Pacific Advanced Prostate Cancer Consensus Conference 2023. Asia Pac J Clin Oncol 2024; 20:481-490. [PMID: 38628049 DOI: 10.1111/ajco.14064] [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: 11/02/2023] [Accepted: 03/20/2024] [Indexed: 07/24/2024]
Abstract
AIM The aim of the third Asia-Pacific Advanced Prostate Cancer Consensus Conference (APAC APCCC 2023) was to discuss the application in the Asia-Pacific (APAC) region of consensus statements from the 4th Advanced Prostate Cancer Consensus Conference (APCCC 2022). METHODS The one-day meeting in July 2023 brought together 27 experts from 14 APAC countries. The meeting covered five topics: (1) Intermediate- and high-risk and locally advanced prostate cancer; (2) Management of newly diagnosed metastatic hormone-sensitive prostate cancer; (3) Management of non-metastatic castration-resistant prostate cancer; (4) Homologous recombination repair mutation testing; (5) Management of metastatic castration-resistant prostate cancer. Pre- and post-symposium polling gathered APAC-specific responses to APCCC consensus questions and insights on current practices and challenges in the APAC region. RESULTS APAC APCCC highlights APAC-specific considerations in an evolving landscape of diagnostic technologies and treatment innovations for advanced prostate cancer. While new technologies are available in the region, cost and reimbursement continue to influence practice significantly. Individual patient considerations, including the impact of chemophobia on Asian patients, also influence decision-making. CONCLUSION The use of next-generation imaging, genetic testing, and new treatment combinations is increasing the complexity and duration of prostate cancer management. Familiarity with new diagnostic and treatment options is growing in the APAC region. Insights highlight the continued importance of a multidisciplinary approach that includes nuclear medicine, genetic counseling, and quality-of-life expertise. The APAC APCCC meeting provides an important opportunity to share practice and identify APAC-specific issues and considerations in areas of low evidence where clinical experience is growing.
Collapse
Affiliation(s)
- Edmund Chiong
- Department of Urology, National University Hospital, and Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Nicholas Buchan
- Department of Urology, Christchurch Public Hospital, Christchurch, New Zealand
- Monash University, Melbourne, Australia, Eastern Health, Melbourne, Australia
| | - Kenneth Chen
- Department of Urology, Singapore General Hospital, Singapore, Singapore
| | - Sarah S Chen
- Department of Nursing, Tan Tock Seng Hospital, Singapore, Singapore
| | - Melvin L K Chua
- Department of Head Neck and Thoracic Cancers, Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Agus Rizal Hamid
- Department of Urology, Faculty of Medicine Universitas Indonesia - CiptoMangunkusumo Hospital, Jakarta, Indonesia
| | - Ravindran Kanesvaran
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Makarand Khochikar
- Department of Urology/Uro-oncology, Ushakal Abhinav Institute of Medical Sciences, Sangli, India
| | - Jason Letran
- Department of Urology, Chinese General Hospital and Medical Center, Manila, Philippines
| | | | - Indranil Mallik
- Department of Radiation Oncology, Tata Medical Center, Kolkata, India
| | - Chee Fai Ng
- SH Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Teng Aik Ong
- Department of Surgery, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Darren M C Poon
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Yeong-Shiau Pu
- Department of Urology, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Marniza Saad
- Department of Clinical Oncology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kathryn Schubach
- Monash University, Melbourne, Australia, Eastern Health, Melbourne, Australia
- Australian and New Zealand Urology Nurses (ANZUNS), Melbourne, Australia
| | | | - Jeremy Tey
- Department of Radiation Oncology, National University Cancer Institute Singapore, Singapore, Singapore
| | - Sue-Ping Thang
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Poh Choo Toh
- Department of Urology, National University Hospital, and Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Levent Türkeri
- Department of Urology, Altunizade Hospital, Acibadem M.A. Aydinlar University, Istanbul, Turkey
| | - Nguyễn Tuấn Vinh
- Department of Urology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
| | - Scott Williams
- Monash University, Melbourne, Australia, Eastern Health, Melbourne, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | | |
Collapse
|
138
|
Quintanal-Villalonga A, Kawasaki K, Redin E, Uddin F, Rakhade S, Durani V, Sabet A, Shafer M, Karthaus WR, Zaidi S, Zhan YA, Manoj P, Sridhar H, Kinyua D, Zhong H, Mello BP, Ciampricotti M, Bhanot UK, Linkov I, Qiu J, Patel RA, Morrissey C, Mehta S, Barnes J, Haffner MC, Socci ND, Koche RP, de Stanchina E, Molina-Pinelo S, Salehi S, Yu HA, Chan JM, Rudin CM. CDC7 inhibition impairs neuroendocrine transformation in lung and prostate tumors through MYC degradation. Signal Transduct Target Ther 2024; 9:189. [PMID: 39054323 PMCID: PMC11272780 DOI: 10.1038/s41392-024-01908-y] [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: 02/19/2024] [Revised: 06/20/2024] [Accepted: 06/30/2024] [Indexed: 07/27/2024] Open
Abstract
Neuroendocrine (NE) transformation is a mechanism of resistance to targeted therapy in lung and prostate adenocarcinomas leading to poor prognosis. Up to date, even if patients at high risk of transformation can be identified by the occurrence of Tumor Protein P53 (TP53) and Retinoblastoma Transcriptional Corepressor 1 (RB1) mutations in their tumors, no therapeutic strategies are available to prevent or delay histological transformation. Upregulation of the cell cycle kinase Cell Division Cycle 7 (CDC7) occurred in tumors during the initial steps of NE transformation, already after TP53/RB1 co-inactivation, leading to induced sensitivity to the CDC7 inhibitor simurosertib. CDC7 inhibition suppressed NE transdifferentiation and extended response to targeted therapy in in vivo models of NE transformation by inducing the proteasome-mediated degradation of the MYC Proto-Oncogen (MYC), implicated in stemness and histological transformation. Ectopic overexpression of a degradation-resistant MYC isoform reestablished the NE transformation phenotype observed on targeted therapy, even in the presence of simurosertib. CDC7 inhibition also markedly extended response to standard cytotoxics (cisplatin, irinotecan) in lung and prostate small cell carcinoma models. These results nominate CDC7 inhibition as a therapeutic strategy to constrain lineage plasticity, as well as to effectively treat NE tumors de novo or after transformation. As simurosertib clinical efficacy trials are ongoing, this concept could be readily translated for patients at risk of transformation.
Collapse
Affiliation(s)
- Alvaro Quintanal-Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Kenta Kawasaki
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Esther Redin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fathema Uddin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Swanand Rakhade
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Vidushi Durani
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Amin Sabet
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Moniquetta Shafer
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Wouter R Karthaus
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Samir Zaidi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingqian A Zhan
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parvathy Manoj
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harsha Sridhar
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dennis Kinyua
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hong Zhong
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barbara P Mello
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Metamia Ciampricotti
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Umesh K Bhanot
- Pathology Core Facility, Department of Pathology and Laboratory Medicine, MSKCC, New York, NY, USA
| | - Irina Linkov
- Pathology Core Facility, Department of Pathology and Laboratory Medicine, MSKCC, New York, NY, USA
| | - Juan Qiu
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Radhika A Patel
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Sanjoy Mehta
- Gene Editing & Screening Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jesse Barnes
- Gene Editing & Screening Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael C Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Nicholas D Socci
- Bioinformatics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sonia Molina-Pinelo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, Seville, Spain
| | - Sohrab Salehi
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Helena A Yu
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Joseph M Chan
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
139
|
Murphy KC, DeMarco KD, Zhou L, Lopez-Diaz Y, Ho YJ, Li J, Bai S, Simin K, Zhu LJ, Mercurio AM, Ruscetti M. MYC and p53 alterations cooperate through VEGF signaling to repress cytotoxic T cell and immunotherapy responses in prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604943. [PMID: 39091883 PMCID: PMC11291169 DOI: 10.1101/2024.07.24.604943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Patients with castration-resistant prostate cancer (CRPC) are generally unresponsive to tumor targeted and immunotherapies. Whether genetic alterations acquired during the evolution of CRPC impact immune and immunotherapy responses is largely unknown. Using our innovative electroporation-based mouse models, we generated distinct genetic subtypes of CRPC found in patients and uncovered unique immune microenvironments. Specifically, mouse and human prostate tumors with MYC amplification and p53 disruption had weak cytotoxic lymphocyte infiltration and an overall dismal prognosis. MYC and p53 cooperated to induce tumor intrinsic secretion of VEGF, which by signaling through VEGFR2 expressed on CD8+ T cells, could directly inhibit T cell activity. Targeting VEGF-VEGFR2 signaling in vivo led to CD8+ T cell-mediated tumor and metastasis growth suppression and significantly increased overall survival in MYC and p53 altered CPRC. VEGFR2 blockade also led to induction of PD-L1, and in combination with PD-L1 immune checkpoint blockade produced anti-tumor efficacy in multiple preclinical CRPC mouse models. Thus, our results identify a genetic mechanism of immune suppression through VEGF signaling in prostate cancer that can be targeted to reactivate immune and immunotherapy responses in an aggressive subtype of CRPC. Significance Though immune checkpoint blockade (ICB) therapies can achieve curative responses in many treatment-refractory cancers, they have limited efficacy in CRPC. Here we identify a genetic mechanism by which VEGF contributes to T cell suppression, and demonstrate that VEGFR2 blockade can potentiate the effects of PD-L1 ICB to immunologically treat CRPC.
Collapse
Affiliation(s)
- Katherine C. Murphy
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelly D. DeMarco
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lin Zhou
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yvette Lopez-Diaz
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yu-jui Ho
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Junhui Li
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shi Bai
- Department of Pathology, University of Massachusetts Medical Center, Worcester, MA, USA
| | - Karl Simin
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Arthur M. Mercurio
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Immunology and Microbiology Program, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
140
|
Frank S, Persse T, Coleman I, Bankhead A, Li D, De-Sarkar N, Wilson D, Rudoy D, Vashisth M, Galipeau P, Yang M, Hanratty B, Dumpit R, Morrissey C, Corey E, Montgomery RB, Haffner MC, Pritchard C, Vasioukhin V, Ha G, Nelson PS. Molecular consequences of acute versus chronic CDK12 loss in prostate carcinoma nominates distinct therapeutic strategies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603734. [PMID: 39071291 PMCID: PMC11275783 DOI: 10.1101/2024.07.16.603734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Genomic loss of the transcriptional kinase CDK12 occurs in ~6% of metastatic castration-resistant prostate cancers (mCRPC) and correlates with poor patient outcomes. Prior studies demonstrate that acute CDK12 loss confers a homologous recombination (HR) deficiency (HRd) phenotype via premature intronic polyadenylation (IPA) of key HR pathway genes, including ATM. However, mCRPC patients have not demonstrated benefit from therapies that exploit HRd such as inhibitors of polyADP ribose polymerase (PARP). Based on this discordance, we sought to test the hypothesis that an HRd phenotype is primarily a consequence of acute CDK12 loss and the effect is greatly diminished in prostate cancers adapted to CDK12 loss. Analyses of whole genome sequences (WGS) and RNA sequences (RNAseq) of human mCRPCs determined that tumors with biallelic CDK12 alterations (CDK12 BAL ) lack genomic scar signatures indicative of HRd, despite carrying bi-allelic loss and the appearance of the hallmark tandem-duplicator phenotype (TDP). Experiments confirmed that acute CDK12 inhibition resulted in aberrant polyadenylation and downregulation of long genes (including BRCA1 and BRCA2) but such effects were modest or absent in tumors adapted to chronic CDK12 BAL . One key exception was ATM, which did retain transcript shortening and reduced protein expression in the adapted CDK12 BAL models. However, CDK12 BAL cells demonstrated intact HR as measured by RAD51 foci formation following irradiation. CDK12 BAL cells showed a vulnerability to targeting of CDK13 by sgRNA or CDK12/13 inhibitors and in vivo treatment of prostate cancer xenograft lines showed that tumors with CDK12 BAL responded to the CDK12/13 inhibitor SR4835, while CDK12-intact lines did not. Collectively, these studies show that aberrant polyadenylation and long HR gene downregulation is primarily a consequence of acute CDK12 deficiency, which is largely compensated for in cells that have adapted to CDK12 loss. These results provide an explanation for why PARPi monotherapy has thus far failed to consistently benefit patients with CDK12 alterations, though alternate therapies that target CDK13 or transcription are candidates for future research and testing.
Collapse
Affiliation(s)
- Sander Frank
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Thomas Persse
- Divison of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Ilsa Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Armand Bankhead
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Dapei Li
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Navonil De-Sarkar
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226
- Research Member, Medical College of Wisconsin Cancer Center, WI-53226
| | - Divin Wilson
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, 53226
- Research Member, Medical College of Wisconsin Cancer Center, WI-53226
| | - Dmytro Rudoy
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Manasvita Vashisth
- Divison of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Patty Galipeau
- Divison of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Michael Yang
- Divison of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Brian Hanratty
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Ruth Dumpit
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA 98195
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195
| | | | - Michael C. Haffner
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195
| | - Colin Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195
| | - Valera Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Gavin Ha
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
- Divison of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98119
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98119
- Divison of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98119
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA 98119
- Department of Urology, University of Washington, Seattle, WA 98195
- Department of Medicine, University of Washington, Seattle, WA 98195
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195
| |
Collapse
|
141
|
Ku SY, Wang Y, Garcia MM, Yamada Y, Mizuno K, Long MD, Rosario S, Chinnam M, Al Assaad M, Puca L, Kim MJ, Bakht MK, Venkadakrishnan VB, Robinson BD, Acosta AM, Wadosky KM, Mosquera JM, Goodrich DW, Beltran H. Notch signaling suppresses neuroendocrine differentiation and alters the immune microenvironment in advanced prostate cancer. J Clin Invest 2024; 134:e175217. [PMID: 39024561 PMCID: PMC11364388 DOI: 10.1172/jci175217] [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: 08/28/2023] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
Notch signaling can have either an oncogenic or tumor-suppressive function in cancer depending on the cancer type and cellular context. While Notch can be oncogenic in early prostate cancer, we identified significant downregulation of the Notch pathway during prostate cancer progression from adenocarcinoma to neuroendocrine (NE) prostate cancer, where it functions as a tumor suppressor. Activation of Notch in NE and Rb1/Trp53-deficient prostate cancer models led to phenotypic conversion toward a more indolent, non-NE state with glandular features and expression of luminal lineage markers. This was accompanied by upregulation of MHC and type I IFN and immune cell infiltration. Overall, these data support Notch signaling as a suppressor of NE differentiation in advanced prostate cancer and provide insights into how Notch signaling influences lineage plasticity and the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Sheng-Yu Ku
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Maria Mica Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Yasutaka Yamada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kei Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Mark D. Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Spencer Rosario
- Department of Pharmacology and Therapeutics and
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | | | | | - Loredana Puca
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Min Jin Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Martin K. Bakht
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | - Andrés M. Acosta
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | | | - David W. Goodrich
- Department of Pharmacology and Therapeutics and
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
142
|
Broderick A, Pan E, Li J, Chu A, Hwang C, Barata PC, Cackowski FC, Labriola M, Ghose A, Bilen MA, Kilari D, Thapa B, Piero M, Graham L, Tripathi A, Garje R, Koshkin VS, Hernandez E, Dorff TB, Schweizer MT, Alva AS, McKay RR, Armstrong AJ. Clinical implications of Wnt pathway genetic alterations in men with advanced prostate cancer. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00869-1. [PMID: 39019980 PMCID: PMC11739431 DOI: 10.1038/s41391-024-00869-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Aberrant Wnt signaling has been implicated in prostate cancer tumorigenesis and metastasis in preclinical models but the impact of genetic alterations in Wnt signaling genes in men with advanced prostate cancer is unknown. METHODS We utilized the Prostate Cancer Precision Medicine Multi-Institutional Collaborative Effort (PROMISE) clinical-genomic database for this retrospective analysis. Patients with activating mutations in CTNNB1 or RSPO2 or inactivating mutations in APC, RNF43, or ZNRF3 were defined as Wnt-altered, while those lacking such alterations were defined as Wnt non-altered. We compared patient characteristics and clinical outcomes as well as co-occurring genetic alterations according to Wnt alteration status. RESULTS Of the 1498 patients included, 193 (12.9%) were Wnt-altered. These men had a statistically significant 2-fold increased prevalence of liver and lung metastases as compared with Wnt non-altered patients at the time of initial diagnosis, (4.66% v 2.15% ; 6.22% v 3.07%), first metastatic disease diagnosis (10.88% v 5.29%; 13.99% v 6.21%), and CRPC development (11.40% v 6.36%; 12.95% v 5.29%). Wnt alterations were associated with more co-occurring alterations in RB1 (10.4% v 6.2%), AR (38.9% vs 25.7%), SPOP (13.5% vs 4.1%), FOXA1 (6.7% vs 2.8%), and PIK3CA (10.9% vs 5.1%). We found no significant differences in overall survival or other clinical outcomes from initial diagnosis, first metastatic disease, diagnosis of CRPC, or with AR inhibition for mCRPC between the Wnt groups. CONCLUSIONS Wnt-altered patients with prostate cancer have a higher prevalence of visceral metastases and are enriched in RB1, AR, SPOP, FOXA1, and PIK3CA alterations. Despite these associations, Wnt alterations were not associated with worse survival or treatment outcomes in men with advanced prostate cancer.
Collapse
Affiliation(s)
- Amanda Broderick
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA
| | - Elizabeth Pan
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Jinju Li
- Rogel Cancer Center, Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Alec Chu
- Pathology Department, University of Michigan, Ann Arbor, MI, USA
| | - Clara Hwang
- Division of Hematology/Oncology, Department of Internal Medicine, Henry Ford Cancer Institute, Detroit, MI, USA
| | - Pedro C Barata
- Department of Internal Medicine, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | | | - Matthew Labriola
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA
| | - Alyssa Ghose
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | | | - Deepak Kilari
- Department of Medicine, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bicky Thapa
- Department of Medicine, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael Piero
- Department of Medicine, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Laura Graham
- University of Colorado Cancer Center Anschutz Medical Campus, Aurora, CO, USA
| | - Abhishek Tripathi
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Rohan Garje
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Vadim S Koshkin
- University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Erik Hernandez
- University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Tanya B Dorff
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Ajjai Shivaram Alva
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
143
|
Weiss S, Lamy P, Rusan M, Nørgaard M, Ulhøi BP, Knudsen M, Kassentoft CG, Farajzadeh L, Jensen JB, Pedersen JS, Borre M, Sørensen KD. Exploring the tumor genomic landscape of aggressive prostate cancer by whole-genome sequencing of tissue or liquid biopsies. Int J Cancer 2024; 155:298-313. [PMID: 38602058 DOI: 10.1002/ijc.34949] [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: 08/15/2023] [Revised: 01/19/2024] [Accepted: 03/12/2024] [Indexed: 04/12/2024]
Abstract
Treatment resistance remains a major issue in aggressive prostate cancer (PC), and novel genomic biomarkers may guide better treatment selection. Circulating tumor DNA (ctDNA) can provide minimally invasive information about tumor genomes, but the genomic landscape of aggressive PC based on whole-genome sequencing (WGS) of ctDNA remains incompletely characterized. Thus, we here performed WGS of tumor tissue (n = 31) or plasma ctDNA (n = 10) from a total of 41 aggressive PC patients, including 11 hormone-naïve, 15 hormone-sensitive, and 15 castration-resistant patients. Across all variant types, we found progressively more altered tumor genomic profiles in later stages of aggressive PC. The potential driver genes most frequently affected by single-nucleotide variants or insertions/deletions included the known PC-related genes TP53, CDK12, and PTEN and the novel genes COL13A1, KCNH3, and SENP3. Etiologically, aggressive PC was associated with age-related and DNA repair-related mutational signatures. Copy number variants most frequently affected 14q11.2 and 8p21.2, where no well-recognized PC-related genes are located, and also frequently affected regions near the known PC-related genes MYC, AR, TP53, PTEN, and BRCA1. Structural variants most frequently involved not only the known PC-related genes TMPRSS2 and ERG but also the less extensively studied gene in this context, PTPRD. Finally, clinically actionable variants were detected throughout all stages of aggressive PC and in both plasma and tissue samples, emphasizing the potential clinical applicability of WGS of minimally invasive plasma samples. Overall, our study highlights the feasibility of using liquid biopsies for comprehensive genomic characterization as an alternative to tissue biopsies in advanced/aggressive PC.
Collapse
Affiliation(s)
- Simone Weiss
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Philippe Lamy
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Maria Rusan
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Maibritt Nørgaard
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Michael Knudsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Gødstrup Hospital, Gødstrup, Denmark
| | - Jakob Skou Pedersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Michael Borre
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Karina Dalsgaard Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
144
|
Chaudhary U, Banerjee S. Decoding the Non-coding: Tools and Databases Unveiling the Hidden World of "Junk" RNAs for Innovative Therapeutic Exploration. ACS Pharmacol Transl Sci 2024; 7:1901-1915. [PMID: 39022352 PMCID: PMC11249652 DOI: 10.1021/acsptsci.3c00388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024]
Abstract
Non-coding RNAs are pivotal regulators of gene and protein expression, exerting crucial influences on diverse biological processes. Their dysregulation is frequently implicated in the onset and progression of diseases, notably cancer. A profound comprehension of the intricate mechanisms governing ncRNAs is imperative for devising innovative therapeutic interventions against these debilitating conditions. Significantly, nearly 80% of our genome comprises ncRNAs, underscoring their centrality in cellular processes. The elucidation of ncRNA functions is pivotal for grasping the complexities of gene regulation and its implications for human health. Modern genome sequencing techniques yield vast datasets, stored in specialized databases. To harness this wealth of information and to understand the crosstalk of non-coding RNAs, knowledge of available databases is required, and many new sophisticated computational tools have emerged. These tools play a pivotal role in the identification, prediction, and annotation of ncRNAs, thereby facilitating their experimental validation. This Review succinctly outlines the current understanding of ncRNAs, emphasizing their involvement in disease development. It also highlights the databases and tools instrumental in classifying, annotating, and evaluating ncRNAs. By extracting meaningful biological insights from seemingly "junk" data, these tools empower scientists to unravel the intricate roles of ncRNAs in shaping human health.
Collapse
Affiliation(s)
- Uma Chaudhary
- Department of Biotechnology,
School of Biosciences and Technology, Vellore
Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Satarupa Banerjee
- Department of Biotechnology,
School of Biosciences and Technology, Vellore
Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| |
Collapse
|
145
|
Hofstad M, Woods A, Parra K, Sychev ZE, Mazzagatti A, Yu L, Gilbreath C, Ly P, Drake JM, Kittler R. Dual inhibition of ATR and DNA-PKcs radiosensitizes ATM-mutant prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602941. [PMID: 39026771 PMCID: PMC11257504 DOI: 10.1101/2024.07.10.602941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
In advanced castration resistant prostate cancer (CRPC), mutations in the DNA damage response (DDR) gene ataxia telangiectasia mutated ( ATM ) are common. While poly(ADP-ribose) polymerase inhibitors are approved in this context, their clinical efficacy remains limited. Thus, there is a compelling need to identify alternative therapeutic avenues for ATM mutant prostate cancer patients. Here, we generated matched ATM-proficient and ATM-deficient CRPC lines to elucidate the impact of ATM loss on DDR in response to DNA damage via irradiation. Through unbiased phosphoproteomic screening, we unveiled that ATM-deficient CRPC lines maintain dependence on downstream ATM targets through activation of ATR and DNA-PKcs kinases. Dual inhibition of ATR and DNA-PKcs effectively inhibited downstream γH2AX foci formation in response to irradiation and radiosensitized ATM-deficient lines to a greater extent than either ATM-proficient controls or single drug treatment. Further, dual inhibition abrogated residual downstream ATM pathway signaling and impaired replication fork dynamics. To circumvent potential toxicity, we leveraged the RUVBL1/2 ATPase inhibitor Compound B, which leads to the degradation of both ATR and DNA-PKcs kinases. Compound B effectively radiosensitized ATM-deficient CRPC in vitro and in vivo , and impacted replication fork dynamics. Overall, dual targeting of both ATR and DNA-PKcs is necessary to block DDR in ATM-deficient CRPC, and Compound B could be utilized as a novel therapy in combination with irradiation in these patients.
Collapse
|
146
|
Foley GR, Marthick JR, Lucas SE, Raspin K, Banks A, Stanford JL, Ostrander EA, FitzGerald LM, Dickinson JL. Germline Sequencing of DNA Damage Repair Genes in Two Hereditary Prostate Cancer Cohorts Reveals New Disease Risk-Associated Gene Variants. Cancers (Basel) 2024; 16:2482. [PMID: 39001544 PMCID: PMC11240467 DOI: 10.3390/cancers16132482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Rare, inherited variants in DNA damage repair (DDR) genes have a recognised role in prostate cancer (PrCa) susceptibility. In addition, these genes are therapeutically targetable. While rare variants are informing clinical management in other common cancers, defining the rare disease-associated variants in PrCa has been challenging. Here, whole-genome and -exome sequencing data from two independent, high-risk Australian and North American familial PrCa datasets were interrogated for novel DDR risk variants. Rare DDR gene variants (predicted to be damaging and present in two or more family members) were identified and subsequently genotyped in 1963 individuals (700 familial and 459 sporadic PrCa cases, 482 unaffected relatives, and 322 screened controls), and association analyses accounting for relatedness (MQLS) undertaken. In the combined datasets, rare ERCC3 (rs145201970, p = 2.57 × 10-4) and BRIP1 (rs4988345, p = 0.025) variants were significantly associated with PrCa risk. A PARP2 (rs200603922, p = 0.028) variant in the Australian dataset and a MUTYH (rs36053993, p = 0.031) variant in the North American dataset were also associated with risk. Evaluation of clinicopathological characteristics provided no evidence for a younger age or higher-grade disease at diagnosis in variant carriers, which should be taken into consideration when determining genetic screening eligibility criteria for targeted, gene-based treatments in the future. This study adds valuable knowledge to our understanding of PrCa-associated DDR genes, which will underpin effective clinical screening and treatment strategies.
Collapse
Affiliation(s)
- Georgea R Foley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - James R Marthick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Sionne E Lucas
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Kelsie Raspin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Annette Banks
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Janet L Stanford
- Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., M4-B874, Seattle, WA 98109, USA
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liesel M FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
147
|
Hänze J, Mengen LM, Mernberger M, Tiwari DK, Plagge T, Nist A, Subtil FSB, Theiss U, Eberle F, Roth K, Lauth M, Hofmann R, Engenhart-Cabillic R, Stiewe T, Hegele A. Transcriptomic response of prostate cancer cells to carbon ion and photon irradiation with focus on androgen receptor and TP53 signaling. Radiat Oncol 2024; 19:85. [PMID: 38956684 PMCID: PMC11218163 DOI: 10.1186/s13014-024-02480-z] [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: 01/03/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Radiotherapy is essential in the treatment of prostate cancer. An alternative to conventional photon radiotherapy is the application of carbon ions, which provide a superior intratumoral dose distribution and less induced damage to adjacent healthy tissue. A common characteristic of prostate cancer cells is their dependence on androgens which is exploited therapeutically by androgen deprivation therapy in the advanced prostate cancer stage. Here, we aimed to analyze the transcriptomic response of prostate cancer cells to irradiation by photons in comparison to carbon ions, focusing on DNA damage, DNA repair and androgen receptor signaling. METHODS Prostate cancer cell lines LNCaP (functional TP53 and androgen receptor signaling) and DU145 (dysfunctional TP53 and androgen receptor signaling) were irradiated by photons or carbon ions and the subsequent DNA damage was assessed by immuno-cytofluorescence. Furthermore, the cells were treated with an androgen-receptor agonist. The effects of irradiation and androgen treatment on the gene regulation and the transcriptome were investigated by RT-qPCR and RNA sequencing, followed by bioinformatic analysis. RESULTS Following photon or carbon ion irradiation, both LNCaP and DU145 cells showed a dose-dependent amount of visible DNA damage that decreased over time, indicating occurring DNA repair. In terms of gene regulation, mRNAs involved in the TP53-dependent DNA damage response were significantly upregulated by photons and carbon ions in LNCaP but not in DU145 cells, which generally showed low levels of gene regulation after irradiation. Both LNCaP and DU145 cells responded to photons and carbon ions by downregulation of genes involved in DNA repair and cell cycle, partially resembling the transcriptome response to the applied androgen receptor agonist. Neither photons nor carbon ions significantly affected canonical androgen receptor-dependent gene regulation. Furthermore, certain genes that were specifically regulated by either photon or carbon ion irradiation were identified. CONCLUSION Photon and carbon ion irradiation showed a significant congruence in terms of induced signaling pathways and transcriptomic responses. These responses were strongly impacted by the TP53 status. Nevertheless, irradiation mode-dependent distinct gene regulations with undefined implication for radiotherapy outcome were revealed. Androgen receptor signaling and irradiations shared regulation of certain genes with respect to DNA-repair and cell-cycle.
Collapse
Affiliation(s)
- Jörg Hänze
- Department of Urology, Faculty of Medicine, Philipps University Marburg, Baldingerstraße, 35043, Marburg, Germany.
| | - Lilly M Mengen
- Department of Urology, Faculty of Medicine, Philipps University Marburg, Baldingerstraße, 35043, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Genomics Core Facility, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Dinesh Kumar Tiwari
- Department of Radiotherapy and Radiooncology, Philipps University Marburg, Marburg, Germany
| | - Thomas Plagge
- Department of Urology, Faculty of Medicine, Philipps University Marburg, Baldingerstraße, 35043, Marburg, Germany
| | - Andrea Nist
- Institute of Molecular Oncology, Genomics Core Facility, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Florentine S B Subtil
- Department of Radiotherapy and Radiooncology, Philipps University Marburg, Marburg, Germany
| | - Ulrike Theiss
- Department of Radiotherapy and Radiooncology, Philipps University Marburg, Marburg, Germany
- Marburg Ion-Beam Therapy Center (MIT), Department of Radiotherapy and Radiation Oncology, Marburg University Hospital, Marburg, Germany
| | - Fabian Eberle
- Department of Radiotherapy and Radiooncology, Philipps University Marburg, Marburg, Germany
- Marburg Ion-Beam Therapy Center (MIT), Department of Radiotherapy and Radiation Oncology, Marburg University Hospital, Marburg, Germany
| | - Katrin Roth
- Core Facility Cellular Imaging, Philipps University Marburg, Marburg, Germany
| | - Matthias Lauth
- Center for Tumor and Immune Biology, Philipps University Marburg, Marburg, Germany
| | - Rainer Hofmann
- Department of Urology, Faculty of Medicine, Philipps University Marburg, Baldingerstraße, 35043, Marburg, Germany
| | - Rita Engenhart-Cabillic
- Department of Radiotherapy and Radiooncology, Philipps University Marburg, Marburg, Germany
- Marburg Ion-Beam Therapy Center (MIT), Department of Radiotherapy and Radiation Oncology, Marburg University Hospital, Marburg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Genomics Core Facility, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Axel Hegele
- Department of Radiotherapy and Radiooncology, Philipps University Marburg, Marburg, Germany
- Urological Center Mittelhessen, DRK Hospital Biedenkopf, Biedenkopf, Germany
| |
Collapse
|
148
|
Charlton PV, O'Reilly D, Philippou Y, Rao SR, Lamb ADG, Mills IG, Higgins GS, Hamdy FC, Verrill C, Buffa FM, Bryant RJ. Molecular analysis of archival diagnostic prostate cancer biopsies identifies genomic similarities in cases with progression post-radiotherapy, and those with de novo metastatic disease. Prostate 2024; 84:977-990. [PMID: 38654435 PMCID: PMC11253896 DOI: 10.1002/pros.24715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/18/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND It is important to identify molecular features that improve prostate cancer (PCa) risk stratification before radical treatment with curative intent. Molecular analysis of historical diagnostic formalin-fixed paraffin-embedded (FFPE) prostate biopsies from cohorts with post-radiotherapy (RT) long-term clinical follow-up has been limited. Utilizing parallel sequencing modalities, we performed a proof-of-principle sequencing analysis of historical diagnostic FFPE prostate biopsies. We compared patients with (i) stable PCa (sPCa) postprimary or salvage RT, (ii) progressing PCa (pPCa) post-RT, and (iii) de novo metastatic PCa (mPCa). METHODS A cohort of 19 patients with diagnostic prostate biopsies (n = 6 sPCa, n = 5 pPCa, n = 8 mPCa) and mean 4 years 10 months follow-up (diagnosed 2009-2016) underwent nucleic acid extraction from demarcated malignancy. Samples underwent 3'RNA sequencing (3'RNAseq) (n = 19), nanoString analysis (n = 12), and Illumina 850k methylation (n = 8) sequencing. Bioinformatic analysis was performed to coherently identify differentially expressed genes and methylated genomic regions (MGRs). RESULTS Eighteen of 19 samples provided useable 3'RNAseq data. Principal component analysis (PCA) demonstrated similar expression profiles between pPCa and mPCa cases, versus sPCa. Coherently differentially methylated probes between these groups identified ~600 differentially MGRs. The top 50 genes with increased expression in pPCa patients were associated with reduced progression-free survival post-RT (p < 0.0001) in an external cohort. CONCLUSIONS 3'RNAseq, nanoString and 850k-methylation analyses are each achievable from historical FFPE diagnostic pretreatment prostate biopsies, unlocking the potential to utilize large cohorts of historic clinical samples. Profiling similarities between individuals with pPCa and mPCa suggests biological similarities and historical radiological staging limitations, which warrant further investigation.
Collapse
Affiliation(s)
- Philip Vincent Charlton
- Department of OncologyUniversity of OxfordOxfordUK
- Department of OncologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | | | - Yiannis Philippou
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Srinivasa Rao Rao
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | - Alastair David Gordon Lamb
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | | | - Geoff Stuart Higgins
- Department of OncologyUniversity of OxfordOxfordUK
- Department of OncologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Freddie Charles Hamdy
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | - Clare Verrill
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
- Department of PathologyOxford University Hospitals NHS Foundation TrustOxfordUK
| | | | - Richard John Bryant
- Department of UrologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| |
Collapse
|
149
|
Guida A, Mosillo C, Mammone G, Caserta C, Sirgiovanni G, Conteduca V, Bracarda S. The 5-WS of targeting DNA-damage repair (DDR) pathways in prostate cancer. Cancer Treat Rev 2024; 128:102766. [PMID: 38763054 DOI: 10.1016/j.ctrv.2024.102766] [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: 02/05/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
DNA-damage repair (DDR) pathways alterations, a growing area of interest in oncology, are detected in about 20% of patient with prostate cancer and are associated with improved sensitivity to poly(ADP ribose) polymerases (PARP) inhibitors. In May 2020, the Food and Drug Administration (FDA) approved two PARP inhibitors (olaparib and rucaparib) for prostate cancer treatment. Moreover, germline aberrations in DDR pathways genes have also been related to familial or hereditary prostate cancer, requiring tailored health-care programs. These emerging scenarios are rapidly changing diagnostic, prognostic and therapeutic approaches in prostate cancer management. The aim of this review is to highlight the five W-points of DDR pathways in prostate cancer: why targeting DDR pathways in prostate cancer; what we should test for genomic profiling in prostate cancer; "where" testing genetic assessment in prostate cancer (germline or somatic, solid or liquid biopsy); when genetic testing is appropriate in prostate cancer; who could get benefit from PARP inhibitors; how improve patients outcome with combinations strategies.
Collapse
|
150
|
Paralkar D, Akbari A, Aron M. Prostatic adenocarcinoma: molecular underpinnings and treatment-related options. Urol Oncol 2024; 42:203-210. [PMID: 38508940 DOI: 10.1016/j.urolonc.2024.03.003] [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: 09/19/2023] [Revised: 01/28/2024] [Accepted: 03/03/2024] [Indexed: 03/22/2024]
Abstract
Prostate cancer is heterogeneous with varied pathologic features and presents with a wide spectrum of clinical manifestations from indolent to advanced cancer. Interrogation of the molecular landscape of prostate cancer has unveiled the complex genomic alterations in these tumors, which significantly impacts tumor biology. The documented array of chromosomal alterations, gene fusions, and epigenetic changes not only play a crucial role in oncogenesis and disease progression, but also impacts response and resistance to various therapeutic modalities. Various gene expression assays have been developed and are currently recommended in aiding clinical decision making in these clinically and molecularly heterogeneous cancer. In this review, we provide an overview of the molecular underpinnings of prostate cancer, and briefly review the current status of molecular testing and therapeutic options in the management of these tumors.
Collapse
Affiliation(s)
- Divyangi Paralkar
- Department of Urology, Keck School of Medicine, University of Southern California, 1500 San Pablo Street, Room 2409, HC4, Los Angeles, California
| | - Amir Akbari
- Department of Pathology, Keck School of Medicine, University of Southern California, 1500 San Pablo Street, Room 2409, HC4, Los Angeles, California
| | - Manju Aron
- Department of Urology, Keck School of Medicine, University of Southern California, 1500 San Pablo Street, Room 2409, HC4, Los Angeles, California; Department of Pathology, Keck School of Medicine, University of Southern California, 1500 San Pablo Street, Room 2409, HC4, Los Angeles, California.
| |
Collapse
|