1
|
Hong X, Zhang Y, Chi Z, Xu Q, Lin W, Huang Y, Lin T, Zhang Y. Efficacy and Safety of Programmed Death-1 (PD-1)/Programmed Death-Ligand 1 (PD-L1) Checkpoint Inhibitors in Patients With Metastatic Castration-resistant Prostate Cancer: A Systematic Review and Meta-analysis. Clin Oncol (R Coll Radiol) 2024; 36:e20-e30. [PMID: 37993317 DOI: 10.1016/j.clon.2023.11.034] [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: 02/17/2023] [Revised: 09/14/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
AIMS The aim of this systematic review with meta-analysis was to evaluate the efficacy and safety of programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) checkpoint inhibitors in patients with metastatic castration-resistant prostate cancer (mCRPC). MATERIALS AND METHODS We searched PubMed, Embase and Cochrane Library until 1 July 2022 for mCRPC trials testing PD-1/PD-L1 checkpoint inhibitors. We measured the efficacy and safety using overall survival, progression-free survival (PFS), overall response rates (ORR), prostate-specific antigen (PSA) response rate or treatment-related adverse events (TRAEs). When possible, data were meta-analysed. RESULTS Thirteen studies involving 2533 participants were included in this meta-analysis. The pooled hazard ratio for overall survival was 0.81 (95% confidence interval 0.42-1.20, I2 = 80.3%, PHeterogeneity<0.001) and for PFS was 0.65 (95% confidence interval 0.38-0.92, I2 = 72.2%, PHeterogeneity = 0.013). Furthermore, better ORR (relative risk = 2.77, 95% confidence interval 1.25-6.13, I2 = 0%, PHeterogeneity = 0.699) was found in PD-L1-expressing tumours. However, no statistical trends between PD-L1 status on PSA response rate (relative risk = 0.79, 95% confidence interval 0.5-1.25, I2 = 0%, PHeterogeneity = 0.953) and tumour mutational burden on ORR (relative risk = 2.53, 95% confidence interval 0.49-13.12, I2 = 74.5%, PHeterogeneity = 0.02) were observed. The pooled proportions of TRAEs and ≥ grade 3 TRAEs were 85.1% (95% confidence interval = 71.7-98.5%) and 31.6% (95% confidence interval = 18.9-44.4%), respectively. CONCLUSIONS This meta-analysis showed that among selected populations of men with mCRPC, anti-PD-1/PD-L1 combination treatment may significantly increase the PFS benefits. However, overall survival in mCRPC warrants further testing.
Collapse
Affiliation(s)
- X Hong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Department of Urology, Shantou Central Hospital, Shantou, PR China
| | - Y Zhang
- Department of Urology, Shantou Central Hospital, Shantou, PR China
| | - Z Chi
- Department of Urology, Shantou Central Hospital, Shantou, PR China
| | - Q Xu
- Department of Urology, Shantou Central Hospital, Shantou, PR China
| | - W Lin
- Department of Urology, Shantou Central Hospital, Shantou, PR China
| | - Y Huang
- Department of Urology, Shantou Central Hospital, Shantou, PR China
| | - T Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China.
| | - Y Zhang
- Department of Urology, Shantou Central Hospital, Shantou, PR China.
| |
Collapse
|
2
|
Semeniuk-Wojtaś A, Poddębniak-Strama K, Modzelewska M, Baryła M, Dziąg-Dudek E, Syryło T, Górnicka B, Jakieła A, Stec R. Tumour microenvironment as a predictive factor for immunotherapy in non-muscle-invasive bladder cancer. Cancer Immunol Immunother 2023:10.1007/s00262-023-03376-9. [PMID: 36928373 DOI: 10.1007/s00262-023-03376-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 01/09/2023] [Indexed: 03/18/2023]
Abstract
Bladder cancer (BC) can be divided into two subgroups depending on invasion of the muscular layer: non-muscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC). Its aggressiveness is associated, inter alia, with genetic aberrations like losses of 1p, 6q, 9p, 9q and 13q; gain of 5p; or alterations in the p53 and p16 pathways. Moreover, there are reported metabolic disturbances connected with poor diagnosis-for example, enhanced aerobic glycolysis, gluconeogenesis or haem catabolism.Currently, the primary way of treatment method is transurethral resection of the bladder tumour (TURBT) with adjuvant Bacillus Calmette-Guérin (BCG) therapy for NMIBC or radical cystectomy for MIBC combined with chemotherapy or immunotherapy. However, intravesical BCG immunotherapy and immune checkpoint inhibitors are not efficient in every case, so appropriate biomarkers are needed in order to select the proper treatment options. It seems that the success of immunotherapy depends mainly on the tumour microenvironment (TME), which reflects the molecular disturbances in the tumour. TME consists of specific conditions like hypoxia or local acidosis and different populations of immune cells including tumour-infiltrating lymphocytes, natural killer cells, neutrophils and B lymphocytes, which are responsible for shaping the response against tumour neoantigens and crucial pathways like the PD-L1/PD-1 axis.In this review, we summarise holistically the impact of the immune system, genetic alterations and metabolic changes that are key factors in immunotherapy success. These findings should enable better understanding of the TME complexity in case of NMIBC and causes of failures of current therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Tomasz Syryło
- Department of General, Active and Oncological Urology, Military Institute of Medicine, Warsaw, Poland
| | - Barbara Górnicka
- Pathomorphology Department, Medical University of Warsaw, Warsaw, Poland
| | - Anna Jakieła
- Oncology Department, 4 Military Clinical Hospital with a Polyclinic, Wroclaw, Poland
| | - Rafał Stec
- Oncology Department, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
3
|
Developing New Treatment Options for Castration-Resistant Prostate Cancer and Recurrent Disease. Biomedicines 2022; 10:biomedicines10081872. [PMID: 36009418 PMCID: PMC9405166 DOI: 10.3390/biomedicines10081872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer (PCa) is a major diagnosed cancer among men globally, and about 20% of patients develop metastatic prostate cancer (mPCa) in the initial diagnosis. PCa is a typical androgen-dependent disease; thus, hormonal therapy is commonly used as a standard care for mPCa by inhibiting androgen receptor (AR) activities, or androgen metabolism. Inevitably, almost all PCa will acquire resistance and become castration-resistant PCa (CRPC) that is associated with AR gene mutations or amplification, the presence of AR variants, loss of AR expression toward neuroendocrine phenotype, or other hormonal receptors. Treating CRPC poses a great challenge to clinicians. Research efforts in the last decade have come up with several new anti-androgen agents to prolong overall survival of CRPC patients. In addition, many potential targeting agents have been at the stage of being able to translate many preclinical discoveries into clinical practices. At this juncture, it is important to highlight the emerging strategies including small-molecule inhibitors to AR variants, DNA repair enzymes, cell survival pathway, neuroendocrine differentiation pathway, radiotherapy, CRPC-specific theranostics and immune therapy that are underway or have recently been completed.
Collapse
|
4
|
Rallis KS, Makrakis D, Ziogas IA, Tsoulfas G. Immunotherapy for advanced hepatocellular carcinoma: From clinical trials to real-world data and future advances. World J Clin Oncol 2022; 13:448-472. [PMID: 35949435 PMCID: PMC9244967 DOI: 10.5306/wjco.v13.i6.448] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/27/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-associated mortality worldwide. HCC is an inflammation-associated immunogenic cancer that frequently arises in chronically inflamed livers. Advanced HCC is managed with systemic therapies; the tyrosine kinase inhibitor (TKI) sorafenib has been used in 1st-line setting since 2007. Immunotherapies have emerged as promising treatments across solid tumors including HCC for which immune checkpoint inhibitors (ICIs) are licensed in 1st- and 2nd-line treatment setting. The treatment field of advanced HCC is continuously evolving. Several clinical trials are investigating novel ICI candidates as well as new ICI regimens in combination with other therapeutic modalities including systemic agents, such as other ICIs, TKIs, and anti-angiogenics. Novel immunotherapies including adoptive cell transfer, vaccine-based approaches, and virotherapy are also being brought to the fore. Yet, despite advances, several challenges persist. Lack of real-world data on the use of immunotherapy for advanced HCC in patients outside of clinical trials constitutes a main limitation hindering the breadth of application and generalizability of data to this larger and more diverse patient cohort. Consequently, issues encountered in real-world practice include patient ineligibly for immunotherapy because of contraindications, comorbidities, or poor performance status; lack of response, efficacy, and safety data; and cost-effectiveness. Further real-world data from high-quality large prospective cohort studies of immunotherapy in patients with advanced HCC is mandated to aid evidence-based clinical decision-making. This review provides a critical and comprehensive overview of clinical trials and real-world data of immunotherapy for HCC, with a focus on ICIs, as well as novel immunotherapy strategies underway.
Collapse
Affiliation(s)
- Kathrine S Rallis
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, United Kingdom
- Surgery Working Group, Society of Junior Doctors, Athens 15123, Greece
| | - Dimitrios Makrakis
- Surgery Working Group, Society of Junior Doctors, Athens 15123, Greece
- Division of Oncology, University of Washington School of Medicine, Seattle, WA 98195, United States
| | - Ioannis A Ziogas
- Surgery Working Group, Society of Junior Doctors, Athens 15123, Greece
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Georgios Tsoulfas
- Department of Transplantation Surgery, Aristotle University School of Medicine, Thessaloniki 54622, Greece
| |
Collapse
|
5
|
Nivolumab plus docetaxel in patients with chemotherapy-naïve metastatic castration-resistant prostate cancer: results from the phase II CheckMate 9KD trial. Eur J Cancer 2022; 160:61-71. [PMID: 34802864 DOI: 10.1016/j.ejca.2021.09.043] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND Docetaxel has immunostimulatory effects that may promote an immunoresponsive prostate tumour microenvironment, providing a rationale for combination with nivolumab (programmed death-1 inhibitor) for metastatic castration-resistant prostate cancer (mCRPC). METHODS In the non-randomised, multicohort, global phase II CheckMate 9KD trial, 84 patients with chemotherapy-naive mCRPC, ongoing androgen deprivation therapy and ≤2 prior novel hormonal therapies (NHTs) received nivolumab 360 mg and docetaxel 75 mg/m2 every 3 weeks with prednisone 5 mg twice daily (≤10 cycles) and then nivolumab 480 mg every 4 weeks (≤2 years). The co-primary end-points were objective response rate (ORR) and prostate-specific antigen response rate (PSA50-RR; ≥50% decrease from baseline). RESULTS The confirmed ORR (95% confidence interval [CI]) was 40.0% (25.7-55.7), and the confirmed PSA50-RR (95% CI) was 46.9% (35.7-58.3). The median (95% CI) radiographic progression-free survival (rPFS) and overall survival (OS) were 9.0 (8.0-11.6) and 18.2 (14.6-20.7) months, respectively. In subpopulations with versus without prior NHT, the ORR was 38.7% versus 42.9%, the PSA50-RR was 39.6% versus 60.7%, the median rPFS was 8.5 versus 12.0 months and the median OS was 16.2 months versus not reached. Homologous recombination deficiency status or tumour mutational burden did not appear to impact efficacy. The most common any-grade and grade 3-4 treatment-related adverse events were fatigue (39.3%) and neutropenia (16.7%), respectively. Three treatment-related deaths occurred (1 pneumonitis related to nivolumab; 2 pneumonias related to docetaxel). CONCLUSIONS Nivolumab plus docetaxel has clinical activity in patients with chemotherapy-naïve mCRPC. Safety was consistent with the individual components. These results support further investigation in the ongoing phase III CheckMate 7DX trial. CLINICALTRIALS. GOV REGISTRATION NCT03338790.
Collapse
|
6
|
Naseer F, Ahmad T, Kousar K, Anjum S. Advanced Therapeutic Options for Treatment of Metastatic Castration Resistant Prostatic Adenocarcinoma. Front Pharmacol 2021; 12:728054. [PMID: 34899292 PMCID: PMC8660108 DOI: 10.3389/fphar.2021.728054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/21/2021] [Indexed: 12/04/2022] Open
Abstract
The initial stage of prostatic adenocarcinoma (PaC) has been treated with surgery and radiation therapy, but the advanced stages need systemic novel treatment. Since 2010, several advanced therapeutic innovations have been introduced in various randomized clinical trials to improve survival and reduce morbidity and mortality. Several of these therapeutics have shown substantial survival assistance globally, even in the advanced stages of metastatic castration-resistant prostatic adenocarcinoma (mCRPC). This article describes advanced PaC therapy regimens including chemotherapeutic options, hormonal therapies (abiraterone, enzalutamide), immunotherapeutic agents, and bone-modifying agents. We discussed various pros and cons of gene therapy approaches including Crispr/Cas9 mediation, oncolytic viruses, suicidal genes, and micro-RNA based antitumor therapy. The mCRPC microenvironment is characterized by elevated prostate-specific antigen (PSA) levels, which ultimately trigger the androgen receptor (AR) and its dependent signaling pathways. The advanced therapeutics target these receptors and inhibit the steroidogenic enzymes that play an important role in increasing testosterone (T) and dihydrotestosterone (DHT) levels in the body. These advanced therapeutic novelties also target AR-independent oncogenic signaling pathways by focusing on DNA damage repair (DDR) pathways and their mechanisms. Some of these options appear to be very attractive strategies for acute and chronic stages of PaC and mCRPC treatment by overcoming the mechanisms of resistance.
Collapse
Affiliation(s)
- Faiza Naseer
- Industrial Biotechnology (IBT), Atta-ur-Rehman School of Applied Biosciences (ASAB), National University of Science and Technology, Islamabad, Pakistan.,Basic Medical Sciences, Shifa Tameer e Millat University (STMU), Islamabad, Pakistan
| | - Tahir Ahmad
- Industrial Biotechnology (IBT), Atta-ur-Rehman School of Applied Biosciences (ASAB), National University of Science and Technology, Islamabad, Pakistan
| | - Kousain Kousar
- Industrial Biotechnology (IBT), Atta-ur-Rehman School of Applied Biosciences (ASAB), National University of Science and Technology, Islamabad, Pakistan
| | - Sadia Anjum
- Department of Biology, University of Hail, Ha'il, Saudi Arabia
| |
Collapse
|
7
|
Ravindranathan D, Alhalabi O, Rafei H, Shah AY, Bilen MA. Landscape of Immunotherapy in Genitourinary Malignancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1342:143-192. [PMID: 34972965 PMCID: PMC11235092 DOI: 10.1007/978-3-030-79308-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The past decade has witnessed a revolution in the development of immune checkpoint inhibitors for the treatment of multiple tumor types, including genitourinary cancers. Immune checkpoint inhibitors have notably improved the treatment outcomes of patients with metastatic renal cell carcinoma and metastatic urothelial carcinoma. In prostate cancer, the role of immunotherapy with checkpoint inhibitors is not yet established except for microsatellite instability high (MSI-H) tumors. Other immunotherapeutic approaches that have been explored in these malignancies include cytokines, vaccines, and cellular therapy. Ongoing studies are exploring the use of immunotherapy combinations as well as combination with chemotherapy and targeted therapy in these types of tumors. The use of immunotherapy beyond the metastatic setting is an active area of research. Moreover, there is great interest in biomarker development to predict response to immunotherapy and risk of toxicity. This book chapter is a comprehensive review of immunotherapeutic approaches, both approved and investigational, for the treatment of renal cell carcinoma, urothelial carcinoma, and prostate cancer.
Collapse
Affiliation(s)
- Deepak Ravindranathan
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Omar Alhalabi
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hind Rafei
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amishi Yogesh Shah
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
8
|
Fizazi K, Drake CG, Beer TM, Kwon ED, Scher HI, Gerritsen WR, Bossi A, den Eertwegh AJMV, Krainer M, Houede N, Santos R, Mahammedi H, Ng S, Danielli R, Franke FA, Sundar S, Agarwal N, Bergman AM, Ciuleanu TE, Korbenfeld E, Sengeløv L, Hansen S, McHenry MB, Chen A, Logothetis C. Final Analysis of the Ipilimumab Versus Placebo Following Radiotherapy Phase III Trial in Postdocetaxel Metastatic Castration-resistant Prostate Cancer Identifies an Excess of Long-term Survivors. Eur Urol 2020; 78:822-830. [PMID: 32811715 PMCID: PMC8428575 DOI: 10.1016/j.eururo.2020.07.032] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/27/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND The phase 3 trial CA184-043 evaluated radiotherapy to bone metastases followed by Ipilimumab or placebo in men with metastatic castrate-resistant prostate cancer (mCRPC) who had received docetaxel previously. In a prior analysis, the trial's primary endpoint (overall survival [OS]) was not improved significantly. OBJECTIVE To report the final analysis of OS. DESIGN, SETTING, AND PARTICIPANTS A total of 799 patients were randomized to receive a single dose of radiotherapy to one or more bone metastases followed by either Ipilimumab (n = 399) or placebo (n = 400). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS OS was analyzed in the intention-to-treat population. Prespecified and exploratory subset analyses based on Kaplan-Meier/Cox methodology were performed. RESULTS AND LIMITATIONS During an additional follow-up of approximately 2.4 yr since the primary analysis, 721/799 patients have died. Survival analysis showed crossing of the curves at 7-8 mo, followed by persistent separation of the curves beyond that point, favoring the ipilimumab arm. Given the lack of proportional hazards, a piecewise hazard model showed that the hazard ratio (HR) changed over time: the HR was 1.49 (95% confidence interval 1.12, 1.99) for 0-5 mo, 0.66 (0.51, 0.86) for 5-12 mo, and 0.66 (0.52, 0.84) beyond 12 mo. OS rates were higher in the ipilimumab versus placebo arms at 2 yr (25.2% vs 16.6%), 3 yr (15.3% vs 7.9%), 4 yr (10.1% vs 3.3%), and 5 yr (7.9% vs. 2.7%). Disease progression was the most frequent cause of death in both arms. In seven patients (1.8%) in the ipilimumab arm and one (0.3%) in the placebo arm, the primary cause of death was reported as study drug toxicity. No long-term safety signals were identified. CONCLUSIONS In this preplanned long-term analysis, OS favored ipilimumab plus radiotherapy versus placebo plus radiotherapy for patients with postdocetaxel mCRPC. OS rates at 3, 4, and 5 yr were approximately two to three times higher in the ipilimumab arm. PATIENT SUMMARY After longer follow-up, survival favored the group of men who received ipilimumab, with overall survival rates being two to three times higher at 3 yr and beyond.
Collapse
Affiliation(s)
- Karim Fizazi
- Gustave Roussy, University of Paris Sud, Villejuif, France.
| | - Charles G Drake
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Tomasz M Beer
- Oregon Health & Science University Knight Cancer Institute, Portland, OR, USA
| | | | - Howard I Scher
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Weill-Cornell Medical College, New York, NY, USA
| | | | | | | | | | - Nadine Houede
- CHU of Nimes, Nimes, France; Montpellier University, Montpellier, France
| | | | | | - Siobhan Ng
- St John Of God Hospital, Subiaco, Australia
| | | | | | | | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - André M Bergman
- The Netherlands Cancer Institute and Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | | | - Lisa Sengeløv
- Herlev Hospital, Region Hovedstadens Apotek, Herlev, Denmark
| | | | | | - Allen Chen
- Bristol-Myers Squibb Company, Wallingford, CT, USA
| | | |
Collapse
|
9
|
Ma X, Zou L, Chen Z, Li X, Wei L, Wu X. Demethylation of miR-195 suppresses prostate cancer cell proliferation, migration and invasion. FEBS Open Bio 2020; 10:525-534. [PMID: 31977154 PMCID: PMC7137791 DOI: 10.1002/2211-5463.12799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer (PCa) is the most prevalent cancer among men and the second leading cause of tumor-associated deaths worldwide, with increasing incidence rates over the last 10 years. Recently, miR-195 was reported to be hypermethylated at its promoter CpG island and down-regulated in hepatocellular carcinoma. However, the function of miR-195 and the underlying mechanisms in PCa remain unknown. Here, we report that a significant down-regulation of microRNA-195 (miR-195) in PCa tissues and cell lines was associated with promoter methylation status. Overexpression of miR-195 significantly suppressed cell proliferation, migration, invasion and epithelial-mesenchymal transition (increased E-cadherin and decreased N-cadherin) in PCa cells. We further demonstrated that transfection with a miR-195 inhibitor reversed the inhibitory effect of the DNA methyltransferase inhibitor 5-azacytidine on the proliferation, migration and invasion ability of PCa cells. In summary, our findings suggest that miR-195 may function as a crucial tumor suppressor in PCa.
Collapse
Affiliation(s)
- Xiaokun Ma
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liyuan Zou
- Department of Prevention and Health Care, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhanhong Chen
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xing Li
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li Wei
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiangyuan Wu
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
10
|
Jiang Z, Zhang Y, Chen X, Wu P, Chen D. Inactivation of the Wnt/β-catenin signaling pathway underlies inhibitory role of microRNA-129-5p in epithelial-mesenchymal transition and angiogenesis of prostate cancer by targeting ZIC2. Cancer Cell Int 2019; 19:271. [PMID: 31649488 PMCID: PMC6805653 DOI: 10.1186/s12935-019-0977-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022] Open
Abstract
Background Prostate cancer (PCa) is a common disease that often occurs among older men and a frequent cause of malignancy associated death in this group. microRNA (miR)-129-5p has been identified as an essential regulator with a significant role in the prognosis of PC. Therefore, this study aimed to investigate roles of miR-129-5p in PCa. Methods Microarray analysis was conducted to identify PCa-related genes. The expression of miR-129-5p and ZIC2 in PCa tissues was investigated. To understand the role of miR-129-5p and ZIC2 in PCa, DU145 cells were transfected with mimic or inhibitor of miR-129-5p, or si-ZIC2 and the expression of Wnt, β-catenin, E-cadherin, vimentin, N-cadherin, vascular endothelial growth factor (VEGF), and CD31, as well as the extent of β-catenin phosphorylation was determined. In addition, cell proliferation, migration, invasion, angiogenesis, apoptosis and tumorigenesis were detected. Results miR-129-5p was poorly expressed and ZIC2 was highly expressed in PCa tissues. Down-regulation of ZIC2 or overexpression of miR-129-5p reduced the expression of ZIC2, Wnt, β-catenin, N-cadherin, vimentin, and β-catenin phosphorylation but increased the expression of E-cadherin. Importantly, miR-129-5p overexpression significantly reduced cell migration, invasion, angiogenesis and tumorigenesis while increasing cell apoptosis. Conclusions The findings of the present study indicated that overexpression of miR-129-5p or silencing of ZIC2 could inhibit epithelial–mesenchymal transition (EMT) and angiogenesis in PCa through blockage of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Zhenming Jiang
- 1Department of Urology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China
| | - Yuxi Zhang
- 1Department of Urology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China.,Department of Urology, People's Hospital of Datong Hui and Tu Autonomous County, No. 1, Wenhua Road, Qiaotou Town, Datong Hui and Tu Autonomous County, Xining, 810100 Qinghai People's Republic of China
| | - Xi Chen
- 3Department of Pharmacy, The First Hospital of China Medical University, Shenyang, 110001 People's Republic of China
| | - Pingeng Wu
- 1Department of Urology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China
| | - Dong Chen
- 4Central Lab, The First Hospital of China Medical University, Shenyang, 110001 People's Republic of China
| |
Collapse
|
11
|
Cao J, Kong FH, Liu X, Wang XB. Immunotherapy with dendritic cells and cytokine-induced killer cells for hepatocellular carcinoma: A meta-analysis. World J Gastroenterol 2019; 25:3649-3663. [PMID: 31367163 PMCID: PMC6658393 DOI: 10.3748/wjg.v25.i27.3649] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/18/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has been revealed as the second most common cause of cancer-related deaths worldwide. The introduction of cell-based immunotherapy, including dendritic cells (DCs) and cytokine-induced killer cells (CIKs), has brought HCC patients an effective benefit. However, the efficacy and necessity of cellular immunotherapy after different interventional therapy remains to be further explored. AIM To investigate the efficacy of cellular immunotherapy, involving DCs and CIKs, combined with different conventional treatments of HCC. METHODS We performed a literature search on PubMed and Web of Science up to February 15, 2019. Long-term efficacy (overall survival and recurrence) and short-term adverse effects were investigated to assess the effectiveness of immunotherapy with DCs and/or CIKs. Review Manager 5.3 was used to perform the analysis. RESULTS A total of 22 studies involving 3756 patients selected by eligibility inclusion criteria were forwarded for meta-analysis. Combined with the conventional clinical treatment, immunotherapy with DCs and/or CIKs was demonstrated to significantly improve overall survival at 6 mo [risk ratio (RR) = 1.07; 95% confidence interval (CI): 1.01-1.13, P = 0.02], 1 year (RR = 1.12; 95%CI: 1.07-1.17, P < 0.00001), 3 years (RR = 1.23; 95%CI: 1.15-1.31, P < 0.00001) and 5 years (RR = 1.26; 95%CI: 1.15-1.37, P < 0.00001). Recurrence rate was significantly reduced by cellular immunotherapy at 6 mo (RR = 0.50; 95%CI: 0.36-0.69, P < 0.0001) and 1 year (RR = 0.82; 95%CI: 0.75-0.89, P < 0.00001). Adverse effect assessment addressed that immunotherapy with DCs and/or CIKs was accepted as a safe, feasible treatment. CONCLUSION Combination immunotherapy with DCs, CIKs and DC/CIK with various routine treatments for HCC was evidently suggested to improve patients' prognosis by increasing overall survival and reducing cancer recurrence.
Collapse
Affiliation(s)
- Jing Cao
- Department of Surgery, Technical University of Munich, Munich 80333, Germany
| | - Fan-Hua Kong
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xi Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiao-Bo Wang
- Department of Surgery, Technical University of Munich, Munich 80333, Germany
| |
Collapse
|
12
|
Shah R, Botteman M, Solem CT, Luo L, Doan J, Cella D, Motzer RJ. A Quality-adjusted Time Without Symptoms or Toxicity (Q-TWiST) Analysis of Nivolumab Versus Everolimus in Advanced Renal Cell Carcinoma (aRCC). Clin Genitourin Cancer 2019; 17:356-365.e1. [PMID: 31272883 PMCID: PMC8262523 DOI: 10.1016/j.clgc.2019.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/10/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
This study assessed the net health benefits of treatment with nivolumab versus everolimus among patients with advanced renal cell carcinoma by assessing the quality (ie, patient preferences) and quantity of survival (ie, time spent with significant toxicities, in progression, or before progression and without significant toxicities). Nivolumab resulted in a 3.3-month quality-adjusted survival gain versus everolimus that was statistically significant and clearly clinically meaningful.
Collapse
Affiliation(s)
| | | | | | | | | | - David Cella
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | |
Collapse
|
13
|
Zhang J, Wang L, You X, Xian T, Wu J, Pang J. Nanoparticle Therapy for Prostate Cancer: Overview and Perspectives. Curr Top Med Chem 2019; 19:57-73. [PMID: 30686255 DOI: 10.2174/1568026619666190125145836] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/27/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022]
Abstract
Traditional prostate cancer therapy and especially chemotherapy has faced many challenges. Low accumulation levels, rapid clearance or drug resistance at the tumor site have been central to why the effect of chemotherapy drugs has declined. Applications of nanotechnology to biomedicine have enabled the development of nanoparticle therapeutic carriers suited for the delivery of chemotherapeutics in cancer therapy. This review describes the current nature of nanoparticle therapeutic carriers for prostate cancer. It describes typical nanocarriers commonly used for the delivery of chemotherapy or for imaging examination. Targeting strategies and related influencing factors are investigated to find ways of enhancing treatment effects of nanoparticles. The overall purpose of this review is to further understanding and to offer recommendations on the design and development of therapeutic nanoparticles for prostate cancer.
Collapse
Affiliation(s)
- Junfu Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.,Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Liying Wang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Xinru You
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Tuzeng Xian
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510006, China.,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, 518057, China
| | - Jun Pang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
14
|
Zheng XM, Zhang P, Liu MH, Chen P, Zhang WB. MicroRNA-30e inhibits adhesion, migration, invasion and cell cycle progression of prostate cancer cells via inhibition of the activation of the MAPK signaling pathway by downregulating CHRM3. Int J Oncol 2019; 54:443-454. [PMID: 30483762 PMCID: PMC6317654 DOI: 10.3892/ijo.2018.4647] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PCa) testing is currently based on measurement of serum prostate‑specific antigen levels and digital rectal examination, which are limited by a low predictive value and the adverse effects associated with overdiagnosis and overtreatment. Recent studies have reported that the abnormal expression of microRNAs (miRNAs) is associated with the mechanism underlying the development of PCa. Thus, the aim of the present study was to investigate the effects of miR‑30e and its target gene, M3 muscarinic acetylcholine receptor (CHRM3), on the adhesion, migration, invasion and cell cycle distribution of PCa cells via the mitogen‑activated protein kinase (MAPK) signaling pathway. The differentially expressed genes were screened in the Gene Expression Omnibus database from a gene expression microarray (GSE55945) of PCa. PCa tissues and adjacent tissues were collected from patients with PCa. The PC‑3 and DU145 human PCa cell lines were treated with activator, inhibitor and siRNAs. The effects of miR‑30e on cell adhesion, migration, invasion and cell cycle distribution with the involvement of CHRM3 and the MAPK signaling pathway were investigated. The bioinformatics results demonstrated that the CHRM3 gene and the MAKP signaling pathway were involved in the progression of PCa, and has‑miR‑30e was selected for further study. The levels of miR‑30e were significantly downregulated, while the levels of CHRM3 were obviously upregulated in PCa. CHRM3 was verified as a target gene of miR‑30e. Upregulation of miR‑30e and downregulation of CHRM3 decreased the levels of p‑P38, p‑extracellular signal‑regulated kinase, p‑c‑Jun N‑terminal kinase, p‑c‑fos and p‑c‑JUN, cell adhesion, migration and invasion ability, and the number of cells in the S phase, while they increased the number of cells in the G0 and G1 phases. The findings of the present study suggest that miR‑30e inhibited the adhesion, migration, invasion and cell cycle entry of PCa cells by suppressing the activation of the MAPK signaling pathway and inhibiting CHRM3 expression. Thus, miR‑30e may serve as a candidate target for the treatment of PCa.
Collapse
Affiliation(s)
- Xin-Min Zheng
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| | - Peng Zhang
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| | - Man-Hua Liu
- The Second Department of Surgery, Jingan People’s Hospital, Jingan, Jiangxi 330600, P.R. China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| | - Wei-Bing Zhang
- Department of Urology, Zhongnan Hospital of WuHan University, Wuhan, Hubei 430071
| |
Collapse
|
15
|
Hood SP, Foulds GA, Imrie H, Reeder S, McArdle SEB, Khan M, Pockley AG. Phenotype and Function of Activated Natural Killer Cells From Patients With Prostate Cancer: Patient-Dependent Responses to Priming and IL-2 Activation. Front Immunol 2019; 9:3169. [PMID: 30761160 PMCID: PMC6362408 DOI: 10.3389/fimmu.2018.03169] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/24/2018] [Indexed: 12/27/2022] Open
Abstract
Background: Although immunotherapy has emerged as the “next generation” of cancer treatments, it has not yet been shown to be successful in the treatment of patients with prostate cancer, for whom therapeutic options remain limited to radiotherapy and androgen (hormone) deprivation therapy. Previous studies have shown that priming natural killer (NK) cells isolated from healthy individuals via co-incubation with CTV-1 cells derived from an acute lymphoblastic leukemia (ALL) enhances their cytotoxicity against human DU145 (metastatic) prostate cancer cells, but it remains unknown to what extent NK cells from patients with prostate cancer can be triggered to kill. Herein, we explore the phenotype of peripheral blood NK cells in patients with prostate cancer and compare the capacity of CTV-1 cell-mediated priming and IL-2 stimulation to trigger NK cell-mediated killing of the human PC3 (metastatic) prostate cancer cell line. Methods: The phenotype of resting, primed (co-incubation with CTV-1 cells for 17 h) and IL-2 activated (100 IU/ml IL-2 for 17 h) NK cells isolated from frozen-thawed peripheral blood mononuclear cell (PBMC) preparations from patients with benign disease (n = 6) and prostate cancer (n = 18) and their cytotoxicity against PC3 and K562 cells was determined by flow cytometry. Relationship(s) between NK cell phenotypic features and cytotoxic potential were interrogated using Spearman Rank correlation matrices. Results and Conclusions: NK cell priming and IL-2 activation of patient-derived NK cells resulted in similar levels of cytotoxicity, but distinct NK cell phenotypes. Importantly, the capacity of priming and IL-2 stimulation to trigger cytotoxicity was patient-dependent and mutually exclusive, in that NK cells from ~50% of patients preferentially responded to priming whereas NK cells from the remaining patients preferentially responded to cytokine stimulation. In addition to providing more insight into the biology of primed and cytokine-stimulated NK cells, this study supports the use of autologous NK cell-based immunotherapies for the treatment of prostate cancer. However, our findings also indicate that patients will need to be stratified according to their potential responsiveness to individual therapeutic approaches.
Collapse
Affiliation(s)
- Simon P Hood
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Gemma A Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Heather Imrie
- School of Animal Rural and Environmental Sciences, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephen Reeder
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stéphanie E B McArdle
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Masood Khan
- Department of Urology, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Alan Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
16
|
[Immunotherapy for the treatment of prostate cancer-a comeback?]. Urologe A 2018; 57:1342-1345. [PMID: 30341689 DOI: 10.1007/s00120-018-0790-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Prostate cancer (PCA) seems to be more of an immunologic desert than other tumor entities. It is striking that only rarely does prostate cancer show abundant immune cells and a proimmunogenic microenvironment. OBJECTIVES Is immunotherapy in PCA effective and which patients can benefit. MATERIALS AND METHODS A review of the literature and recent congress data are presented. RESULTS Preliminary results with sipuleucel-T for PCA cancer were very promising showing a significant overall survival benefit in randomised phase III studies and the US Federal Drug Administration (FDA) approval for this individualised vaccine. Contrary to other tumor entities this was not the immediate breakthrough to a new therapeutic era of immunotherapy but remained an isolated case and restricted to the USA. More recently, several trials evaluated immunotherapeutic agents but missed their preliminary endpoints. Interestingly, individual patients did benefit and showed long-term remission. CONCLUSIONS Genome sequencing and new biomarkers are also paving a novel pathway towards individualised immunotherapy for PCA. On-going research and clinical trials are exploring the question of which patients will benefit.
Collapse
|
17
|
Baciarello G, Gizzi M, Fizazi K. Advancing therapies in metastatic castration-resistant prostate cancer. Expert Opin Pharmacother 2018; 19:1797-1804. [PMID: 30311804 DOI: 10.1080/14656566.2018.1527312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Prostate cancer is the second most common cause of cancer worldwide and is the most frequently detected cancer in the European Union in men over 50 years of age. Androgen deprivation therapy remains the cornerstone of treatment for recurrent or metastatic disease. Unfortunately, nearly all patients will develop resistance to androgen blockade leading to castration-resistant prostate cancer (CRPC). Over the last 10 years, new treatments have dramatically improved overall survival of men with mCRPC. Current therapies are based on AR-axis inhibitors and taxane-based chemotherapies, as well as radiopharmaceuticals and Sipuleucel T. AREAS COVERED The authors provide a review of the current field of systemic therapy in metastatic CRPC. This is followed by an in-depth analysis of recent developments in treatment, and the biological rationale behind these therapies. EXPERT OPINION Since several trials with docetaxel or novel hormonal agents showed improvement in overall survival in metastatic castration-sensitive prostate cancer, as well as in non-metastatic castration-resistant patients, it is expected that a growing subgroup of patients will be exposed earlier to chemotherapy and to AR targeted agents. It becomes then fundamental to find novel strategies to overcome drug resistance and further improve survival.
Collapse
Affiliation(s)
- Giulia Baciarello
- a Department of Medical Oncology , Institut Gustave Roussy, University of Paris Sud , Villejuif , France
| | - Marco Gizzi
- b Service d'Oncologie , Grand Hôpital de Charleroi & Cliniques universitaires Saint Luc , Bruxelles , Belgium
| | - Karim Fizazi
- a Department of Medical Oncology , Institut Gustave Roussy, University of Paris Sud , Villejuif , France
| |
Collapse
|
18
|
Alaia C, Boccellino M, Zappavigna S, Amler E, Quagliuolo L, Rossetti S, Facchini G, Caraglia M. Ipilimumab for the treatment of metastatic prostate cancer. Expert Opin Biol Ther 2017; 18:205-213. [PMID: 29271259 DOI: 10.1080/14712598.2018.1420777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Immunotherapy with checkpoint inhibitors is beginning to be recognized as a valid weapon for the treatment of metastatic prostate cancer (PCa) when chemotherapy fails. Ipilimumab (ipi) is a fully humanized monoclonal antibody that blocks the activity of CTLA4. It also has a molecular weight of 148 kDa and is water-soluble at physiological pH. Ipi was first approved by the FDA for the treatment of malignant melanoma and is currently being studied in metastatic castration-resistant prostate cancer, with promising early results. Areas covered: The aim of this review is to collate the most significant preclinical and clinical studies available that look at ipi to propose new strategies for the future. Expert opinion: Additional studies are required to reduce toxicity and increase the activity of ipi in PCa. A possible strategy is to combine ipi with standard anti-cancer therapeutics such as vaccines, PDL1 inhibitors, antiandrogen drugs, and chemotherapy agents. Several initial results have suggested that combination strategies are useful to increase the activity in mCRPC, even if the toxicity of the treatment can increase. The activity of combined treatments is still not predictable, but considering the ongoing studies, we believe that they have good potential that will lead to the discovery of an optimal therapeutic strategy.
Collapse
Affiliation(s)
- Concetta Alaia
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Mariarosaria Boccellino
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Silvia Zappavigna
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Evzen Amler
- b Department of Biophysics, 2nd Faculty of Medicine , Charles University Prague , Prague , Czech Republic.,c Laboratory of Tissue Engineering, Institute of Experimental Medicine , Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Lucio Quagliuolo
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Sabrina Rossetti
- d Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale" , Napoli , Italy.,e Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo, Uro-Gynaechological Department of the National Institute of Tumours "G. Pascale", Regione Campania , Naples , Italy
| | - Gaetano Facchini
- d Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale" , Napoli , Italy
| | - Michele Caraglia
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| |
Collapse
|
19
|
Fizazi K, Ulys A, Sengeløv L, Moe M, Ladoire S, Thiery-Vuillemin A, Flechon A, Guida A, Bellmunt J, Climent MA, Chowdhury S, Dumez H, Matouskova M, Penel N, Liutkauskiene S, Stachurski L, Sternberg CN, Baton F, Germann N, Daugaard G. A randomized, double-blind, placebo-controlled phase II study of maintenance therapy with tasquinimod in patients with metastatic castration-resistant prostate cancer responsive to or stabilized during first-line docetaxel chemotherapy. Ann Oncol 2017; 28:2741-2746. [PMID: 29059273 PMCID: PMC6246397 DOI: 10.1093/annonc/mdx487] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND This phase II study was conducted to assess clinical efficacy of tasquinimod maintenance therapy in patients with metastatic castrate-resistant prostate cancer not progressing during first-line docetaxel-based therapy. PATIENTS AND METHODS Patients were randomly assigned (1 : 1) to receive tasquinimod (0.25-1.0 mg/day orally) or placebo. The primary end point was radiologic progression-free survival (rPFS); secondary efficacy end points included: overall survival (OS); PFS on next-line therapy (PFS 2) and symptomatic PFS, assessed using the Brief Pain Inventory (BPI) questionnaire and analgesic use. Quality of life was measured by the Functional Assessment of Cancer Therapy-Prostate (FACT-P) questionnaire and by the EuroQol-5 Dimension Quality of Life Instrument (EQ-5D). Adverse events were recorded. RESULTS A total of 219 patients were screened and 144 patients randomized. The median duration of treatment was 18.7 weeks (range 0.6-102.7 weeks) for the tasquinimod arm and 19.2 weeks (range 0.4-80.0 weeks) for the placebo arm. Median (90% CI) rPFS was 31.7 (24.3-53.7) and 22.7 (16.1-25.9) weeks in the tasquinimod and placebo arms, respectively [HR (90% CI) 0.6 (0.4-0.9), P = 0.0162]. The median OS was not reached because only 14 deaths occurred by the cut-off date. No statistically significant differences between treatment arms were noted for symptomatic PFS, PFS 2, BPI score, FACT-P score, or EQ-5D. The incidence of any treatment emergent adverse event (TEAE) was similar in the tasquinimod and placebo arms (97.2% versus 94.3%, respectively), whereas severe TEAEs (NCI-CTC Grade 3-5) incidence was higher in the tasquinimod group (50.7% versus 27.1%). CONCLUSIONS Randomized trials testing new drugs as maintenance can be successfully conducted after chemotherapy in castrate-resistant prostate cancer. Maintenance tasquinimod therapy significantly reduced the risk of rPFS by 40%. CLINICALTRIALS gov identifier NCT01732549.
Collapse
Affiliation(s)
- K Fizazi
- Department of Cancer Medicine, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| | - A Ulys
- National Cancer Institute, Vilnius, Lithuania
| | - L Sengeløv
- Department of Oncology, Herlev Hospital, Herlev
| | - M Moe
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | - S Ladoire
- Department of Medical Oncology, Centre Georges François Leclerc, Dijon
| | | | - A Flechon
- Medicine, Centre Léon Bérard, Lyon, France
| | - A Guida
- Department of Oncology & Hematology, Azienda University Hospital, Modena, Italy
| | - J Bellmunt
- Dana-Farber/Brigham and Women's Cancer Center, Boston, USA
| | - M A Climent
- Department of Medical Oncology, Instituto Valenciano de Oncología, Valencia, Spain
| | - S Chowdhury
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - H Dumez
- Department of General Medical Oncology, University Hospitals Leuven, Leuven; KU Leuven, Leuven, Belgium
| | | | - N Penel
- General Oncology Department, Centre Oscar Lambret, Lille, France
| | - S Liutkauskiene
- Lithuanian University of Health Sciences Hospital, Kaunas, Lithuania
| | | | - C N Sternberg
- Department of Medical Oncology, San Camillo and Forlanini Hospitals, Rome, Italy
| | - F Baton
- Ipsen Innovation, Les Ulis, France
| | | | - G Daugaard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
20
|
Koti M, Xu AS, Ren KYM, Visram K, Ren R, Berman DM, Siemens DR. Tertiary Lymphoid Structures Associate with Tumour Stage in Urothelial Bladder Cancer. Bladder Cancer 2017; 3:259-267. [PMID: 29152550 PMCID: PMC5676768 DOI: 10.3233/blc-170120] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Urothelial bladder cancer (UBC) is a highly prevalent disease in North America, however its optimal management remains elusive. The contribution of B cell associated responses is poorly understood in bladder cancer. Lymphoid neogenesis is a hallmark of an active immune response at tumor sites that sometimes leads to formation of tertiary lymphoid structures (TLS) that resemble germinal centers formed in secondary lymphoid organs. OBJECTIVE This study was conducted with an aim to investigate the presence and characteristics of TLS in UBC with a focus to compare and contrast the TLS formation in treatment naive low grade non-muscle invasive (NMIBC) and muscle invasive bladder cancers (MIBC). METHODS The study cohort consisted of transurethral bladder resection tumour (TURBT) specimens from 28 patients. Sections showing lymphoid aggregates in hematoxylin and eosin (H&E) stained TURBT specimens were further subjected to multi-color immunohistochemistry using immune cell markers specific to CD20+ B cells, CD3+ and CD8+ T cells, PNAd+ high endothelial venules, CD208+ mature dendritic cells, CD21+ follicular dendritic cells to confirm the hallmarks of classical germinal centers. RESULTS Our pilot study investigating the presence of TLS in bladder cancer patients is the first to demonstrate that well-formed TLS are more common in aggressive high grade MIBC tumors compared to low grade NIMBC. CONCLUSIONS These novel findings suggest B cell mediated anti-tumour humoral immune responses in bladder cancer progression.
Collapse
Affiliation(s)
- Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
- Department of Obstetrics and Gynecology, Queen’s University, Kingston, ON, Canada
- Cancer Biology and Genetics Division, Queen’s Cancer Research Institute, Queen’s University, Kingston, ON, Canada
| | - Amanda Shuo Xu
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Kevin Yi Mi Ren
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Kash Visram
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Runhan Ren
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - David M. Berman
- Cancer Biology and Genetics Division, Queen’s Cancer Research Institute, Queen’s University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - D. Robert Siemens
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
- Department of Urology Queen’s University, Kingston, ON, Canada
| |
Collapse
|
21
|
ALBERTI C. Prostate cancer immunotherapy, particularly in combination with androgen deprivation or radiation treatment. Customized pharmacogenomic approaches to overcome immunotherapy cancer resistance. G Chir 2017; 37:225-235. [PMID: 28098061 PMCID: PMC5256907 DOI: 10.11138/gchir/2016.37.5.225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Conventional therapeutic approaches for advanced prostate cancer - such as androgen deprivation, chemotherapy, radiation - come up often against lack of effectiveness because of possible arising of correlative cancer cell resistance and/or inadequate anti-tumor immune conditions. Whence the timeliness of resorting to immune-based treatment strategies including either therapeutic vaccination-based active immunotherapy or anti-tumor monoclonal antibody-mediated passive immunotherapy. Particularly attractive, as for research studies and clinical applications, results to be the cytotoxic T-lymphocyte check point blockade by the use of anti-CTLA-4 and PD-1 monoclonal antibodies, particularly when combined with androgen deprivation therapy or radiation. Unlike afore said immune check point inhibitors, both cell-based (by the use of prostate specific antigen carriers autologous dendritic cells or even whole cancer cells) and recombinant viral vector vaccines are able to induce immune-mediated focused killing of specific antigen-presenting prostate cancer cells. Such vaccines, either used alone or concurrently/sequentially combined with above-mentioned conventional therapies, led to generally reach, in the field of various clinical trials, reasonable results particularly as regards the patient's overall survival. Adoptive trasferred T-cells, as adoptive T-cell passive immunotherapy, and monoclonal antibodies against specific antigen-endowed prostate cancer cells can improve immune micro-environmental conditions. On the basis of a preliminary survey about various immunotherapy strategies, are here also outlined their effects when combined with androgen deprivation therapy or radiation. What's more, as regard the immune-based treatment effectiveness, it has to be pointed out that suitable personalized epigenetic/gene profile-achieved pharmacogenomic approaches to target identified gene aberrations, may lead to overcome - as well as for conventional therapies - possible prostate cancer resistance to immunotherapy.
Collapse
|
22
|
Abstract
Immunotherapy for castration-resistant prostate cancer has continued to be an area of active research over the last several years. The enthusiasm of this approach has been based on the assumption of better tolerability and that using the body's own immune system may be more effective than either hormonal or chemotherapy. Sipuleucel-T, a dendritic cell-based vaccine, is the only approved agent in this class for the management of castrate-resistant prostate cancer. Although sipuleucel-T increases overall survival without any significant changes in progression-free survival, other forms of immunotherapy such as PSA-TRICOM, ipilimumab, and chimeric antigen receptor T cell therapy are in advanced stages of clinical development. Immune biomarkers are being developed to assess response to these treatments and also to understand how the immune system responds to these respective therapies. Combinations of immunotherapy with androgen deprivation, radiation therapy, and chemotherapy have also been explored with varying results. This review discusses the mechanisms, key preclinical and clinical data, and perspectives for immunotherapeutic agents in the treatment scheme for castrate-resistant prostate cancer.
Collapse
|
23
|
Cabel L, Loir E, Gravis G, Lavaud P, Massard C, Albiges L, Baciarello G, Loriot Y, Fizazi K. Long-term complete remission with Ipilimumab in metastatic castrate-resistant prostate cancer: case report of two patients. J Immunother Cancer 2017; 5:31. [PMID: 28428880 PMCID: PMC5394619 DOI: 10.1186/s40425-017-0232-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Prostate cancer is one of the most common cancers in men and the fourth leading cause of cancer mortality worldwide. Although major progress has been achieved in the last years for patients with metastatic castrate-resistant prostate cancer (mCRPC), thanks to next-generation androgen receptor axis targeted drugs, taxanes, and bone-targeted agents, immunotherapy has not been widely approved and used for the treatment of prostate cancer. Two large studies with ipilimumab, an anti-CTLA-4 (cytotoxic T-lymphocyte antigen 4) antibody reported improved progression-free survival, but not statistically improved overall survival at the primary analysis (CA184 043 and CA184 095). CASE PRESENTATION Here, we report on two patients who received ipilimumab in these trials and are still in long-term complete remission with a follow-up of 64 and 52 months respectively after the initiation of ipilimumab. Immunohistochemical staining for hMLH1, hMSH2, hMSH6 and PMS2 was performed on archival prostate biopsy samples from one of the two patients; they exhibited normal protein expression. Interestingly for this patient, a high CD3+ and CD8+ T cell infiltration was observed on archival prostate biopsies as well as Treg FoxP3+ T cells. CONCLUSION Ipilimumab produces clinical activity in patients with CRPC, including very long responders with no detectable residual disease.
Collapse
Affiliation(s)
- Luc Cabel
- Institut Gustave Roussy, University of Paris-Sud, Department of Cancer Medicine, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| | - Elika Loir
- Institut Paoli-Calmettes, Department of Medical Oncology, 13009 Marseille, France
| | - Gwenaelle Gravis
- Institut Paoli-Calmettes, Department of Medical Oncology, 13009 Marseille, France
| | - Pernelle Lavaud
- Institut Gustave Roussy, University of Paris-Sud, Department of Cancer Medicine, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| | - Christophe Massard
- Institut Gustave Roussy, University of Paris-Sud, Department of Cancer Medicine, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| | - Laurence Albiges
- Institut Gustave Roussy, University of Paris-Sud, Department of Cancer Medicine, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| | - Giulia Baciarello
- Institut Gustave Roussy, University of Paris-Sud, Department of Cancer Medicine, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| | - Yohann Loriot
- Institut Gustave Roussy, University of Paris-Sud, Department of Cancer Medicine, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris-Sud, Department of Cancer Medicine, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| |
Collapse
|
24
|
McNeel DG, Bander NH, Beer TM, Drake CG, Fong L, Harrelson S, Kantoff PW, Madan RA, Oh WK, Peace DJ, Petrylak DP, Porterfield H, Sartor O, Shore ND, Slovin SF, Stein MN, Vieweg J, Gulley JL. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of prostate carcinoma. J Immunother Cancer 2016; 4:92. [PMID: 28031820 PMCID: PMC5170901 DOI: 10.1186/s40425-016-0198-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/23/2016] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed malignancy and second leading cause of cancer death among men in the United States. In recent years, several new agents, including cancer immunotherapies, have been approved or are currently being investigated in late-stage clinical trials for the management of advanced prostate cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) convened a multidisciplinary panel, including physicians, nurses, and patient advocates, to develop consensus recommendations for the clinical application of immunotherapy for prostate cancer patients. To do so, a systematic literature search was performed to identify high-impact papers from 2006 until 2014 and was further supplemented with literature provided by the panel. Results from the consensus panel voting and discussion as well as the literature review were used to rate supporting evidence and generate recommendations for the use of immunotherapy in prostate cancer patients. Sipuleucel-T, an autologous dendritic cell vaccine, is the first and currently only immunotherapeutic agent approved for the clinical management of metastatic castrate resistant prostate cancer (mCRPC). The consensus panel utilized this model to discuss immunotherapy in the treatment of prostate cancer, issues related to patient selection, monitoring of patients during and post treatment, and sequence/combination with other anti-cancer treatments. Potential immunotherapies emerging from late-stage clinical trials are also discussed. As immunotherapy evolves as a therapeutic option for the treatment of prostate cancer, these recommendations will be updated accordingly.
Collapse
Affiliation(s)
- Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, 7007 WIMR, 1111 Highland Avenue, Madison, WI 53705 USA
| | - Neil H Bander
- Weill Medical College of Cornell University, Laboratory of Urological Oncology E-300, 525 East 68th Street, New York, NY 10021 USA
| | - Tomasz M Beer
- Oregon Health and Science University Knight Cancer Institute, 3181 SW Sam Jackson Park Road, Portland, OR 97239 USA
| | - Charles G Drake
- Johns Hopkins University, 1650 Orleans Street Room 410, Baltimore, MD 21287 USA
| | - Lawrence Fong
- University of California, San Francisco, 513 Parnassus Ave, Room HSF 301, Box 1270, San Francisco, CA 94143 USA
| | - Stacey Harrelson
- Carolina Urologic Research Center, 823 82nd Parkway, Suite B, Myrtle Beach, SC 29572 USA
| | - Philip W Kantoff
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021 USA
| | - Ravi A Madan
- National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892 USA
| | - William K Oh
- Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1079, New York, NY 10029 USA
| | - David J Peace
- University of Illinois, 840 S Wood Street, Suite 820, Chicago, IL 60612 USA
| | | | - Hank Porterfield
- Alliance for Prostate Cancer Prevention, 17660 Tamiami Trail, Suite 106, Fort Myers, FL 33908 USA
| | - Oliver Sartor
- Tulane University School of Medicine, 1430 Tulane Ave, SL-42, New Orleans, LA 70112 USA
| | - Neal D Shore
- Carolina Urologic Research Center, 823 82nd Parkway, Suite B, Myrtle Beach, SC 29572 USA
| | - Susan F Slovin
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021 USA
| | - Mark N Stein
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903 USA
| | - Johannes Vieweg
- Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328 USA
| | - James L Gulley
- Genitourinary Malignancies Branch, 10 Center Drive, 13N240, Bethesda, MD 20892 USA
| |
Collapse
|
25
|
Yap TA, Smith AD, Ferraldeschi R, Al-Lazikani B, Workman P, de Bono JS. Drug discovery in advanced prostate cancer: translating biology into therapy. Nat Rev Drug Discov 2016; 15:699-718. [DOI: 10.1038/nrd.2016.120] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
26
|
Falasca L, Torino F, Marconi M, Costantini M, Pompeo V, Sentinelli S, De Salvo L, Patrizio M, Padula C, Gallucci M, Piacentini M, Malorni W. AMBRA1 and SQSTM1 expression pattern in prostate cancer. Apoptosis 2016; 20:1577-86. [PMID: 26423274 PMCID: PMC4602066 DOI: 10.1007/s10495-015-1176-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Prostate cancer is among the most commonly diagnosed male diseases and a leading cause of cancer mortality in men. There is emerging evidence that autophagy plays an important role in malignant cell survival and offers protection from the anti-cancer drugs in prostate cancer cells. AMBRA1 and the autophagic protein sequestosome-1 (SQSTM1; p62) expression were evaluated by immunohistochemistry and western blot on tissue samples from both benign and malignant prostatic lesions. The data reported in this pilot study demonstrated an increased expression of AMBRA1 and SQSTM1, which were also associated with an accumulation of LC3II in prostate cancer but not in benign lesion. In the present study we found that: (i) at variance with benign lesion, prostate cancer cells underwent SQSTM1 accumulation, i.e., clearly displayed a defective autophagic process but, also, (ii) prostate cancer accumulated AMBRA1 and (iii) this increase positively correlated with the Gleason score. These results underscore a possible implication of autophagy in prostate cancer phenotype and of AMBRA1 as possible cancer progression biomarker in this malignancy.
Collapse
Affiliation(s)
- Laura Falasca
- Laboratory of Cell Biology and Electron Microscopy, National Institute for Infectious Diseases I.R.C.C.S. 'L. Spallanzani', Rome, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Matteo Marconi
- Department of Drug Research and Medicine Evaluation, Istituto Superiore di Sanita', Rome, Italy
| | - Manuela Costantini
- Department of Urology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy.,Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Vincenzo Pompeo
- Department of Urology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Steno Sentinelli
- Department of Pathology, Regina Elena National Cancer, Institute, IRCCS, Rome, Italy
| | - Laura De Salvo
- Department of Pathology, Regina Elena National Cancer, Institute, IRCCS, Rome, Italy
| | - Mario Patrizio
- Department of Drug Research and Medicine Evaluation, Istituto Superiore di Sanita', Rome, Italy
| | | | - Michele Gallucci
- Department of Urology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Mauro Piacentini
- Laboratory of Cell Biology and Electron Microscopy, National Institute for Infectious Diseases I.R.C.C.S. 'L. Spallanzani', Rome, Italy.,Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Walter Malorni
- Department of Drug Research and Medicine Evaluation, Istituto Superiore di Sanita', Rome, Italy. .,Istituto San Raffaele Pisana, Rome, Italy.
| |
Collapse
|
27
|
Lorion R, Bladou F, Spatz A, van Kempen L, Irani J. [Prostate cancer microenvironment: Its structure, functions and therapeutic applications]. Prog Urol 2016; 26:464-76. [PMID: 27423973 DOI: 10.1016/j.purol.2016.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 06/11/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION In the field of prostate cancer there is a growing tendency for more and more studies to emphasise the predominant role of the zone situated between the tumour and the host: the tumour microenvironment. The aim of this article is to describe the structure and the functions of the prostate cancer microenvironment as well as the principal treatments that are being applied to it. MATERIAL AND METHODS PubMed and ScienceDirect databases have been interrogated using the association of keywords "tumour microenvironment" and "neoplasm therapy" along with "microenvironnement tumoral" and "traitements". Of the 593 articles initially found, 50 were finally included. RESULTS The tumour microenvironment principally includes host elements that are diverted from their primary functions and encourage the development of the tumour. In it we find immunity cells, support tissue as well as vascular and lymphatic neovascularization. Highlighting the major role played by this microenvironment has led to the development of specific treatments, notably antiangiogenic therapy and immunotherapy. CONCLUSION The tumour microenvironment, the tumour and the host influence themselves mutually and create a variable situation over time. Improvement of the knowledge of the prostate cancer microenvironment gradually enables us to pass from an approach centred on the tumour to a broader approach to the whole tumoral ecosystem. This enabled the emergence of new treatments whose place in the therapeutic arsenal still need to be found.
Collapse
Affiliation(s)
- R Lorion
- Service d'urologie, centre hospitalo-universitaire la Milétrie, Poitiers, France; X chromosome and cancer laboratory, Lady Davis institute for medical research, Jewish general hospital, Montreal, Canada.
| | - F Bladou
- Department of urology, Jewish general hospital, Montreal, Canada; X chromosome and cancer laboratory, Lady Davis institute for medical research, Jewish general hospital, Montreal, Canada
| | - A Spatz
- Department of pathology, Jewish general hospital, Montreal, Canada; X chromosome and cancer laboratory, Lady Davis institute for medical research, Jewish general hospital, Montreal, Canada
| | - L van Kempen
- X chromosome and cancer laboratory, Lady Davis institute for medical research, Jewish general hospital, Montreal, Canada
| | - J Irani
- Service d'urologie, centre hospitalier universitaire de Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
28
|
Hernandez-Hoyos G, Sewell T, Bader R, Bannink J, Chenault RA, Daugherty M, Dasovich M, Fang H, Gottschalk R, Kumer J, Miller RE, Ravikumar P, Wiens J, Algate PA, Bienvenue D, McMahan CJ, Natarajan SK, Gross JA, Blankenship JW. MOR209/ES414, a Novel Bispecific Antibody Targeting PSMA for the Treatment of Metastatic Castration-Resistant Prostate Cancer. Mol Cancer Ther 2016; 15:2155-65. [PMID: 27406985 DOI: 10.1158/1535-7163.mct-15-0242] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/14/2016] [Indexed: 11/16/2022]
Abstract
Treatment of metastatic, castration-resistant prostate cancer (mCRPC) remains a highly unmet medical need and current therapies ultimately result in disease progression. Immunotherapy is a rapidly growing approach for treatment of cancer but has shown limited success to date in the treatment of mCRPC. We have developed a novel humanized bispecific antibody, MOR209/ES414, built on the ADAPTIR (modular protein technology) platform, to redirect T-cell cytotoxicity toward prostate cancer cells by specifically targeting T cells through CD3ε to prostate cancer cells expressing PSMA (prostate-specific membrane antigen). In vitro cross-linking of T cells with PSMA-expressing tumor cells by MOR209/ES414 triggered potent target-dependent tumor lysis and induction of target-dependent T-cell activation and proliferation. This activity occurred at low picomolar concentrations of MOR209/ES414 and was effective at low T-effector to tumor target cell ratios. In addition, cytotoxic activity was equivalent over a wide range of PSMA expression on target cells, suggesting that as few as 3,700 PSMA receptors per cell are sufficient for tumor lysis. In addition to high sensitivity and in vitro activity, MOR209/ES414 induced limited production of cytokines compared with other bispecific antibody formats. Pharmacokinetic analysis of MOR209/ES414 demonstrated a serum elimination half-life in NOD/SCID γ (NSG) mice of 4 days. Administration of MOR209/ES414 in murine xenograft models of human prostate cancer significantly inhibited tumor growth, prolonged survival, and decreased serum prostate-specific antigen levels only in the presence of adoptively transferred human T cells. On the basis of these preclinical findings, MOR209/ES414 warrants further investigation as a potential therapeutic for the treatment of CRPC. Mol Cancer Ther; 15(9); 2155-65. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hang Fang
- Emergent BioSolutions, Seattle, Washington
| | | | - John Kumer
- Emergent BioSolutions, Seattle, Washington
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Advanced hepatocellular carcinoma (HCC) is a serious therapeutic challenge and targeted therapies only provide a modest benefit in terms of overall survival. Novel approaches are urgently needed for the treatment of this prevalent malignancy. Evidence demonstrating the antigenicity of tumour cells, the discovery that immune checkpoint molecules have an essential role in immune evasion of tumour cells, and the impressive clinical results achieved by blocking these inhibitory receptors, are revolutionizing cancer immunotherapy. Here, we review the data on HCC immunogenicity, the mechanisms for HCC immune subversion and the different immunotherapies that have been tested to treat HCC. Taking into account the multiplicity of hyperadditive immunosuppressive forces acting within the HCC microenvironment, a combinatorial approach is advised. Strategies include combinations of systemic immunomodulation and gene therapy, cell therapy or virotherapy.
Collapse
|
30
|
Wirth MP, Froehner M. Prostate-specific Antigen Pox Virus Vaccination for Recurrent Prostate Cancer. Eur Urol 2015; 68:372-3. [DOI: 10.1016/j.eururo.2015.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 02/20/2015] [Indexed: 11/29/2022]
|
31
|
Arcangeli S, Zilli T, De Bari B, Alongi F. "Hit the primary": A paradigm shift in the treatment of metastatic prostate cancer? Crit Rev Oncol Hematol 2015; 97:231-7. [PMID: 26375435 DOI: 10.1016/j.critrevonc.2015.08.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 01/17/2023] Open
Abstract
Patients with metastatic prostate cancer (PC) represent a heterogeneous group with survival rates varying between 13 and 75 months. The current standard treatment in this setting is hormonal therapy, with or without docetaxel-based chemotherapy. In the era of individualized medicine, however, maximizing treatment options, especially in long-term surviving patients with limited disease burden, is of capital importance. Emerging data, mainly from retrospective surgical series, show survival benefits in men diagnosed with metastatic PC following definitive therapy for the prostate. Whether the irradiation of primary tumor in a metastatic disease might improve the therapeutic ratio in association with systemic treatments remains investigational. In this scenario, modern radiation therapy (RT) can play a significant role owing to its intrinsic capability to act as a more general immune response modifier, as well as to the potentially better toxicity profile compared to surgery. Preclinical data, clinical experience, and challenges in local treatment in de novo metastatic PC are reviewed and discussed.
Collapse
Affiliation(s)
- Stefano Arcangeli
- Radiation Oncology, San Camillo and Forlanini Hospital - Rome, Italy.
| | - Thomas Zilli
- Radiation Oncology Department, Hôpitaux Universitaires de Genève (HUG), Geneva, Switzerland
| | - Berardino De Bari
- Radiation Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV) - Lausanne, Switzerland
| | - Filippo Alongi
- Radiation Oncology Department, Sacro Cuore - Don Calabria Hospital - Negrar, (Verona), Italy
| |
Collapse
|