1
|
Pérez-de-Oliveira ME, Wagner VP, Bingle CD, Vargas PA, Bingle L. Disruption of oncogenic pathways in mucoepidermoid carcinoma: CREB inhibitor 666.15 as a potential therapeutic agent. Oral Oncol 2024; 159:107029. [PMID: 39332274 DOI: 10.1016/j.oraloncology.2024.107029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/07/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVES Mucoepidermoid carcinoma (MEC) is the most common malignant salivary gland tumour with around 50 % of cases carrying the CRTC1-MAML2 translocation. The CREB pathway has been associated with the transforming activity of this translocation. The aim of this study was to determine the effects of CREB inhibition on MEC cell behaviour in vitro. MATERIAL AND METHODS Two translocation-positive (UM-HMC-2 and H292) and one translocation-negative (H253) MEC cell lines were treated with 666.15, a CREB inhibitor. Drug IC50 doses were determined for each cell line. Clonogenic and spheroid assays were used to assess survival, including percentage of cancer stem cells, and transwell and scratch assays evaluated invasive and migratory capacities, respectively. Immunofluorescence staining was used to determine E-cadherin expression. RESULTS CREB inhibition significantly reduced the number of surviving colonies and spheroids and delayed cell invasion in all cell lines, but this was more significant in the fusion positive, UM-HMC-2 cells. The expression of E-cadherin was significantly higher in treated UM-HMC-2 and H292 cells. CONCLUSION CREB inhibition with 666.15 impaired key MEC oncogenic behaviours associated with metastasis and drug resistance, including cell invasion and survival.
Collapse
Affiliation(s)
- Maria Eduarda Pérez-de-Oliveira
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Vivian Petersen Wagner
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom; Department of Oral Medicine, School of Dentistry, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Colin D Bingle
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas, Piracicaba, São Paulo, Brazil
| | - Lynne Bingle
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
2
|
Liu GH, Yao ZQ, Chen GQ, Li YL, Liang B. Potential Benefits of Green Tea in Prostate Cancer Prevention and Treatment: A Comprehensive Review. Chin J Integr Med 2024; 30:1045-1055. [PMID: 38561489 DOI: 10.1007/s11655-024-4100-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 04/04/2024]
Abstract
Prostate cancer is a prevalent and debilitating disease that necessitates effective prevention and treatment strategies. Green tea, a well-known beverage derived from the Camellia sinensis plant, contains bioactive compounds with potential health benefits, including catechins and polyphenols. This comprehensive review aims to explore the potential benefits of green tea in prostate cancer prevention and treatment by examining existing literature. Green tea possesses antioxidant, anti-inflammatory, and anti-carcinogenic properties attributed to its catechins, particularly epigallocatechin gallate. Epidemiological studies have reported an inverse association between green tea consumption and prostate cancer risk, with potential protection against aggressive forms of the disease. Laboratory studies demonstrate that green tea components inhibit tumor growth, induce apoptosis, and modulate signaling pathways critical to prostate cancer development and progression. Clinical trials and human studies further support the potential benefits of green tea. Green tea consumption has been found to be associated with a reduction in prostate-specific antigen levels, tumor markers, and played a potential role in slowing disease progression. However, challenges remain, including optimal dosage determination, formulation standardization, and conducting large-scale, long-term clinical trials. The review suggests future research should focus on combinatorial approaches with conventional therapies and personalized medicine strategies to identify patient subgroups most likely to benefit from green tea interventions.
Collapse
Affiliation(s)
- Gui-Hong Liu
- Department of Urology, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, 572000, China
| | - Ze-Qin Yao
- Department of Urology, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, 572000, China
| | - Guo-Qiang Chen
- Department of Urology, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, 572000, China
| | - Ya-Lang Li
- Department of Urology, Yuzhou People's Hospital, Xuchang City, Henan Province, 461670, China
| | - Bing Liang
- Department of Urology, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, 572000, China.
| |
Collapse
|
3
|
Albarracín-Navas L, Almonte-Becerril M, Guerrero E, Rivadeneira J, Telechea-Fernández M, Guzmán E, Calderón F, Hernández-Leal MJ, Otzen T, Manterola C, Duque G, Riffo-Campos ÁL. Differential Protein-Coding Gene Expression Profile in Patients with Prostate Cancer. Biomedicines 2024; 12:2509. [PMID: 39595075 PMCID: PMC11592252 DOI: 10.3390/biomedicines12112509] [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: 07/17/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Prostate cancer is the second most common neoplasm in men, with projections estimating over one million new cases by 2045. Differentially expressed genes can significantly enhance the diagnosis, treatment, monitoring, and prognosis of this disease. PURPOSE to systematically review and analyze validated differentially expressed mRNAs in prostate cancer patients to propose a robust molecular profile for clinical diagnostics. METHODS A systematic review was conducted following PRISMA guidelines, searching literature databases for mRNAs with validated differential expression in adult prostate cancer patients. Identified mRNAs were analyzed using STRING, Cytoscape, and DrugBank to explore protein-protein interactions and potential drug targets. RESULTS A total of 5003 participants from Europe, Asia, America, and Oceania were included, and 144 mRNAs (p < 0.05) were reported across 75 primary articles, predominantly validated using RT-qPCR with tissue samples. Among these, at least 36 mRNAs were identified as targets for cancer-related drugs. Enrichment analysis revealed the top pathways were associated with cancer, including specific prostate cancer terms. Key nodes emerged as hubs in the protein-protein interaction network. CONCLUSION Based on our comprehensive in silico analysis of validated differentially expressed mRNAs, we propose a molecular profile of twenty-five mRNAs with significant potential for clinical diagnosis of prostate cancer. These findings offer a valuable foundation for developing precision oncology strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Lorena Albarracín-Navas
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Comprehensive Medical Services (SERMEDIC), Cuenca 010111, Ecuador
| | - Maylin Almonte-Becerril
- Executive Direction of Research and Advanced Studies, Universidad de la Salud, Mexico City 01210, Mexico;
| | - Enmanuel Guerrero
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Comprehensive Medical Services (SERMEDIC), Cuenca 010111, Ecuador
- Faculty of Medicine, Universidad de Cuenca, Cuenca 010201, Ecuador
- SOLCA Cancer Institute, Cuenca 010111, Ecuador
| | - Josue Rivadeneira
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Zero Biomedical Research, Quito 170103, Ecuador
| | | | - Elizabeth Guzmán
- Comprehensive Medical Services (SERMEDIC), Cuenca 010111, Ecuador
| | | | - María José Hernández-Leal
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- University of Navarra, School of Nursing, Department of Community, Maternity and Pediatric Nursing, 31006 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Tamara Otzen
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| | - Carlos Manterola
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
- Universidad de La Frontera, Center for Morphological and Surgical Studies (CEMyQ), Temuco 4811230, Chile
| | - Galo Duque
- Faculty of Medicine, Universidad del Azuay, Cuenca 010107, Ecuador;
| | - Ángela L. Riffo-Campos
- Universidad de La Frontera, Ph.D. Program in Medical Sciences, Temuco 4811230, Chile; (L.A.-N.); (E.G.); (J.R.); (M.J.H.-L.); (T.O.); (C.M.)
- Universidad de La Frontera, Millennium Nucleus on Sociomedicine (SocioMed), Temuco 4811230, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| |
Collapse
|
4
|
Kwon WA, Song YS, Lee MK. Strategic Advances in Combination Therapy for Metastatic Castration-Sensitive Prostate Cancer: Current Insights and Future Perspectives. Cancers (Basel) 2024; 16:3187. [PMID: 39335158 PMCID: PMC11430187 DOI: 10.3390/cancers16183187] [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: 08/23/2024] [Revised: 09/14/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
The contemporary treatment for metastatic castration-sensitive prostate cancer (mCSPC) has evolved significantly, building on successes in managing metastatic castration-resistant prostate cancer (mCRPC). Although androgen deprivation therapy (ADT) alone has long been the cornerstone of mCSPC treatment, combination therapies have emerged as the new standard of care based on recent advances, offering improved survival outcomes. Landmark phase 3 trials demonstrated that adding chemotherapy (docetaxel) and androgen receptor pathway inhibitors to ADT significantly enhances overall survival, particularly for patients with high-volume, high-risk, or de novo metastatic disease. Despite these advancements, a concerning gap between evidence-based guidelines and real-world practice remains, with many patients not receiving recommended combination therapies. The challenge in optimizing therapy sequences, considering both disease control and treatment burdens, and identifying clinical and biological subgroups that could benefit from personalized treatment strategies persists. The advent of triplet therapy has shown promise in extending survival, but the uro-oncology community must narrow the gap between evidence and practice to deliver the most effective care. Current research is focused on refining treatment approaches and utilizing biomarkers to guide therapy selection, aiming to offer more personalized and adaptive strategies for mCSPC management. Thus, aligning clinical practices with the evolving evidence is urgently needed to improve outcomes for patients facing this incurable disease.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Min-Kyung Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| |
Collapse
|
5
|
Fieni C, Sorrentino C, Ciummo SL, Fontana A, Lotti LV, Scialis S, Calvo Garcia D, Caulo M, Di Carlo E. Immunoliposome-based targeted delivery of the CRISPR/Cas9gRNA-IL30 complex inhibits prostate cancer and prolongs survival. Exp Mol Med 2024; 56:2033-2051. [PMID: 39232121 PMCID: PMC11447253 DOI: 10.1038/s12276-024-01310-2] [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: 02/05/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 09/06/2024] Open
Abstract
The development of selective and nontoxic immunotherapy targeting prostate cancer (PC) is challenging. Interleukin (IL)30 plays immunoinhibitory and oncogenic roles in PC, and its tumor-specific suppression may have significant clinical implications. CRISPR/Cas9-mediated IL30 gene deletion in PC xenografts using anti-PSCA antibody-driven lipid nanocomplexes (Cas9gRNA-hIL30-PSCA NxPs) revealed significant genome editing efficiency and circulation stability without off-target effects or organ toxicity. Biweekly intravenous administration of Cas9gRNA-hIL30-PSCA NxPs to PC-bearing mice inhibited tumor growth and metastasis and improved survival. Mechanistically, Cas9gRNA-hIL30-PSCA NxPs suppressed ANGPTL 1/2/4, IL1β, CCL2, CXCL1/6, SERPINE1-F1, EFNB2, PLG, PF4, VEGFA, VEGFD, ANG, TGFβ1, EGF and HGF expression in human PC cells while upregulated CDH1, DKK3 and PTEN expression, leading to low proliferation and extensive ischemic necrosis. In the syngeneic PC model, IL30-targeting immunoliposomes downregulated NFKB1 expression and prevented intratumoral influx of CD11b+Gr-1+MDCs, Foxp3+Tregs, and NKp46+RORγt+ILC3, and prolonged host survival by inhibiting tumor progression. This study serves as a proof of principle that immunoliposome-based targeted delivery of Cas9gRNA-IL30 represent a potentially safe and effective strategy for PC treatment.
Collapse
Affiliation(s)
- Cristiano Fieni
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Stefania Livia Ciummo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Antonella Fontana
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UDA-TECHLAB Research Center, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | | | - Sofia Scialis
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Darien Calvo Garcia
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
6
|
Phongsuwichetsak C, Suksrichavalit T, Chatupheeraphat C, Eiamphungporn W, Yainoy S, Yamkamon V. Diospyros rhodocalyx Kurz induces mitochondrial-mediated apoptosis via BAX, Bcl-2, and caspase-3 pathways in LNCaP human prostate cancer cell line. PeerJ 2024; 12:e17637. [PMID: 38966207 PMCID: PMC11223595 DOI: 10.7717/peerj.17637] [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: 12/22/2023] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
Background Prostate cancer (PCa) is one of the causes of death in men worldwide. Although treatment strategies have been developed, the recurrence of the disease and consequential side effects remain an essential concern. Diospyros rhodocalyx Kurz, a traditional Thai medicine, exhibits diverse therapeutic properties, including anti-cancer activity. However, its anti-cancer activity against prostate cancer has not been thoroughly explored. This study aims to evaluate the anti-cancer activity and underlying mechanisms of the ethyl acetate extract of D. rhodocalyx Kurz (EADR) related to apoptosis induction in the LNCaP human prostate cancer cell line. Methods Ethyl acetate was employed to extract the dried bark of D. rhodocalyx Kurz. The cytotoxicity of EADR on both LNCaP and WPMY-1 cells (normal human prostatic myofibroblast cell line) was evaluated using MTS assay. The effect of EADR on the cell cycle, apoptosis induction, and alteration in mitochondrial membrane potential (MMP) was assessed by the staining with propidium iodide (PI), Annexin V-FITC/PI, and JC-1 dye, respectively. Subsequent analysis was conducted using flow cytometry. The expression of cleaved caspase-3, BAX, and Bcl-2 was examined by Western blotting. The phytochemical profiling of the EADR was performed using gas chromatography-mass spectrometry (GC-MS). Results EADR exhibited a dose-dependent manner cytotoxic effect on LNCaP cells, with IC50 values of 15.43 and 12.35 µg/mL after 24 and 48 h, respectively. Although it also exhibited a cytotoxic effect on WPMY-1 cells, the effect was comparatively lower, with the IC50 values of 34.61 and 19.93 µg/mL after 24 and 48 h of exposure, respectively. Cell cycle analysis demonstrated that EADR did not induce cell cycle arrest in either LNCaP or WPMY-1 cells. However, it significantly increased the sub-G1 population in LNCaP cells, indicating a potential induction of apoptosis. The Annexin V-FITC/PI staining indicated that EADR significantly induced apoptosis in LNCaP cells. Subsequent investigation into the underlying mechanism of EADR-induced apoptosis revealed a reduction in MMP as evidenced by JC-1 staining. Moreover, Western blotting demonstrated that EADR treatment resulted in the upregulation of BAX, downregulation of BCL-2, and elevation of caspase-3 cleavage in LNCaP cells. Notably, the epilupeol was a prominent compound in EADR as identified by GC-MS. Conclusion The EADR exhibits anti-cancer activity against the LNCaP human prostate cancer cell line by inducing cytotoxicity and apoptosis. Our findings suggest that EADR promotes apoptosis by upregulating pro-apoptotic BAX, whereas downregulation of anti-apoptotic Bcl-2 results in the reduction of MMP and the activation of caspase-3. Of particular interest is the presence of epilupeol, a major compound identified in EADR, which may hold promise as a candidate for the development of therapeutic agents for prostate cancer.
Collapse
Affiliation(s)
- Chayisara Phongsuwichetsak
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Thummaruk Suksrichavalit
- Department of Clinical Chemistry, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Chawalit Chatupheeraphat
- Center for Research Innovation and Biomedical Information, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Warawan Eiamphungporn
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Sakda Yainoy
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Vichanan Yamkamon
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
7
|
Kulasegaran T, Oliveira N. Metastatic Castration-Resistant Prostate Cancer: Advances in Treatment and Symptom Management. Curr Treat Options Oncol 2024; 25:914-931. [PMID: 38913213 PMCID: PMC11236885 DOI: 10.1007/s11864-024-01215-2] [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] [Accepted: 05/06/2024] [Indexed: 06/25/2024]
Abstract
OPINION STATEMENT The management of metastatic castrate-resistant prostate cancer (mCRPC) has evolved in the past decade due to substantial advances in understanding the genomic landscape and biology underpinning this form of prostate cancer. The implementation of various therapeutic agents has improved overall survival but despite the promising advances in therapeutic options, mCRPC remains incurable. The focus of treatment should be not only to improve survival but also to preserve the patient's quality of life (QoL) and ameliorate cancer-related symptoms such as pain. The choice and sequence of therapy for mCRPC patients are complex and influenced by various factors, such as side effects, disease burden, treatment history, comorbidities, patient preference and, more recently, the presence of actionable genomic alterations or biomarkers. Docetaxel is the first-line treatment for chemo-naïve patients with good performance status and those who have yet to progress on docetaxel in the castration-sensitive setting. Novel androgen agents (NHAs), such as abiraterone and enzalutamide, are effective treatment options that are utilized as second-line options. These medications can be considered upfront in frail patients or patients who are NHA naïve. Current guidelines recommend genetic testing in mCRPC for mutations in DNA repair deficiency genes to inform treatment decisions, as for example in breast cancer gene mutation testing. Other potential biomarkers being investigated include phosphatase and tensin homologues and homologous recombination repair genes. Despite a growing number of studies incorporating biomarkers in their trial designs, to date, only olaparib in the PROFOUND study and lutetium-177 in the VISION trial have improved survival. This is an unmet need, and future trials should focus on biomarker-guided treatment strategies. The advent of novel noncytotoxic agents has enhanced targeted drug delivery and improved treatment responses with favourable toxicity profiling. Trials should continue to incorporate and report health-related QoL scores and functional assessments into their trial designs.
Collapse
Affiliation(s)
- Tivya Kulasegaran
- Mater Hospital Brisbane, Cancer Centre, Raymond Terrace, South Brisbane, QLD, 4104, Australia.
- School of Clinical Medicine, Mater Clinical Unit, The University of Queensland, Brisbane, Queensland, Australia, Raymond Terrace, South Brisbane, QLD, 4101, Australia.
| | - Niara Oliveira
- Mater Hospital Brisbane, Cancer Centre, Raymond Terrace, South Brisbane, QLD, 4104, Australia
- School of Clinical Medicine, Mater Clinical Unit, The University of Queensland, Brisbane, Queensland, Australia, Raymond Terrace, South Brisbane, QLD, 4101, Australia
| |
Collapse
|
8
|
Kelly WK, Danila DC, Lin CC, Lee JL, Matsubara N, Ward PJ, Armstrong AJ, Pook D, Kim M, Dorff TB, Fischer S, Lin YC, Horvath LG, Sumey C, Yang Z, Jurida G, Smith KM, Connarn JN, Penny HL, Stieglmaier J, Appleman LJ. Xaluritamig, a STEAP1 × CD3 XmAb 2+1 Immune Therapy for Metastatic Castration-Resistant Prostate Cancer: Results from Dose Exploration in a First-in-Human Study. Cancer Discov 2024; 14:76-89. [PMID: 37861461 PMCID: PMC10784743 DOI: 10.1158/2159-8290.cd-23-0964] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023]
Abstract
Xaluritamig (AMG 509) is a six-transmembrane epithelial antigen of the prostate 1 (STEAP1)-targeted T-cell engager designed to facilitate lysis of STEAP1-expressing cancer cells, such as those in advanced prostate cancer. This first-in-human study reports monotherapy dose exploration for patients with metastatic castration-resistant prostate cancer (mCRPC), primarily taxane pretreated. Ninety-seven patients received ≥1 intravenous dose ranging from 0.001 to 2.0 mg weekly or every 2 weeks. MTD was identified as 1.5 mg i.v. weekly via a 3-step dose. The most common treatment-related adverse events were cytokine release syndrome (CRS; 72%), fatigue (45%), and myalgia (34%). CRS occurred primarily during cycle 1 and improved with premedication and step dosing. Prostate-specific antigen (PSA) and RECIST responses across cohorts were encouraging [49% PSA50; 24% objective response rate (ORR)], with greater frequency at target doses ≥0.75 mg (59% PSA50; 41% ORR). Xaluritamig is a novel immunotherapy for prostate cancer that has shown encouraging results supporting further development. SIGNIFICANCE Xaluritamig demonstrated encouraging responses (PSA and RECIST) compared with historical established treatments for patients with late-line mCRPC. This study provides proof of concept for T-cell engagers as a potential treatment for prostate cancer, validates STEAP1 as a target, and supports further clinical investigation of xaluritamig in prostate cancer. See related commentary by Hage Chehade et al., p. 20. See related article by Nolan-Stevaux et al., p. 90. This article is featured in Selected Articles from This Issue, p. 5.
Collapse
Affiliation(s)
- William K. Kelly
- Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, Pennsylvania
- Sarah Cannon Research Institute, Nashville, Tennessee
| | - Daniel C. Danila
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Chia-Chi Lin
- National Taiwan University Hospital, Taipei, Taiwan
| | - Jae-Lyun Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | | - Patrick J. Ward
- Sarah Cannon Research Institute, Nashville, Tennessee
- Oncology Hematology Care, Cincinnati, Ohio
| | - Andrew J. Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, North Carolina
| | - David Pook
- Monash Health, Clayton, Victoria, Australia
| | - Miso Kim
- Seoul National University Hospital, Seoul, South Korea
| | | | - Stefanie Fischer
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Yung-Chang Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Lisa G. Horvath
- Chris O'Brien Lifehouse, University of Sydney, Sydney, New South Wales, Australia
| | | | - Zhao Yang
- Amgen Inc., Thousand Oaks, California
| | | | | | | | | | | | | |
Collapse
|
9
|
Fenor de la Maza MD, Pérez Gracia JL, Miñana B, Castro E. PARP inhibitors alone or in combination for prostate cancer. Ther Adv Urol 2024; 16:17562872241272929. [PMID: 39184454 PMCID: PMC11344902 DOI: 10.1177/17562872241272929] [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: 12/08/2024] [Accepted: 06/20/2024] [Indexed: 08/27/2024] Open
Abstract
DNA repair genomic aberrations in the Homologous Recombination pathway are identifiable in up to 25% of patients with advanced prostate cancer, making them more likely to benefit from treatment with poly (ADP-ribose) polymerase inhibitors (PARPi) alone or in combination with other therapies, particularly when BRCA driver genomic aberrations are documented. Although several clinical trials have demonstrated the efficacy of this approach, the validation of reliable biomarkers predictive of response still needs further improvement to refine patient selection. In this setting, the characterization of resistance mechanisms and the validation of novel biomarkers are critical to maximize clinical benefit and to develop novel treatment combinations to improve outcomes. In this review, we summarize the development of PARPi in prostate cancer as single agent as well as the efficacy of their combination with other drugs, and the future directions for their implementation in the management of advanced prostate cancer.
Collapse
Affiliation(s)
| | | | - Bernardino Miñana
- Department of Urology, Clínica Universidad de Navarra, Madrid, Spain
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de octubre, Av. Córdoba s/n, Madrid 28041, Spain
| |
Collapse
|
10
|
Grizzi F, Hegazi MAAA, Zanoni M, Vota P, Toia G, Clementi MC, Mazzieri C, Chiriva-Internati M, Taverna G. Prostate Cancer Microvascular Routes: Exploration and Measurement Strategies. Life (Basel) 2023; 13:2034. [PMID: 37895416 PMCID: PMC10608780 DOI: 10.3390/life13102034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Angiogenesis is acknowledged as a pivotal feature in the pathology of human cancer. Despite the absence of universally accepted markers for gauging the comprehensive angiogenic activity in prostate cancer (PCa) that could steer the formulation of focused anti-angiogenic treatments, the scrutiny of diverse facets of tumoral blood vessel development may furnish significant understanding of angiogenic processes. Malignant neoplasms, encompassing PCa, deploy a myriad of strategies to secure an adequate blood supply. These modalities range from sprouting angiogenesis and vasculogenesis to intussusceptive angiogenesis, vascular co-option, the formation of mosaic vessels, vasculogenic mimicry, the conversion of cancer stem-like cells into tumor endothelial cells, and vascular pruning. Here we provide a thorough review of these angiogenic mechanisms as they relate to PCa, discuss their prospective relevance for predictive and prognostic evaluations, and outline the prevailing obstacles in quantitatively evaluating neovascularization via histopathological examinations.
Collapse
Affiliation(s)
- Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
| | - Mohamed A. A. A. Hegazi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Matteo Zanoni
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Paolo Vota
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Giovanni Toia
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Maria Chiara Clementi
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Cinzia Mazzieri
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| | - Maurizio Chiriva-Internati
- Departments of Gastroenterology, Hepatology & Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Gianluigi Taverna
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- Department of Urology, Humanitas Mater Domini, Castellanza, 21053 Varese, Italy; (M.Z.); (P.V.); (G.T.); (M.C.C.); (C.M.)
| |
Collapse
|
11
|
Bugoye FC, Torrorey-Sawe R, Biegon R, Dharsee N, Mafumiko FMS, Patel K, Mining SK. Mutational spectrum of DNA damage and mismatch repair genes in prostate cancer. Front Genet 2023; 14:1231536. [PMID: 37732318 PMCID: PMC10507418 DOI: 10.3389/fgene.2023.1231536] [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: 05/30/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023] Open
Abstract
Over the past few years, a number of studies have revealed that a significant number of men with prostate cancer had genetic defects in the DNA damage repair gene response and mismatch repair genes. Certain of these modifications, notably gene alterations known as homologous recombination (HRR) genes; PALB2, CHEK2 BRCA1, BRCA2, ATM, and genes for DNA mismatch repair (MMR); MLH1, MSH2, MSH6, and PMS2 are connected to a higher risk of prostate cancer and more severe types of the disease. The DNA damage repair (DDR) is essential for constructing and diversifying the antigen receptor genes required for T and B cell development. But this DDR imbalance results in stress on DNA replication and transcription, accumulation of mutations, and even cell death, which compromises tissue homeostasis. Due to these impacts of DDR anomalies, tumor immunity may be impacted, which may encourage the growth of tumors, the release of inflammatory cytokines, and aberrant immune reactions. In a similar vein, people who have altered MMR gene may benefit greatly from immunotherapy. Therefore, for these treatments, mutational genetic testing is indicated. Mismatch repair gene (MMR) defects are also more prevalent than previously thought, especially in patients with metastatic disease, high Gleason scores, and diverse histologies. This review summarizes the current information on the mutation spectrum and clinical significance of DDR mechanisms, such as HRR and MMR abnormalities in prostate cancer, and explains how patient management is evolving as a result of this understanding.
Collapse
Affiliation(s)
- Fidelis Charles Bugoye
- Government Chemist Laboratory Authority, Directorate of Forensic Science and DNA Services, Dar es Salaam, Tanzania
- Department of Pathology, Moi Teaching and Referral Hospital, Moi University, Eldoret, Kenya
| | - Rispah Torrorey-Sawe
- Department of Pathology, Moi Teaching and Referral Hospital, Moi University, Eldoret, Kenya
| | - Richard Biegon
- Department of Pathology, Moi Teaching and Referral Hospital, Moi University, Eldoret, Kenya
| | | | - Fidelice M. S. Mafumiko
- Government Chemist Laboratory Authority, Directorate of Forensic Science and DNA Services, Dar es Salaam, Tanzania
| | - Kirtika Patel
- Department of Pathology, Moi Teaching and Referral Hospital, Moi University, Eldoret, Kenya
| | - Simeon K. Mining
- Department of Pathology, Moi Teaching and Referral Hospital, Moi University, Eldoret, Kenya
| |
Collapse
|