151
|
Ashraf MU, Farwa U, Siddiqa M, Sarfraz A, Azeem N, Sarfraz Z. Has the Landscape of Immunotherapy for Prostate Cancer Changed? A Systematic Review and Post Hoc Analysis. Am J Mens Health 2023; 17:15579883231165140. [PMID: 37002863 PMCID: PMC10069001 DOI: 10.1177/15579883231165140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-causing death in the United States. As the most common malignancy in men, it is pertinent to explore whether novel immunotherapies may improve the quality of life and overall survival (OS) of patient populations. This systematic review and post hoc analysis curates a patient-by-patient pool of evidence adhering to PRISMA Statement 2020 guidelines. In total, 24 patients were analyzed for treatment history and associated variables including prostate-specific antigen (PSA) levels at diagnosis and post-treatment, Gleason score, secondary tumor locations, success/failure of therapy, and post-immunotherapy outcomes including OS. In total, 10 types of immunotherapies were identified with Pembrolizumab (among 8 patients) followed by IMM-101 (among 6 patients) being the most commonly administered. The mean OS for all patients was 27.8 months (24 patients) with the relatively highest mean OS reported with IMM-101 (56 months) followed by tumor-infiltrating lymphocytes (30 months). This research article provides critical insights into the evolving landscape of immunotherapies being tested for PCa and addresses gaps in oncological research to advance the understanding of PCa.
Collapse
Affiliation(s)
| | - Ume Farwa
- University Medical and Dental College, Faisalabad, Pakistan
| | - Maryam Siddiqa
- University Medical and Dental College, Faisalabad, Pakistan
| | | | - Nishwa Azeem
- Schwarzman College, Tsinghua University, Beijing, China
| | | |
Collapse
|
152
|
Naito Y, Kato M, Kawanishi H, Yamamoto A, Sakamoto F, Hirabayashi H, Kobayashi M, Matsukawa Y, Kimura T, Araki H, Nishikimi T, Kondo A, Yoshino Y, Hashimoto Y, Nakano Y, Tsuzuki T. Clinical utility of intraductal carcinoma of the prostate in treatment selection for metastatic hormone-sensitive prostate cancer. Prostate 2023; 83:307-315. [PMID: 36420892 DOI: 10.1002/pros.24462] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/25/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND In recent years, the usefulness of androgen receptor axis-targeted agents (ARATs) such as abiraterone, enzalutamide, and apalutamide for the upfront treatment of metastatic hormone-sensitive prostate cancer (mHSPC) has been demonstrated. However, it remains unclear which patients would truly benefit from these treatments. Furthermore, intraductal carcinoma of the prostate (IDC-P) is a known poor prognostic factor in patients with prostate cancer. We investigated the association between the presence of IDC-P and response to therapy in patients with mHSPC. METHODS This retrospective analysis included 318 patients with mHSPC who received treatment at Nagoya University and its 12 affiliated institutions between 2014 and 2021. Their biopsy specimens were evaluated for the presence of IDC-P. The patients were classified according to their first-line treatment into the ARAT (n = 100, receiving a combination of androgen-deprivation therapy [ADT] and ARAT) or conventional therapy (n = 218, receiving ADT with or without standard antiandrogen agents) group. We compared the overall survival (OS) and second progression-free survival (PFS2) between the ARAT and conventional groups according to the presence of IDC-P to evaluate whether presence of IDC-P predicts the response to each treatment. PFS2 was defined as the period from mHSPC diagnosis to disease progression on second-line treatment or death. Propensity score matching with one-to-one nearest-neighbor matching was used to minimize the potential effects of selection bias and confounding factors. The clinicopathological variables of the patients were well-balanced after propensity score matching. RESULTS Most patients in the ARAT (79%) and conventional therapy (71%) groups were ICD-P positive. In the propensity score-matched cohort, the OS and PFS2 of IDC-P-positive patients were significantly longer in the ARAT group than in the conventional group (OS: hazard ratio [HR], 0.36; p = 0.047; PFS2: HR, 0.30; p < 0.001). In contrast, no difference in OS and PFS2 was observed between the ARAT and conventional groups in IDC-P-negative patients (OS: HR, 1.09; p = 0.920; PFS2: HR, 0.40; p = 0.264). CONCLUSIONS The findings highlight a high prevalence of IDC-P among patients with mHSPC and suggest that IDC-P positivity may be a reliable indicator that ARAT should be implemented as first-line treatment.
Collapse
Affiliation(s)
- Yushi Naito
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Kato
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideji Kawanishi
- Department of Urology, Aichi Medical University Hospital, Nagakute, Japan
| | - Akiyuki Yamamoto
- Department of Urology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | | | - Hiroki Hirabayashi
- Department of Urology, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Japan
| | | | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tohru Kimura
- Department of Urology, Japan Community Healthcare Organization Chukyo Hospital, Nagoya, Japan
| | | | - Toshinori Nishikimi
- Department of Urology, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Atsuya Kondo
- Department of Urology, Kariya Toyota General Hospital, Kariya, Japan
| | - Yasushi Yoshino
- Department of Urology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | | | - Yojiro Nakano
- Department of Urology, Tosei General Hospital, Seto, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Japan
| |
Collapse
|
153
|
Reizine N, Socco S, Weng E, Atueyi U, Mehta V, Patnaik A. Utilization of Next-Generation Sequencing in Conjunction With Immunohistochemistry to Predict Exceptional Response to Combination Immune Checkpoint Therapy in a Heavily Pretreated Patient With Castration-Resistant Prostate Cancer. JCO Precis Oncol 2023; 7:e2200483. [PMID: 36996374 DOI: 10.1200/po.22.00483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/10/2023] [Indexed: 04/01/2023] Open
Affiliation(s)
- Natalie Reizine
- Division of Hematology and Oncology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Samantha Socco
- College of Pharmacy, The University of Illinois at Chicago, Chicago, IL
| | - Eric Weng
- Division of Hematology and Oncology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Uka Atueyi
- Department of Radiology, The University of Illinois at Chicago, Chicago, IL
| | - Vikas Mehta
- Department of Pathology, The University of Illinois at Chicago, Chicago, IL
| | - Akash Patnaik
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL
| |
Collapse
|
154
|
Mehra N, Kloots I, Vlaming M, Aluwini S, Dewulf E, Oprea-Lager DE, van der Poel H, Stoevelaar H, Yakar D, Bangma CH, Bekers E, van den Bergh R, Bergman AM, van den Berkmortel F, Boudewijns S, Dinjens WN, Fütterer J, van der Hulle T, Jenster G, Kroeze LI, van Kruchten M, van Leenders G, van Leeuwen PJ, de Leng WW, van Moorselaar RJA, Noordzij W, Oldenburg RA, van Oort IM, Oving I, Schalken JA, Schoots IG, Schuuring E, Smeenk RJ, Vanneste BG, Vegt E, Vis AN, de Vries K, Willemse PPM, Wondergem M, Ausems M. Genetic Aspects and Molecular Testing in Prostate Cancer: A Report from a Dutch Multidisciplinary Consensus Meeting. EUR UROL SUPPL 2023; 49:23-31. [PMID: 36874601 PMCID: PMC9975012 DOI: 10.1016/j.euros.2022.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 01/27/2023] Open
Abstract
Background Germline and tumour genetic testing in prostate cancer (PCa) is becoming more broadly accepted, but testing indications and clinical consequences for carriers in each disease stage are not yet well defined. Objective To determine the consensus of a Dutch multidisciplinary expert panel on the indication and application of germline and tumour genetic testing in PCa. Design setting and participants The panel consisted of 39 specialists involved in PCa management. We used a modified Delphi method consisting of two voting rounds and a virtual consensus meeting. Outcome measurements and statistical analysis Consensus was reached if ≥75% of the panellists chose the same option. Appropriateness was assessed by the RAND/UCLA appropriateness method. Results and limitations Of the multiple-choice questions, 44% reached consensus. For men without PCa having a relevant family history (familial PCa/BRCA-related hereditary cancer), follow-up by prostate-specific antigen was considered appropriate. For patients with low-risk localised PCa and a family history of PCa, active surveillance was considered appropriate, except in case of the patient being a BRCA2 germline pathogenic variant carrier. Germline and tumour genetic testing should not be done for nonmetastatic hormone-sensitive PCa in the absence of a relevant family history of cancer. Tumour genetic testing was deemed most appropriate for the identification of actionable variants, with uncertainty for germline testing. For tumour genetic testing in metastatic castration-resistant PCa, consensus was not reached for the timing and panel composition. The principal limitations are as follows: (1) a number of topics discussed lack scientific evidence, and therefore the recommendations are partly opinion based, and (2) there was a small number of experts per discipline. Conclusions The outcomes of this Dutch consensus meeting may provide further guidance on genetic counselling and molecular testing related to PCa. Patient summary A group of Dutch specialists discussed the use of germline and tumour genetic testing in prostate cancer (PCa) patients, indication of these tests (which patients and when), and impact of these tests on the management and treatment of PCa.
Collapse
Affiliation(s)
- Niven Mehra
- Department of Medical Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - Iris Kloots
- Department of Medical Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - Michiel Vlaming
- Division Laboratories, Pharmacy and biomedical Genetics, Department of Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Shafak Aluwini
- Department of Radiation Oncology, UMCG, Groningen, The Netherlands
| | - Els Dewulf
- Centre for Decision Analysis & Support, Ismar Healthcare NV, Lier, Belgium
| | - Daniela E. Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Henk van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Herman Stoevelaar
- Centre for Decision Analysis & Support, Ismar Healthcare NV, Lier, Belgium
| | - Derya Yakar
- Department of Radiology, UMCG, Groningen, The Netherlands
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Chris H. Bangma
- Department of Urology, Erasmus MC, Rotterdam, The Netherlands
| | - Elise Bekers
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | - Andries M. Bergman
- Department of Medical Oncology and Oncogenomics, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | - Steve Boudewijns
- Department of Medical Oncology, Bravis Hospital, Roosendaal, The Netherlands
| | | | - Jurgen Fütterer
- Department of Medical Imaging, Radboud UMC, Nijmegen, The Netherlands
| | - Tom van der Hulle
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Guido Jenster
- Department of Urology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Michel van Kruchten
- Department of Medical Oncology, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Pim J. van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | | | - Walter Noordzij
- Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Irma Oving
- Department of Internal Medicine, Ziekenhuis Groep Twente, Almelo, The Netherlands
| | | | - Ivo G. Schoots
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ed Schuuring
- Department of Pathology, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert J. Smeenk
- Department of Radiation Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - Ben G.L. Vanneste
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht UMC, Maastricht, The Netherlands
- Department of Human Structure and Repair, Ghent University Hospital, Ghent, Belgium
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Erik Vegt
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - André N. Vis
- Department of Urology, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Kim de Vries
- Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Maurits Wondergem
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Margreet Ausems
- Division Laboratories, Pharmacy and biomedical Genetics, Department of Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
155
|
Siddiqui BA, Chapin BF, Jindal S, Duan F, Basu S, Yadav SS, Gu AD, Espejo AB, Kinder M, Pettaway CA, Ward JF, Tidwell RSS, Troncoso P, Corn PG, Logothetis CJ, Knoblauch R, Hutnick N, Gottardis M, Drake CG, Sharma P, Subudhi SK. Immune and pathologic responses in patients with localized prostate cancer who received daratumumab (anti-CD38) or edicotinib (CSF-1R inhibitor). J Immunother Cancer 2023; 11:e006262. [PMID: 36948506 PMCID: PMC10040066 DOI: 10.1136/jitc-2022-006262] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND The prostate tumor microenvironment (TME) is immunosuppressive, with few effector T cells and enrichment of inhibitory immune populations, leading to limited responses to treatments such as immune checkpoint therapies (ICTs). The immune composition of the prostate TME differs across soft tissue and bone, the most common site of treatment-refractory metastasis. Understanding immunosuppressive mechanisms specific to prostate TMEs will enable rational immunotherapy strategies to generate effective antitumor immune responses. Daratumumab (anti-CD38 antibody) and edicotinib (colony-stimulating factor-1 receptor (CSF-1R) inhibitor) may alter the balance within the prostate TME to promote antitumor immune responses. HYPOTHESIS Daratumumab or edicotinib will be safe and will alter the immune TME, leading to antitumor responses in localized prostate cancer. PATIENTS AND METHODS In this presurgical study, patients with localized prostate cancer received 4 weekly doses of daratumumab or 4 weeks of daily edicotinib prior to radical prostatectomy (RP). Treated and untreated control (Gleason score ≥8 in prostate biopsy) prostatectomy specimens and patient-matched pre- and post-treatment peripheral blood mononuclear cells (PBMCs) and bone marrow samples were evaluated. The primary endpoint was incidence of adverse events (AEs). The secondary endpoint was pathologic complete remission (pCR) rate. RESULTS Twenty-five patients were treated (daratumumab, n=15; edicotinib, n=10). All patients underwent RP without delays. Grade 3 treatment-related AEs with daratumumab occurred in 3 patients (12%), and no ≥grade 3 treatment-related AEs occurred with edicotinib. No changes in serum prostate-specific antigen (PSA) levels or pCRs were observed. Daratumumab led to a decreased frequency of CD38+ T cells, natural killer cells, and myeloid cells in prostate tumors, bone marrow, and PBMCs. There were no consistent changes in CSF-1R+ immune cells in prostate, bone marrow, or PBMCs with edicotinib. Neither treatment induced T cell infiltration into the prostate TME. CONCLUSIONS Daratumumab and edicotinib treatment was safe and well-tolerated in patients with localized prostate cancer but did not induce pCRs. Decreases in CD38+ immune cells were observed in prostate tumors, bone marrow, and PBMCs with daratumumab, but changes in CSF-1R+ immune cells were not consistently observed with edicotinib. Neither myeloid-targeted agent alone was sufficient to generate antitumor responses in prostate cancer; thus, combinations with agents to induce T cell infiltration (eg, ICTs) will be needed to overcome the immunosuppressive prostate TME.
Collapse
Affiliation(s)
- Bilal A Siddiqui
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brian F Chapin
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sonali Jindal
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fei Duan
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sreyashi Basu
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shalini S Yadav
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ai-Di Gu
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexsandra B Espejo
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michelle Kinder
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Curtis A Pettaway
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John F Ward
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rebecca S S Tidwell
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paul G Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Natalie Hutnick
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Marco Gottardis
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Charles G Drake
- Janssen Research & Development, Spring House, Pennsylvania, USA
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
- Department of Urology, Columbia University Medical Center, New York, New York, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Immunotherapy Platform, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
156
|
Koster KL, Zumstein V, Süveg K, Jochum W, Husarik DB, Omlin A. Case of the month from the Cantonal Hospital, St Gallen, Switzerland: checkpoint inhibition for recurrent prostate cancer. BJU Int 2023; 131:173-176. [PMID: 36468816 DOI: 10.1111/bju.15847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Kira-Lee Koster
- Department of Medical Oncology and Hematology, Cantonal Hospital, St Gallen, Switzerland
| | | | - Krisztian Süveg
- Department of Radiation Oncology, Cantonal Hospital, St Gallen, Switzerland
| | - Wolfram Jochum
- Institute of Pathology, Cantonal Hospital, St Gallen, Switzerland
| | | | - Aurelius Omlin
- Department of Medical Oncology and Hematology, Cantonal Hospital, St Gallen, Switzerland
| |
Collapse
|
157
|
Cao Y, Lu M, Sun Y, Gong J, Li J, Lu Z, Li J, Zhang X, Li Y, Peng Z, Zhou J, Wang X, Shen L. Surufatinib plus toripalimab in patients with advanced solid tumors: a single-arm, open-label, phase 1 trial. J Cancer Res Clin Oncol 2023; 149:779-789. [PMID: 35166929 PMCID: PMC9931771 DOI: 10.1007/s00432-021-03898-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/21/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE This phase 1 trial evaluated the safety, preliminary efficacy, and pharmacokinetics of surufatinib, a small molecular tyrosine kinase inhibitor, combined with toripalimab, a programmed cell death protein-1 antibody, in patients with advanced solid tumors. METHODS This is an open-label, dose-escalation and expansion study in patients with solid tumors who had failed standard therapies or had no effective treatment. In the dose-escalation stage, patients were treated with surufatinib, at dose levels of 200, 250, or 300 mg once daily (QD) in combination with toripalimab 240 mg, every 3 weeks (Q3W), to estimate maximum tolerated dose. Additional patients were enrolled in the dose expansion stage to further assess the efficacy, safety, and pharmacokinetics profile. Recommended phase 2 dose (RP2D) was determined based on the safety, tolerability, and preliminary efficacy from dose-escalation and expansion stages. RESULTS From Feb 14, 2019 to Dec 20, 2020, 33 patients were screened, of which 30 patients were enrolled. One patient in the 300 mg cohort experienced dose limited toxicity, a grade 3 hyperthyroidism. The most frequent treatment-related adverse events of grade ≥ 3 were hypertension (20.0%), transaminases increased (13.3%), and blood bilirubin increased (13.3%). No treatment-related death or treatment discontinuation was identified. The RP2D was determined to be surufatinib 250 mg QD plus toripalimab 240 mg Q3W. Objective response rate was 24.1% (95% confidence interval 10.3‒43.5%) in this study. CONCLUSIONS Surufatinib plus toripalimab was well tolerated, with no unexpected safety signals, and showed preliminary anti-tumor activity in patients with advanced solid tumors. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT03879057; registration date: March 18, 2019.
Collapse
Affiliation(s)
- Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Yu Sun
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Jie Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Yan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Xicheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China.
- Department of Early Drug Development Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
158
|
Brown LC, Armstrong AJ. Germline Testing in Prostate Cancer: Implementation and Disparities of Care. JCO Oncol Pract 2023; 19:221-223. [PMID: 36720077 DOI: 10.1200/op.22.00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC
| |
Collapse
|
159
|
Catalano M, Generali D, Gatti M, Riboli B, Paganini L, Nesi G, Roviello G. DNA repair deficiency as circulating biomarker in prostate cancer. Front Oncol 2023; 13:1115241. [PMID: 36793600 PMCID: PMC9922904 DOI: 10.3389/fonc.2023.1115241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Deleterious aberrations in DNA repair genes are actionable in approximately 25% of metastatic castration-resistant prostate cancers (mCRPC) patients. Homology recombination repair (HRR) is the DNA damage repair (DDR) mechanism most frequently altered in prostate cancer; of note BRCA2 is the most frequently altered DDR gene in this tumor. Poly ADP-ribose polymerase inhibitors showed antitumor activity with a improvement in overall survival in mCRPC carrying somatic and/or germline alterations of HHR. Germline mutations are tested on peripheral blood samples using DNA extracted from peripheral blood leukocytes, while the somatic alterations are assessed by extracting DNA from a tumor tissue sample. However, each of these genetic tests have some limitations: the somatic tests are related to the sample availability and tumor heterogeneity, while the germline testing are mainly related to the inability to detect somatic HRR mutations. Therefore, the liquid biopsy, a non-invasive and easily repeatable test compared to tissue test, could identified somatic mutation detected on the circulating tumor DNA (ctDNA) extracted from a plasma. This approach should better represent the heterogeneity of the tumor compared to the primary biopsy and maybe helpful in monitoring the onset of potential mutations involved in treatment resistance. Furthermore, ctDNA may inform about timing and potential cooperation of multiple driver genes aberration guiding the treatment options in patients with mCRPC. However, the clinical use of ctDNA test in prostate cancer compared to blood and tissue testing are currently very limited. In this review, we summarize the current therapeutic indications in prostate cancer patients with DDR deficiency, the recommendation for germline and somatic-genomic testing in advanced PC and the advantages of the use liquid biopsy in clinical routine for mCRPC.
Collapse
Affiliation(s)
- Martina Catalano
- School of Human Health Sciences, University of Florence, Florence, Italy
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital Trieste, Trieste, Italy
| | - Marta Gatti
- Servizio di Citogenetica e Genetica - Azienda Socio-Sanitaria Territoriale (ASST) di Cremona, Cremona, Italy
| | - Barbara Riboli
- Servizio di Citogenetica e Genetica - Azienda Socio-Sanitaria Territoriale (ASST) di Cremona, Cremona, Italy
| | - Leda Paganini
- Servizio di Citogenetica e Genetica - Azienda Socio-Sanitaria Territoriale (ASST) di Cremona, Cremona, Italy
| | - Gabriella Nesi
- Department of Health Sciences, University of Florence, Florence, Italy
| | | |
Collapse
|
160
|
Cheng X, Zeng Z, Yang H, Chen Y, Liu Y, Zhou X, Zhang C, Wang G. Novel cuproptosis-related long non-coding RNA signature to predict prognosis in prostate carcinoma. BMC Cancer 2023; 23:105. [PMID: 36717792 PMCID: PMC9887873 DOI: 10.1186/s12885-023-10584-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Cuproptosis, an emerging form of programmed cell death, has recently been identified. However, the association between cuproptosis-related long non-coding RNA (lncRNA) signature and the prognosis in prostate carcinoma remains elusive. This study aims to develop the novel cuproptosis-related lncRNA signature in prostate cancer and explore its latent molecular function. METHODS RNA-seq data and clinical information were downloaded from the TCGA datasets. Then, cuproptosis-related gene was identified from the previous literature and further applied to screen the cuproptosis-related differentially expressed lncRNAs. Patients were randomly assigned to the training cohort or the validation cohort with a 1:1 ratio. Subsequently, the machine learning algorithms (Lasso and stepwise Cox (direction = both)) were used to construct a novel prognostic signature in the training cohorts, which was validated by the validation and the entire TCGA cohorts. The nomogram base on the lncRNA signature and several clinicopathological traits were constructed to predict the prognosis. Functional enrichment and immune analysis were performed to evaluate its potential mechanism. Furthermore, differences in the landscape of gene mutation, tumour mutational burden (TMB), microsatellite instability (MSI), drug sensitivity between both risk groups were also assessed to explicit their relationships. RESULTS The cuproptosis-related lncRNA signature was constructed based on the differentially expressed cuproptosis-related lncRNAs, including AC005790.1, AC011472.4, AC099791.2, AC144450.1, LIPE-AS1, and STPG3-AS1. Kaplan-Meier survival and ROC curves demonstrate that the prognosis signature as an independent risk indicator had excellent potential to predict the prognosis in prostate cancer. The signature was closely associated with age, T stage, N stage, and the Gleason score. Immune analysis shows that the high-risk group was in an immunosuppressive microenvironment. Additionally, the significant difference in landscape of gene mutation, tumour mutational burden, microsatellite instability, and drug sensitivity between both risk groups was observed. CONCLUSIONS A novel cuproptosis-related lncRNA signature was constructed using machine learning algorithms to predict the prognosis of prostate cancer. It was closely with associated with several common clinical traits, immune cell infiltration, immune-related functions, immune checkpoints, gene mutation, TMB, MSI, and the drug sensitivity, which may be useful to improve the clinical outcome.
Collapse
Affiliation(s)
- Xiaofeng Cheng
- grid.412604.50000 0004 1758 4073Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000 People’s Republic of China ,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000 China
| | - Zhenhao Zeng
- grid.412604.50000 0004 1758 4073Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000 People’s Republic of China ,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000 China
| | - Heng Yang
- grid.412604.50000 0004 1758 4073Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000 People’s Republic of China ,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000 China
| | - Yujun Chen
- grid.412604.50000 0004 1758 4073Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000 People’s Republic of China ,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000 China
| | - Yifu Liu
- grid.412604.50000 0004 1758 4073Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000 People’s Republic of China ,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000 China
| | - Xiaochen Zhou
- grid.412604.50000 0004 1758 4073Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000 People’s Republic of China ,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000 China
| | - Cheng Zhang
- grid.412604.50000 0004 1758 4073Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000 People’s Republic of China ,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000 China
| | - Gongxian Wang
- Department of Urology, Jiangxi Province, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang City, 330000, People's Republic of China. .,Jiangxi Institute of Urology, Nanchang City, Jiangxi Province, 330000, China.
| |
Collapse
|
161
|
Styk J, Pös Z, Pös O, Radvanszky J, Turnova EH, Buglyó G, Klimova D, Budis J, Repiska V, Nagy B, Szemes T. Microsatellite instability assessment is instrumental for Predictive, Preventive and Personalised Medicine: status quo and outlook. EPMA J 2023; 14:143-165. [PMID: 36866160 PMCID: PMC9971410 DOI: 10.1007/s13167-023-00312-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023]
Abstract
A form of genomic alteration called microsatellite instability (MSI) occurs in a class of tandem repeats (TRs) called microsatellites (MSs) or short tandem repeats (STRs) due to the failure of a post-replicative DNA mismatch repair (MMR) system. Traditionally, the strategies for determining MSI events have been low-throughput procedures that typically require assessment of tumours as well as healthy samples. On the other hand, recent large-scale pan-tumour studies have consistently highlighted the potential of massively parallel sequencing (MPS) on the MSI scale. As a result of recent innovations, minimally invasive methods show a high potential to be integrated into the clinical routine and delivery of adapted medical care to all patients. Along with advances in sequencing technologies and their ever-increasing cost-effectiveness, they may bring about a new era of Predictive, Preventive and Personalised Medicine (3PM). In this paper, we offered a comprehensive analysis of high-throughput strategies and computational tools for the calling and assessment of MSI events, including whole-genome, whole-exome and targeted sequencing approaches. We also discussed in detail the detection of MSI status by current MPS blood-based methods and we hypothesised how they may contribute to the shift from conventional medicine to predictive diagnosis, targeted prevention and personalised medical services. Increasing the efficacy of patient stratification based on MSI status is crucial for tailored decision-making. Contextually, this paper highlights drawbacks both at the technical level and those embedded deeper in cellular/molecular processes and future applications in routine clinical testing.
Collapse
Affiliation(s)
- Jakub Styk
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia ,Comenius University Science Park, 841 04 Bratislava, Slovakia ,Geneton Ltd, 841 04 Bratislava, Slovakia
| | - Zuzana Pös
- Comenius University Science Park, 841 04 Bratislava, Slovakia ,Geneton Ltd, 841 04 Bratislava, Slovakia ,Institute of Clinical and Translational Research, Biomedical Research Centre, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Ondrej Pös
- Comenius University Science Park, 841 04 Bratislava, Slovakia ,Geneton Ltd, 841 04 Bratislava, Slovakia
| | - Jan Radvanszky
- Comenius University Science Park, 841 04 Bratislava, Slovakia ,Institute of Clinical and Translational Research, Biomedical Research Centre, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia ,Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia
| | - Evelina Hrckova Turnova
- Comenius University Science Park, 841 04 Bratislava, Slovakia ,Slovgen Ltd, 841 04 Bratislava, Slovakia
| | - Gergely Buglyó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Daniela Klimova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Jaroslav Budis
- Comenius University Science Park, 841 04 Bratislava, Slovakia ,Geneton Ltd, 841 04 Bratislava, Slovakia ,Slovak Centre of Scientific and Technical Information, 811 04 Bratislava, Slovakia
| | - Vanda Repiska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia ,Medirex Group Academy, NPO, 949 05 Nitra, Slovakia
| | - Bálint Nagy
- Comenius University Science Park, 841 04 Bratislava, Slovakia ,Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tomas Szemes
- Comenius University Science Park, 841 04 Bratislava, Slovakia ,Geneton Ltd, 841 04 Bratislava, Slovakia ,Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia
| |
Collapse
|
162
|
Fang B, Wei Y, Pan J, Zhang T, Ye D, Zhu Y. The Somatic Mutational Landscape of Mismatch Repair Deficient Prostate Cancer. J Clin Med 2023; 12:jcm12020623. [PMID: 36675550 PMCID: PMC9864094 DOI: 10.3390/jcm12020623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Prostate cancers with mismatch repair deficiency (MMR-d) have aggressive clinical and histological features, and they are potentially responsive to immunotherapy. However, its rarity prevents the analysis of the underlying biology. Here, we collected the genomic data of 2664 primary prostate tumors and 1409 metastatic prostate tumors from the GENIE and TCGA databases. A total of 69 (2.59%) primary and 60 (4.26%) metastatic MMR-d tumors were identified among these tumors. Single nucleotide variant (SNV) frequencies of 34 candidate genes (including KMT2D (46.4%), ZFHX3 (33.3%), JAK1 (31.9%), and RNF43 (27.5%)) and 16 candidate genes (including KMT2D (33.3%) and JAK1 (28.3%)) were higher in MMR-d primary tumors and MMR-d metastatic tumors, respectively. The tumor mutation burden (TMB) was higher in primary MMR-d tumors. Homozygous deletions of EPCAM and EPAS1 were enriched in MMR-d primary tumors, while EPCAM deletions were enriched in metastatic MMR-d tumors. For genomic rearrangement events, TMPRSS2-ETS fusions were less frequent in primary MMR-d tumors. Our study indicates MMR-d prostate cancers have unique genomic features. These may play an important role in providing therapeutic targets for the treatment of this subset of prostate cancer patients.
Collapse
Affiliation(s)
- Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tingwei Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: (D.Y.); (Y.Z.); Tel.: +86-21-64175590 (D.Y. & Y.Z.); Fax: +86-21-64434556 (D.Y. & Y.Z.)
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: (D.Y.); (Y.Z.); Tel.: +86-21-64175590 (D.Y. & Y.Z.); Fax: +86-21-64434556 (D.Y. & Y.Z.)
| |
Collapse
|
163
|
Zhang Y, Ji Q, Wang J, Dong Y, Pang M, Fu S, Wei Y, Zhu Q. High expression of KNL1 in prostate adenocarcinoma is associated with poor prognosis and immune infiltration. Front Genet 2023; 13:1100787. [PMID: 36685823 PMCID: PMC9853456 DOI: 10.3389/fgene.2022.1100787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Prostate adenocarcinoma (PRAD) is a common malignancy with increasing morbidity and mortality. Kinetochore scaffold 1 (KNL1) has been reported to be involved in tumor progression and prognosis in other tumors, but its role in PRAD has not been reported in detail. KNL1 expression analysis, clinicopathological parameters analysis, prognostic correlation analysis, molecular interaction network and functional abdominal muscle analysis and immune infiltration analysis by using multiple online databases and downloaded expression profile. The results suggest that KNL1 is highly expressed in PRAD, which is associated with worse prognosis in PRAD patients. KnL1-related genes are highly enriched in mitotic function, which is considered to be highly related to the development of cancer. Finally, KNL1 expression is associated with a variety of tumor infiltrating immune cells, especially Treg and Th2 cells. In conclusion, our findings provide preliminary evidence that KNL1 may be an independent prognostic predictor of PRAD and is associated with immune infiltration.
Collapse
Affiliation(s)
- Yetao Zhang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianying Ji
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jun Wang
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuxiang Dong
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mingyang Pang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shengqiang Fu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Wei
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Yong Wei, ; Qingyi Zhu,
| | - Qingyi Zhu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,*Correspondence: Yong Wei, ; Qingyi Zhu,
| |
Collapse
|
164
|
Sfakianaki M, Tzardi M, Tsantaki K, Koutoulaki C, Messaritakis I, Datseri G, Moustou E, Mavroudis D, Souglakos J. Evaluation of Microsatellite Instability Molecular Analysis versus Immuno-Histochemical Interpretation in Malignant Neoplasms with Different Localizations. Cancers (Basel) 2023; 15:cancers15020353. [PMID: 36672302 PMCID: PMC9856558 DOI: 10.3390/cancers15020353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/09/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
MMR gene germline mutations are considered a major genetic disorder in patients with hereditary nonpolyposis colon cancer (HNPCC) or Lynch syndrome; A total of 15% of sporadic colon carcinomas are MSI-High. MSI has also been observed in other cancers, such as endometrial, gastric, and ovarian cancer. The aim of the current study was to correlate and outline the optimal method between the molecular testing of the instability of microsatellite DNA regions (MSI status) and the loss of protein expression by immunehistochemistry (MMR). A total of 242 paraffin-embedded tissues from gastrointestinal, gynecological, genitourinary, lung, breast, and unknown primary cancer patients were analyzed for the expression of MLH1/MSH2/MSH6/PMS2 by immunohistochemistry, as well as for the molecular analysis of MSI status using PCR-based molecular fragment analysis. A total of 29 MSI-High patients were detected molecularly, while 23 patients were detected by immunohistochemistry, with rates that are comparable according to the literature. Based on the agreement coefficient of the two methods, a substantial agreement emerged (Kappa = 0.675 with standard error = 0.081, p < 0.001). Despite the substantial agreement, both methods ought to be established to determine MSI-H/dMMR status in all cancer types as a first-line screening test.
Collapse
Affiliation(s)
- Maria Sfakianaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Correspondence: (M.S.); (I.M.); Tel.: +30-281-039-4926 (I.M.)
| | - Maria Tzardi
- Department of Pathology, University General Hospital of Heraklion, 71110 Heraklion, Greece
| | - Konstantina Tsantaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Chara Koutoulaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Correspondence: (M.S.); (I.M.); Tel.: +30-281-039-4926 (I.M.)
| | - Galateia Datseri
- Department of Pathology, University General Hospital of Heraklion, 71110 Heraklion, Greece
| | - Eleni Moustou
- Department of Pathology, University General Hospital of Heraklion, 71110 Heraklion, Greece
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Iraklio, Greece
| | - John Souglakos
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 71003 Heraklion, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Iraklio, Greece
| |
Collapse
|
165
|
Guo CC, Czerniak B. Updates of Prostate Cancer from the 2022 World Health Organization Classification of the Urinary and Male Genital Tumors. JOURNAL OF CLINICAL AND TRANSLATIONAL PATHOLOGY 2023; 3:26-34. [PMID: 38605939 PMCID: PMC11007763 DOI: 10.14218/jctp.2022.00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2023]
Abstract
Prostate cancer is a heterogeneous disease with a wide spectrum of pathological, clinical, and molecular features. The diagnosis and classification of prostate cancer have been constantly modified with the incorporation of new data. The 5th edition of the World Health Organization (WHO) Classification of Urinary and Genital Tumors was recently published six years after the 4th edition. In this new edition, the classification of prostate cancer has been refined in the diagnostic criteria, grading, nomenclature, and genomics. This paper reviews significant updates to the new WHO classification of prostate cancer, including high-grade prostatic intraepithelial neoplasia, acinar adenocarcinoma, intraductal carcinoma, ductal carcinoma, and neuroendocrine tumors. Controversial issues in the Gleason grading are discussed, such as intraductal carcinoma and tertiary grade. We also highlight distinct genetic and epigenetic alterations in prostate cancer that may contribute to its diverse clinicopathologic features. Overall, the 5th edition of the WHO classification provides a comprehensive assessment of prostate cancer with morphologic, immunohistochemical, genomic, and clinical data, which may represent an optimal paradigm for diagnosing and treating prostate cancer.
Collapse
Affiliation(s)
- Charles C. Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bogdan Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
166
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
167
|
Borgeaud M, Courtes MG, Tsantoulis P, Bodmer A, Labidi-Galy I, Koessler T. Immunotherapy in Urological, Gynecological and Gastrointestinal Cancers - Current Landscape. PRAXIS 2023; 112:149-155. [PMID: 36855887 DOI: 10.1024/1661-8157/a003974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Immunotherapy is becoming increasingly important in the management of urological, gynecological, and gastrointestinal cancers. Immune checkpoint inhibitor-based combinations have become a standard of care for patients with metastatic renal and liver cancers, as well as for many patients with bladder, cervical, gastric, and esophageal cancers, based on various biomarkers. Some tumor types are less responsive to immunotherapy, such as prostate and colon cancer. In these tumors, however, a subgroup of patients with a microsatellite-instability-high/DNA-mismatch repair deficient molecular phenotype significantly benefits from immunotherapy. Molecular characterization is therefore essential to identify patients who may benefit from these treatments. One of the major challenges is the search for new predictive biomarkers and novel combinations or strategies to further improve patient outcome.
Collapse
Affiliation(s)
- Maxime Borgeaud
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
- Co-first authors
| | | | - Petros Tsantoulis
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Alexandre Bodmer
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | | | - Thibaud Koessler
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| |
Collapse
|
168
|
Mouw KW, Choudhury AD. Development of PARP Inhibitors in Targeting Castration-Resistant Prostate Cancer. Cancer Treat Res 2023; 186:103-124. [PMID: 37978133 DOI: 10.1007/978-3-031-30065-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Prostate cancer is a genetically heterogenous disease and a subset of prostate tumors harbor alterations in DNA damage and repair (DDR) genes. Prostate tumor DDR gene alterations can arise via germline or somatic events and are enriched in high-grade and advanced disease. Alterations in genes in the homologous recombination (HR) repair pathway are associated with sensitivity to PARP inhibition in breast and ovarian cancer, and data from recently completed randomized trials also demonstrate benefit of PARP inhibitor therapy in patients with advanced metastatic castration-resistant prostate cancer (mCRPC) and tumor HR gene alterations. PARP inhibitors have been investigated in first-line mCRPC in biomarker-selected and unselected populations, and are currently under study in earlier disease states in patients with DDR gene alterations. This chapter focuses on the current state of PARP inhibitor development in prostate cancer with particular emphasis on biomarkers and combination therapy approaches.
Collapse
Affiliation(s)
- Kent W Mouw
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham & Women's Hospital, Harvard Medical School, 450 Brookline Ave., HIM 328, Boston, MA, 02215, USA.
| | - Atish D Choudhury
- Harvard Medical School, Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave., Dana 930, Boston, MA, 02215, USA
| |
Collapse
|
169
|
Chan EM, Foster KJ, Bass AJ. WRN Is a Promising Synthetic Lethal Target for Cancers with Microsatellite Instability (MSI). Cancer Treat Res 2023; 186:313-328. [PMID: 37978143 DOI: 10.1007/978-3-031-30065-3_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Microsatellite instability (MSI), a type of genetic hypermutability arising from impaired DNA mismatch repair (MMR), is observed in approximately 3% of all cancers. Preclinical work has identified the RecQ helicase WRN as a promising synthetic lethal target for patients with MSI cancers. WRN depletion substantially impairs the viability of MSI, but not microsatellite stable (MSS), cells. Experimental evidence suggests that this synthetic lethal phenotype is driven by numerous TA dinucleotide repeats that undergo expansion mutations in the setting of long-standing MMR deficiency. The lengthening of TA repeats increases their propensity to form secondary DNA structures that require WRN to resolve. In the absence of WRN helicase activity, these unresolved DNA secondary structures stall DNA replication forks and induce catastrophic DNA damage.
Collapse
Affiliation(s)
- Edmond M Chan
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, USA.
- Broad Institute of MIT and Harvard, Cambridge, USA.
- New York Genome Center, New York, USA.
| | | | - Adam J Bass
- Novartis Institutes for BioMedical Research, Cambridge, USA
| |
Collapse
|
170
|
Hoch D, Rabaglio M, Grob T, von Gunten M, Beyer J, Akhoundova D. Exceptional Response to Pembrolizumab in a Mismatch Repair-Deficient Aggressive Prostate Cancer with Somatic EPCAM, MSH2, and MSH6 Co-Deletion: A Case Report. Case Rep Oncol 2023; 16:1280-1286. [PMID: 37928863 PMCID: PMC10622161 DOI: 10.1159/000534177] [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: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 11/07/2023] Open
Abstract
Mismatch repair-deficient (dMMR) prostate cancer (PCa) is a rare (1-5%) but highly actionable molecular subgroup of PCa, vulnerable to immune checkpoint inhibitors. Our case of sporadic dMMR PCa due to large monoallelic co-deletion of EPCAM, MSH2, and MSH6 features a clinically aggressive disease presentation and a major response to pembrolizumab. We report a 65-year-old patient with primary metastatic PCa, Gleason score 5 + 5 = 10, with penile and lymph node metastases at diagnosis. Patient showed rapid progression on first-line ADT and enzalutamide. Tumor next-generation sequencing (NGS) revealed microsatellite instability and a tumor mutational burden of 40.8 mutations/megabase. Immunohistochemistry showed co-loss of MSH2 and MSH6. Review of NGS row data confirmed large monoallelic deletion in chromosome 2p, including EPCAM, MSH2, and MSH6. No germline alterations in mismatch repair genes were detected. Patient showed excellent response to pembrolizumab, which is still ongoing. We conclude that early molecular tumor profiling is essential to enable personalized management of advanced PCa, especially in patients with aggressive or atypical disease course.
Collapse
Affiliation(s)
- Dennis Hoch
- Department of Medical Oncology and Hematology, University Hospital of Bern, Bern, Switzerland
| | - Manuela Rabaglio
- Department of Medical Oncology and Hematology, University Hospital of Bern, Bern, Switzerland
| | - Tobias Grob
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | | | - Jörg Beyer
- Department of Medical Oncology and Hematology, University Hospital of Bern, Bern, Switzerland
| | - Dilara Akhoundova
- Department of Medical Oncology and Hematology, University Hospital of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
171
|
Atiq MO, Pastor DM, Karzai F, Hankin AR, Turkbey B, Cordes LM, Brownell I, Liu Y, Chesnut GT, Madan RA. First-line pembrolizumab plus androgen deprivation therapy for locally advanced microsatellite instability-high prostate cancer in a patient with Muir-Torre syndrome: A case report. Front Oncol 2023; 13:1126476. [PMID: 36937405 PMCID: PMC10022659 DOI: 10.3389/fonc.2023.1126476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
The risks of development of colorectal and endometrial cancers in individuals with Lynch syndrome (LS) are well known and have been widely studied. In recent years, the potential association of other malignancies, including prostate cancer, with LS has been considered. Decision-making regarding screening for prostate cancer in the generalized population can be complicated; accounting for the possibility of a higher risk of cancer conferred by a potential genetic predisposition confounds the creation of salient guidelines even further. Although tissue-agnostic treatment approvals have been granted to several immune checkpoint inhibitors (ICIs) for their use in the treatment of subsets of patients whose tumors exhibit high levels of microsatellite instability or high tumor mutational burden, a paucity of data exists regarding the use of ICIs in the first line treatment of patients with locally advanced prostate cancer harboring these features. A significant reduction in tumor volume in response to the combination of immune checkpoint inhibition and androgen deprivation therapy is described in this report of a male with Muir-Torre syndrome who was found to have locally advanced adenocarcinoma of the prostate. While anecdotal, the anti-tumor activity of this combination of therapy is notable and calls attention to the importance of considering further investigation of the use of immune checkpoint blockade as a primary therapeutic option in patients with localized prostate cancer.
Collapse
Affiliation(s)
- Mohammad O. Atiq
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Danielle M. Pastor
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Danielle M. Pastor,
| | - Fatima Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Amy R. Hankin
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Baris Turkbey
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Lisa M. Cordes
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Isaac Brownell
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yi Liu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Gregory T. Chesnut
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, United States
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ravi A. Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
172
|
Prevalence of mismatch repair genes mutations and clinical activity of PD-1 therapy in Chinese prostate cancer patients. Cancer Immunol Immunother 2022; 72:1541-1551. [DOI: 10.1007/s00262-022-03347-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
|
173
|
Fujita K, Suzuki H, Hinata N, Miura Y, Edamura K, Tabata KI, Arai G, Matsubara N, Yasumizu Y, Kosaka T, Oya M, Sugimoto M. Management of patients with advanced prostate cancer in Japan: 'real-world' consideration of the results from the Advanced Prostate Cancer Consensus Conference. Transl Androl Urol 2022; 11:1771-1785. [PMID: 36632151 PMCID: PMC9827400 DOI: 10.21037/tau-22-396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 10/14/2022] [Indexed: 11/21/2022] Open
Abstract
A multidisciplinary approach is necessary to manage advanced prostate cancer. The Advanced Prostate Cancer Consensus Conference (APCCC) in 2019 provided a practical guide to help clinicians consider therapeutic options in controversial areas, but healthcare systems vary across the world. At the 109th annual meeting of the Japanese Urological Association in December 2021, Japanese urologists voted on the questions in the APCCC 2019 guidelines regarding prostate-specific membrane antigen-positron emission tomography (PSMA-PET), management of oligometastatic prostate cancer, management of nonmetastatic castration-resistant prostate cancer (CRPC), management of a primary tumor in metastatic settings, systemic treatment of newly diagnosed metastatic castration-sensitive prostate cancer (CSPC), management of metastatic CRPC (mCRPC), and tumor genomic testing. We summarize the "real-world" status of the management of advanced prostate cancer in Japan. Several differences were noted in the management of advanced prostate cancer between Japanese urologists and the APCCC 2019 guidelines. Many Japanese urologists chose conventional imaging modalities for detecting metastasis instead of PSMA-PET. More Japanese urologists prefer androgen-deprivation therapy (ADT) alone in the management of low-volume metastatic CSPC than the APCCC panelists do, In the management of M0 CRPC, darolutamide and enzalutamide were chosen more by Japanese urologists than by the voters at the APCCC 2019. Bicalutamide remains one of the options for the management of mCRPC in Japan. More Japanese urologists do not recommend microsatellite instability (MSI) and BRCA1/2 tests than the voters at the APCCC 2019. Clinical evidence in Japan should be collected to address these discrepancies.
Collapse
Affiliation(s)
- Kazutoshi Fujita
- Department of Urology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Hiroyoshi Suzuki
- Department of Urology, Toho University Sakura Medical Center, Chiba, Japan
| | - Nobuyuki Hinata
- Department of Urology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuji Miura
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Kohei Edamura
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ken-Ichi Tabata
- Department of Urology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Gaku Arai
- Department of Urology, Dokkyo Medical University Saitama Medical Center, Koshigaya, Japan
| | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mikio Sugimoto
- Department of Urology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
174
|
Akhoundova D, Feng FY, Pritchard CC, Rubin MA. Molecular Genetics of Prostate Cancer and Role of Genomic Testing. Surg Pathol Clin 2022; 15:617-628. [PMID: 36344179 DOI: 10.1016/j.path.2022.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Prostate cancer (PCa) is characterized by profound genomic heterogeneity. Recent advances in personalized treatment entail an increasing need of genomic profiling. For localized PCa, gene expression assays can support clinical decisions regarding active surveillance and adjuvant treatment. In metastatic PCa, homologous recombination deficiency, microsatellite instability-high (MSI-H), and CDK12 deficiency constitute main actionable alterations. Alterations in DNA repair genes confer variable sensitivities to poly(ADP-ribose)polymerase inhibitors, and the use of genomic instability assays as predictive biomarker is still incipient. MSI can be assessed by immunohistochemistry To date there is a lack of consensus as to testing standards.
Collapse
Affiliation(s)
- Dilara Akhoundova
- Department for BioMedical Research, University of Bern, Murtenstrasse 24, Bern 3008, Switzerland; Department of Medical Oncology, Inselspital, University Hospital of Bern, Bern 3010, Switzerland
| | - Felix Y Feng
- Department of Radiation Oncology, University of California, 1600 Divisadero Street, Suite H-1031, San Francisco, CA 94115, USA
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, 1959 NE Pacific St Seattle, WA 98195-7110, USA
| | - Mark A Rubin
- Department for BioMedical Research, University of Bern, Murtenstrasse 24, Bern 3008, Switzerland; Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, 3008, Switzerland.
| |
Collapse
|
175
|
Steinkellner L, Luger F, Loidl W. [Importance of genetic testing in prostate cancer]. UROLOGIE (HEIDELBERG, GERMANY) 2022; 61:1392-1398. [PMID: 36378280 DOI: 10.1007/s00120-022-01977-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
Due to the increasing accessibility, molecular diagnostics are becoming more important in urology. There are validated predictive and prognostic biomarkers available for prostate cancer. This has clinical implications for screening diagnosis and personalized treatment options. The best known examples are mutations in the DNA damage repair genes breast cancer 1 (BRCA1) and BRCA2 for which the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib has been approved in 2020. This continuous progress enables a more tumor-specific and personalized treatment for prostate cancer. In order to provide this precision medicine, it is necessary to know the most important basic principles, have a close-knit interdisciplinary cooperation and access to a molecular tumor board due to the increased complexity.
Collapse
Affiliation(s)
- Lukas Steinkellner
- Abteilung für Urologie und Andrologie, Ordensklinikum Linz Elisabethinen, Fadingerstraße 1, 4020, Linz, Österreich
| | - Ferdinand Luger
- Abteilung für Urologie und Andrologie, Ordensklinikum Linz Elisabethinen, Fadingerstraße 1, 4020, Linz, Österreich
| | - Wolfgang Loidl
- Abteilung für Urologie und Andrologie, Ordensklinikum Linz Elisabethinen, Fadingerstraße 1, 4020, Linz, Österreich.
| |
Collapse
|
176
|
Fenor de la Maza MD, Chandran K, Rekowski J, Shui IM, Gurel B, Cross E, Carreira S, Yuan W, Westaby D, Miranda S, Ferreira A, Seed G, Crespo M, Figueiredo I, Bertan C, Gil V, Riisnaes R, Sharp A, Rodrigues DN, Rescigno P, Tunariu N, Liu XQ, Cristescu R, Schloss C, Yap C, de Bono JS. Immune Biomarkers in Metastatic Castration-resistant Prostate Cancer. Eur Urol Oncol 2022; 5:659-667. [PMID: 35491356 DOI: 10.1016/j.euo.2022.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/15/2022] [Accepted: 04/13/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is a heterogeneous disease in which molecular stratification is needed to improve clinical outcomes. The identification of predictive biomarkers can have a major impact on the care of these patients, but the availability of metastatic tissue samples for research in this setting is limited. OBJECTIVE To study the prevalence of immune biomarkers of potential clinical utility to immunotherapy in mCRPC and to determine their association with overall survival (OS). DESIGN, SETTING, AND PARTICIPANTS From 100 patients, mCRPC biopsies were assayed by whole exome sequencing, targeted next-generation sequencing, RNA sequencing, tumor mutational burden, T-cell-inflamed gene expression profile (TcellinfGEP) score (Nanostring), and immunohistochemistry for programmed cell death 1 ligand 1 (PD-L1), ataxia-telangiectasia mutated (ATM), phosphatase and tensin homolog (PTEN), SRY homology box 2 (SOX2), and the presence of neuroendocrine features. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The phi coefficient determined correlations between biomarkers of interest. OS was assessed using Kaplan-Meier curves and adjusted hazard ratios (aHRs) from Cox regression. RESULTS AND LIMITATIONS PD-L1 and SOX2 protein expression was detected by immunohistochemistry (combined positive score ≥1 and >5% cells, respectively) in 24 (33%) and 27 (27%) mCRPC biopsies, respectively; 23 (26%) mCRPC biopsies had high TcellinfGEP scores (>-0.318). PD-L1 protein expression and TcellinfGEP scores were positively correlated (phi 0.63 [0.45; 0.76]). PD-L1 protein expression (aHR: 1.90 [1.05; 3.45]), high TcellinfGEP score (aHR: 1.86 [1.04; 3.31]), and SOX2 expression (aHR: 2.09 [1.20; 3.64]) were associated with worse OS. CONCLUSIONS PD-L1, TcellinfGEP score, and SOX2 are prognostic of outcome from the mCRPC setting. If validated, predictive biomarker studies incorporating survival endpoints need to take these findings into consideration. PATIENT SUMMARY This study presents an analysis of immune biomarkers in biopsies from patients with metastatic prostate cancer. We describe tumor alterations that predict prognosis that can impact future studies.
Collapse
Affiliation(s)
| | - Khobe Chandran
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | | | | | - Bora Gurel
- The Institute of Cancer Research, London, UK
| | - Emily Cross
- The Institute of Cancer Research, London, UK
| | | | - Wei Yuan
- The Institute of Cancer Research, London, UK
| | - Daniel Westaby
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | - Susana Miranda
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | | | - George Seed
- The Institute of Cancer Research, London, UK
| | | | | | | | | | | | - Adam Sharp
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | | | | | - Nina Tunariu
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | | | | | | | | | - Johann S de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK.
| |
Collapse
|
177
|
Kwan EM, Wyatt AW, Chi KN. Towards clinical implementation of circulating tumor DNA in metastatic prostate cancer: Opportunities for integration and pitfalls to interpretation. Front Oncol 2022; 12:1054497. [PMID: 36439451 PMCID: PMC9685669 DOI: 10.3389/fonc.2022.1054497] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/25/2022] [Indexed: 08/13/2023] Open
Abstract
Plasma circulating tumor DNA (ctDNA) represents short fragments of tumor-derived DNA released into the bloodstream primarily from cancer cells undergoing apoptosis. In metastatic castration-resistant prostate cancer (mCRPC), characterizing genomic alterations in ctDNA identifies mutations, copy number alterations, and structural rearrangements with predictive and prognostic biomarker utility. These associations with clinical outcomes have resulted in ctDNA increasingly incorporated into routine clinical care. In this review, we summarize current and emerging applications for ctDNA analysis in metastatic prostate cancer, including outcome prediction, treatment selection, and characterization of treatment resistance. We also discuss potential pitfalls with interpreting ctDNA findings, namely false negatives arising from low tumor content and optimal assay design, including correction for clonal hematopoiesis of indeterminate potential and germline variants. Understanding the influence of these limitations on interpretation of ctDNA results is necessary to overcome barriers to clinical implementation. Nevertheless, as assay availability and technology continue to improve, recognizing both opportunities and shortcomings of ctDNA analysis will retain relevance with informing the implementation of precision-oncology initiatives for metastatic prostate cancer.
Collapse
Affiliation(s)
- Edmond M. Kwan
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
- BC Cancer, Vancouver Centre, Vancouver, BC, Canada
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Kim N. Chi
- Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, Canada
- BC Cancer, Vancouver Centre, Vancouver, BC, Canada
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
178
|
Aisa A, Weng S, Li X, Zhang D, Yuan Y. Immune Checkpoint Inhibitors combined with HER-2 targeted therapy in HER-2 positive Gastroesophageal cancer. Crit Rev Oncol Hematol 2022; 180:103864. [DOI: 10.1016/j.critrevonc.2022.103864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
|
179
|
Kotamarti S, Armstrong AJ, Polascik TJ, Moul JW. Molecular Mechanisms of Castrate-Resistant Prostate Cancer. Urol Clin North Am 2022; 49:615-626. [DOI: 10.1016/j.ucl.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
180
|
Cussenot O, Cancel-Tassin G, Comperat E, Benbouzid S, Lamb A. Total pelvic exenteration surgery for loco-regionally advanced prostate cancer, is it justifiable? BJU Int 2022; 130:582-585. [PMID: 36263588 DOI: 10.1111/bju.15841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Olivier Cussenot
- GRC 5 Predictive Onco-Urology, Sorbonne University, APHP, Paris, France
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- CeRePP, Paris, France
| | - Geraldine Cancel-Tassin
- GRC 5 Predictive Onco-Urology, Sorbonne University, APHP, Paris, France
- CeRePP, Paris, France
| | - Eva Comperat
- GRC 5 Predictive Onco-Urology, Sorbonne University, APHP, Paris, France
- CeRePP, Paris, France
| | - Sabrina Benbouzid
- GRC 5 Predictive Onco-Urology, Sorbonne University, APHP, Paris, France
| | - Alastair Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
181
|
French AFU Cancer Committee Guidelines - Update 2022-2024: prostate cancer - Diagnosis and management of localised disease. Prog Urol 2022; 32:1275-1372. [DOI: 10.1016/j.purol.2022.07.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022]
|
182
|
Pan-Cancer Analysis of the Prognostic and Immunotherapeutic Value of MITD1. Cells 2022; 11:cells11203308. [PMID: 36291174 PMCID: PMC9600621 DOI: 10.3390/cells11203308] [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: 07/30/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Microtubule-interacting and trafficking domain containing 1 (MITD1) is associated with abscission during cytokinesis. However, systematic investigation into its role in cancer is lacking. Therefore, we explored the pan-cancer role of MITD1 using multiple databases. Expression and clinical survival, immunological, and enrichment analyses were performed using R packages and online tools. For breast cancer, single-cell level analysis, immunochemistry, and in vitro experiments were performed to explore the mechanism of MITD1. A nomogram was established to predict the prognosis of patients with breast cancer and evaluate the immunotherapy biomarker based on two datasets. In some cancers, high MITD1 expression was associated with a more favorable prognosis. For instance, it inhibited tumor cell proliferation and migration in breast cancer. MITD1 may regulate cancer development by altering the tumor microenvironment, and MITD1 expression may predict the response to immune checkpoint blockade, platinum, and poly ADP-ribose polymerase inhibitor therapies. Our nomogram was used to determine the prognosis of patients with breast cancer. MITD1 can also predict the response to immunotherapy. Our first pan-cancer study of MITD1 has shown that it plays different roles in cancer development and therapy. In breast cancer, MITD1 inhibited cell proliferation and migration and serves as a new biomarker.
Collapse
|
183
|
Wang G, Liu Y, Liu S, Lin Y, Hu C. Oncolyic Virotherapy for Prostate Cancer: Lighting a Fire in Winter. Int J Mol Sci 2022; 23:12647. [PMID: 36293504 PMCID: PMC9603894 DOI: 10.3390/ijms232012647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/30/2022] [Accepted: 10/07/2022] [Indexed: 11/11/2022] Open
Abstract
As the most common cancer of the genitourinary system, prostate cancer (PCa) is a global men's health problem whose treatments are an urgent research issue. Treatment options for PCa include active surveillance (AS), surgery, endocrine therapy, chemotherapy, radiation therapy, immunotherapy, etc. However, as the cancer progresses, the effectiveness of treatment options gradually decreases, especially in metastatic castration-resistant prostate cancer (mCRPC), for which there are fewer therapeutic options and which have a shorter survival period and worse prognosis. For this reason, oncolytic viral therapy (PV), with its exceptional properties of selective tumor killing, relatively good safety in humans, and potential for transgenic delivery, has attracted increasing attention as a new form of anti-tumor strategy for PCa. There is growing evidence that OV not only kills tumor cells directly by lysis but can also activate anticancer immunity by acting on the tumor microenvironment (TME), thereby preventing tumor growth. In fact, evidence of the efficacy of this strategy has been observed since the late 19th century. However, subsequently, interest waned. The renewed interest in this therapy was due to advances in biotechnological methods and innovations at the end of the 20th century, which was also the beginning of PCa therapy with OV. Moreover, in combination with chemotherapy, radiotherapy, gene therapy or immunotherapy, OV viruses can have a wide range of applications and can provide an effective therapeutic result in the treatment of PCa.
Collapse
Affiliation(s)
- Gongwei Wang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Ying Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Shuoru Liu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yuan Lin
- Department of Pharmacology, Sun Yat-sen University, Guangzhou 528478, China
| | - Cheng Hu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
184
|
Hawkey NM, Broderick A, George DJ, Sartor O, Armstrong AJ. The Value of Phenotypic Precision Medicine in Prostate Cancer. Oncologist 2022; 28:93-104. [PMID: 36200788 PMCID: PMC9907055 DOI: 10.1093/oncolo/oyac198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/12/2022] [Indexed: 11/14/2022] Open
Abstract
Prostate cancer is the most common cancer among men and the second leading cause of cancer-related death. For patients who develop metastatic disease, tissue-based and circulating-tumor-based molecular and genomic biomarkers have emerged as a means of improving outcomes through the application of precision medicine. However, the benefit is limited to a minority of patients. An additional approach to further characterize the biology of advanced prostate cancer is through the use of phenotypic precision medicine, or the identification and targeting of phenotypic features of an individual patient's cancer. In this review article, we will discuss the background, potential clinical benefits, and limitations of genomic and phenotypic precision medicine in prostate cancer. We will also highlight how the emergence of image-based phenotypic medicine may lead to greater characterization of advanced prostate cancer disease burden and more individualized treatment approaches in patients.
Collapse
Affiliation(s)
- Nathan M Hawkey
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Amanda Broderick
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Daniel J George
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, USA,Duke Cancer Institute Center for Prostate and Urologic Cancer, Duke University, Durham, NC, USA
| | - Oliver Sartor
- Tulane Cancer Center, Division of Genitourinary Oncology, New Orleans, LA, USA
| | - Andrew J Armstrong
- Corresponding author: Andrew J. Armstrong, MD, ScM, FACP, Department of Medicine, Surgery, Pharmacology and Cancer Biology, Director of Research, the Duke Cancer Institute Center for Prostate and Urologic Cancers, Divisions of Medical Oncology and Urology, Duke University, DUMC Box 103861, Durham, NC 27710, USA;
| |
Collapse
|
185
|
Abstract
Gastric cancer (GC) is one of the most common lethal malignant neoplasms worldwide, with limited treatment options for both locally advanced and/or metastatic conditions, resulting in a dismal prognosis. Although the widely used morphological classifications may be helpful for endoscopic or surgical treatment choices, they are still insufficient to guide precise and/or personalized therapy for individual patients. Recent advances in genomic technology and high-throughput analysis may improve the understanding of molecular pathways associated with GC pathogenesis and aid in the classification of GC at the molecular level. Advances in next-generation sequencing have enabled the identification of several genetic alterations through single experiments. Thus, understanding the driver alterations involved in gastric carcinogenesis has become increasingly important because it can aid in the discovery of potential biomarkers and therapeutic targets. In this article, we review the molecular classifications of GC, focusing on The Cancer Genome Atlas (TCGA) classification. We further describe the currently available biomarker-targeted therapies and potential biomarker-guided therapies. This review will help clinicians by providing an inclusive understanding of the molecular pathology of GC and may assist in selecting the best treatment approaches for patients with GC.
Collapse
Affiliation(s)
- Moonsik Kim
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea.
| |
Collapse
|
186
|
Gu X, Wei S, Li Z, Xu H. Machine learning reveals two heterogeneous subtypes to assist immune therapy based on lipid metabolism in lung adenocarcinoma. Front Immunol 2022; 13:1022149. [PMID: 36238302 PMCID: PMC9551187 DOI: 10.3389/fimmu.2022.1022149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/12/2022] [Indexed: 12/24/2022] Open
Abstract
Background Lipid metabolism pivotally contributes to the incidence and development of lung adenocarcinoma (LUAD). The interaction of lipid metabolism and tumor microenvironment (TME) has become a new research direction. Methods Using the 1107 LUAD records from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, a comprehensive exploration was performed on the heterogeneous lipid metabolism subtypes based on lipid metabolism genes (LMGs) and immune-related genes (LRGs). The clinical significance, functional status, TME interaction and genomic changes of different subtypes were further studied. A new scoring system, lipid-immune score (LIS), was developed and validated. Results Two heterogeneous subtypes, which express more LMGs and show the characteristics of tumor metabolism and proliferation, are defined as lipid metabolism phenotypes. The prognosis of lipid metabolism phenotype is poor, and it is more common in patients with tumor progression. Expressing more IRGs, enrichment of immunoactive pathways and infiltration of effector immune cells are defined as immunoactive phenotypes. The immunoactive phenotype has a better prognosis and stronger anti-tumor immunity and is more sensitive to immunotherapy. In addition, KEAP1 is a driving mutant gene in the lipid metabolism subtype. Finally, LIS was developed and confirmed to be a robust predictor of overall survival (OS) and immunotherapy in LUAD patients. Conclusion Two heterogeneous subtypes of LUAD (lipid metabolism subtype and immune activity subtype) were identified to evaluate prognosis and immunotherapy sensitivity. Our research promotes the understanding of the interaction between lipid metabolism and TME and offers a novel direction for clinical management and precision therapy aimed to LUAD patients.
Collapse
Affiliation(s)
- Xuyu Gu
- School of Medicine, Southeast University, Nanjing, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhixin Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Huan Xu, ; Zhixin Li,
| | - Huan Xu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Huan Xu, ; Zhixin Li,
| |
Collapse
|
187
|
Yoshida T, Yaegashi H, Toriumi R, Kadomoto S, Iwamoto H, Izumi K, Kadono Y, Ikeda H, Mizokami A. Long response duration to pembrolizumab in metastatic, castration-resistant prostate cancer with microsatellite instability-high and neuroendocrine differentiation: A case report. Front Oncol 2022; 12:912490. [PMID: 36185251 PMCID: PMC9523122 DOI: 10.3389/fonc.2022.912490] [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: 04/04/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe detection of microsatellite instability in urologic cancers is rare, especially in metastatic, castration-resistant prostate cancer with neuroendocrine differentiation.Case presentationThis is a case of a 66-year-old Asian male patient with prostate adenocarcinoma who had metastases at initial presentation. Despite combined androgen deprivation therapy, his prostate-specific antigen (PSA) progressively increased, and prostate re-biopsy revealed small cell carcinoma. He was treated with platinum-based systemic chemotherapy, and his tumor markers, including PSA, remained negative; however, his local symptoms worsened. Subsequently, microsatellite instability-high was detected, and pembrolizumab was administered resulting in complete remission with the resolution of symptoms and continued therapeutic effect for more than 14 months.ConclusionMicrosatellite instability testing should be considered, despite its low detection rate, because the response to pembrolizumab in metastatic, castration-resistant prostate cancer with detectable microsatellite instability is associated with a prolonged duration of response.
Collapse
Affiliation(s)
- Tsukasa Yoshida
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Yaegashi
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
- *Correspondence: Hiroshi Yaegashi,
| | - Ren Toriumi
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Suguru Kadomoto
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Iwamoto
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Kouji Izumi
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yoshifumi Kadono
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hiroko Ikeda
- Department of Pathology, Kanazawa University Hospital, Kanazawa, Japan
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
188
|
Tang Q, Chen Y, Li X, Long S, Shi Y, Yu Y, Wu W, Han L, Wang S. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front Immunol 2022; 13:964442. [PMID: 36177034 PMCID: PMC9513184 DOI: 10.3389/fimmu.2022.964442] [Citation(s) in RCA: 241] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) is a checkpoint receptor expressed on the surface of various immune cells. PD-L1, the natural receptor for PD-1, is mainly expressed in tumor cells. Studies have indicated that PD-1 and PD-L1 are closely associated with the progression of human cancers and are promising biomarkers for cancer therapy. Moreover, the interaction of PD-1 and PD-L1 is one of the important mechanism by which human tumors generate immune escape. This article provides a review on the role of PD-L1/PD-1, mechanisms of immune response and resistance, as well as immune-related adverse events in the treatment of anti-PD-1/PD-L1 immunotherapy in human cancers. Moreover, we summarized a large number of clinical trials to successfully reveal that PD-1/PD-L1 Immune-checkpoint inhibitors have manifested promising therapeutic effects, which have been evaluated from different perspectives, including overall survival, objective effective rate and medium progression-free survival. Finally, we pointed out the current problems faced by PD-1/PD-L1 Immune-checkpoint inhibitors and its future prospects. Although PD-1/PD-L1 immune checkpoint inhibitors have been widely used in the treatment of human cancers, tough challenges still remain. Combination therapy and predictive models based on integrated biomarker determination theory may be the future directions for the application of PD-1/PD-L1 Immune-checkpoint inhibitors in treating human cancers.
Collapse
Affiliation(s)
- Qing Tang
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Clinical and Basic Research Team of Traditional Chinese Medicine (TCM) Prevention and Treatment of Non small cell lung cancer (NSCLC), Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun Chen
- Department of Organ Transplantation, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojuan Li
- Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shunqin Long
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Clinical and Basic Research Team of Traditional Chinese Medicine (TCM) Prevention and Treatment of Non small cell lung cancer (NSCLC), Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yao Shi
- Department of Cerebrovascular Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaya Yu
- Department of Oncology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Wanyin Wu
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Clinical and Basic Research Team of Traditional Chinese Medicine (TCM) Prevention and Treatment of Non small cell lung cancer (NSCLC), Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Wanyin Wu, ; Ling Han, ; Sumei Wang,
| | - Ling Han
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Wanyin Wu, ; Ling Han, ; Sumei Wang,
| | - Sumei Wang
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Clinical and Basic Research Team of Traditional Chinese Medicine (TCM) Prevention and Treatment of Non small cell lung cancer (NSCLC), Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Wanyin Wu, ; Ling Han, ; Sumei Wang,
| |
Collapse
|
189
|
Prostate Cancer Tumor Stroma: Responsibility in Tumor Biology, Diagnosis and Treatment. Cancers (Basel) 2022; 14:cancers14184412. [PMID: 36139572 PMCID: PMC9496870 DOI: 10.3390/cancers14184412] [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: 08/11/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The crosstalk between prostate stroma and its epithelium is essential to tissue homeostasis. Likewise, reciprocal signaling between tumor cells and the stromal compartment is required in tumor progression to facilitate or stimulate key processes such as cell proliferation and invasion. The aim of the present work was to review the current state of knowledge on the significance of tumor stroma in the genesis, progression and therapeutic response of prostate carcinoma. Additionally, we addressed the future therapeutic opportunities. Abstract Prostate cancer (PCa) is a common cancer among males globally, and its occurrence is growing worldwide. Clinical decisions about the combination of therapies are becoming highly relevant. However, this is a heterogeneous disease, ranging widely in prognosis. Therefore, new approaches are needed based on tumor biology, from which further prognostic assessments can be established and complementary strategies can be identified. The knowledge of both the morphological structure and functional biology of the PCa stroma compartment can provide new diagnostic, prognostic or therapeutic possibilities. In the present review, we analyzed the aspects related to the tumor stromal component (both acellular and cellular) in PCa, their influence on tumor behavior and the therapeutic response and their consideration as a new therapeutic target.
Collapse
|
190
|
Zhou M, Ko M, Hoge AC, Luu K, Liu Y, Russell ML, Hannon WW, Zhang Z, Carrot-Zhang J, Beroukhim R, Van Allen EM, Choudhury AD, Nelson PS, Freedman ML, Taplin ME, Meyerson M, Viswanathan SR, Ha G. Patterns of structural variation define prostate cancer across disease states. JCI Insight 2022; 7:e161370. [PMID: 35943799 PMCID: PMC9536266 DOI: 10.1172/jci.insight.161370] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/04/2022] [Indexed: 11/19/2022] Open
Abstract
The complex genomic landscape of prostate cancer evolves across disease states under therapeutic pressure directed toward inhibiting androgen receptor (AR) signaling. While significantly altered genes in prostate cancer have been extensively defined, there have been fewer systematic analyses of how structural variation shapes the genomic landscape of this disease across disease states. We uniformly characterized structural alterations across 531 localized and 143 metastatic prostate cancers profiled by whole genome sequencing, 125 metastatic samples of which were also profiled via whole transcriptome sequencing. We observed distinct significantly recurrent breakpoints in localized and metastatic castration-resistant prostate cancers (mCRPC), with pervasive alterations in noncoding regions flanking the AR, MYC, FOXA1, and LSAMP genes enriched in mCRPC and TMPRSS2-ERG rearrangements enriched in localized prostate cancer. We defined 9 subclasses of mCRPC based on signatures of structural variation, each associated with distinct genetic features and clinical outcomes. Our results comprehensively define patterns of structural variation in prostate cancer and identify clinically actionable subgroups based on whole genome profiling.
Collapse
Affiliation(s)
- Meng Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Minjeong Ko
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Anna C.H. Hoge
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kelsey Luu
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Yuzhen Liu
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Magdalena L. Russell
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - William W. Hannon
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Zhenwei Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Pathology, UMass Memorial Medical Center, Worcester, Massachusetts, USA
| | - Jian Carrot-Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Rameen Beroukhim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Atish D. Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Peter S. Nelson
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Matthew L. Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Srinivas R. Viswanathan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Gavin Ha
- Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
191
|
Wen R, Zhao H, Zhang D, Chiu CL, Brooks JD. Sialylated glycoproteins as biomarkers and drivers of progression in prostate cancer. Carbohydr Res 2022; 519:108598. [PMID: 35691122 DOI: 10.1016/j.carres.2022.108598] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/20/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023]
Abstract
Sialic acids have been implicated in cancer initiation, progression, and immune evasion in diverse human malignancies. Sialylation of terminal glycans on cell surface and secreted glycoproteins is a long-recognized feature of cancer cells. Recently, immune checkpoint inhibitor immunotherapy has tremendously improved the outcomes of patients with various cancers. However, available immunotherapy approaches have had limited efficacy in metastatic castration-resistant prostate cancer. Sialic acid modified glycoproteins in prostate cancers and their interaction with Siglec receptors on tumor infiltrating immune cells might underlie immunosuppressive signaling in prostate cancer. Here, we summarize the function of sialic acids and relevant glycosynthetic enzymes in cancer initiation and progression. We also discuss the possible uses of sialic acids as biomarkers in prostate cancer and the potential methods for targeting Siglec-sialic acid interactions for prostate cancer treatment.
Collapse
Affiliation(s)
- Ru Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
192
|
Targeted Approaches in Metastatic Castration-Resistant Prostate Cancer: Which Data? Cancers (Basel) 2022; 14:cancers14174189. [PMID: 36077726 PMCID: PMC9454420 DOI: 10.3390/cancers14174189] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Castration-resistant prostate cancer (CRPC) remains an incurable disease, but some promising innovative treatment options are under investigation. Recent developments in precision medicine have enabled the identification of new predictive biomarkers and potential targeted agents. The purpose of this review is to summarize and discuss new therapeutic approaches for metastatic CRPC (mCRPC), focusing on pathway description, prognostic and/or predictive role of recently discovered molecular alterations, investigation techniques, and potential clinical implications. Abstract Prostate cancer is the second most common diagnosed cancer and the fifth leading cause of cancer-related deaths in men worldwide. Despite significant advances in the management of castration-sensitive prostate cancer, the majority of patients develop a castration-resistant disease after a median duration of treatment of 18–48 months. The transition to a castrate resistance state could rely on alternative survival pathways, some related to androgen-independent mechanisms. Although several agents have been approved in this setting, metastatic castration-resistant prostate cancer (mCRPC) remains a lethal disease. Recent studies revealed some of the complex pathways underlying inherited and acquired mechanisms of resistance to available treatments. A better understanding of these pathways may lead to significant improvements in survival by providing innovative therapeutic targets. The present comprehensive review attempts to provide an overview of recent progress in novel targeted therapies and near-future directions.
Collapse
|
193
|
Schafer JM, Xiao T, Kwon H, Collier K, Chang Y, Abdel-Hafiz H, Bolyard C, Chung D, Yang Y, Sundi D, Ma Q, Theodorescu D, Li X, Li Z. Sex-biased adaptive immune regulation in cancer development and therapy. iScience 2022; 25:104717. [PMID: 35880048 PMCID: PMC9307950 DOI: 10.1016/j.isci.2022.104717] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The cancer research field is finally starting to unravel the mystery behind why males have a higher incidence and mortality rate than females for nearly all cancer types of the non-reproductive systems. Here, we explain how sex - specifically sex chromosomes and sex hormones - drives differential adaptive immunity across immune-related disease states including cancer, and why males are consequently more predisposed to tumor development. We highlight emerging data on the roles of cell-intrinsic androgen receptors in driving CD8+ T cell dysfunction or exhaustion in the tumor microenvironment and summarize ongoing clinical efforts to determine the impact of androgen blockade on cancer immunotherapy. Finally, we outline a framework for future research in cancer biology and immuno-oncology, underscoring the importance of a holistic research approach to understanding the mechanisms of sex dimorphisms in cancer, so sex will be considered as an imperative factor for guiding treatment decisions in the future.
Collapse
Affiliation(s)
- Johanna M. Schafer
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
| | - Tong Xiao
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
| | - Hyunwoo Kwon
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
- Medical Scientist Training Program, College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Katharine Collier
- Division of Medical Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
| | - Yuzhou Chang
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
- Department of Biomedical Informatics, the Ohio State University, Columbus, OH 43210, USA
| | - Hany Abdel-Hafiz
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Chelsea Bolyard
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, the Ohio State University, Columbus, OH 43210, USA
| | - Yuanquan Yang
- Division of Medical Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
| | - Debasish Sundi
- Department of Urology, the Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, the Ohio State University, Columbus, OH 43210, USA
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xue Li
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center – the James, Columbus, OH 43210, USA
| |
Collapse
|
194
|
Wang I, Song L, Wang BY, Rezazadeh Kalebasty A, Uchio E, Zi X. Prostate cancer immunotherapy: a review of recent advancements with novel treatment methods and efficacy. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2022; 10:210-233. [PMID: 36051616 PMCID: PMC9428569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
Immunotherapy remains to be an appealing treatment option for prostate cancer with some documented promise. Prostate cancer is traditionally considered as an immunologically "cold" tumor with low tumor mutation burden, low expression of PD-L1, sparse T-cell infiltration, and a immunosuppressive tumor microenvironment (TME). Sipuleucel-T (Provenge) is the first FDA approved immunotherapeutic agent for the treatment of asymptomatic or minimally symptomatic metastatic castrate resistant prostate cancer (mCRPC); demonstrating a benefit in overall survival. However various clinical trials by immune checkpoint inhibitors (ICIs) and their combinations with other drugs have shown limited responses in mCRPC. Up to now, only a small subset of patients with mismatch repair deficiency/microsatellite instability high and CDK12 mutations can clinically benefit from ICIs and/or their combinations with other agents, such as DNA damage agents. The existence of a large heterogeneity in genomic alterations and a complex TME in prostate cancer suggests the need for identifying new immunotherapeutic targets. As well as designing personalized immunotherapy strategies based on patient-specific molecular signatures. There is also a need to adjust strategies to overcome histologic barriers such as tissue hypoxia and dense stroma. The racial differences of immunological responses between men of diverse ethnicities also merit further investigation to improve the efficacy of immunotherapy and better patient selection in prostate cancer.
Collapse
Affiliation(s)
- Ian Wang
- Hofstra UniversityHempstead, NY, USA
| | - Liankun Song
- Department of Urology, University of CaliforniaIrvine, Orange, CA 92868, USA
| | - Beverly Y Wang
- Department of Pathology, University of CaliforniaIrvine, Orange, CA 92868, USA
| | | | - Edward Uchio
- Department of Medicine, University of CaliforniaIrvine, Orange, CA 92868, USA
- Chao Family Comprehensive Cancer Center, University of CaliforniaOrange, CA 92868, USA
| | - Xiaolin Zi
- Department of Urology, University of CaliforniaIrvine, Orange, CA 92868, USA
- Department of Medicine, University of CaliforniaIrvine, Orange, CA 92868, USA
- Chao Family Comprehensive Cancer Center, University of CaliforniaOrange, CA 92868, USA
- Department of Pharmaceutical Sciences, University of CaliforniaIrvine, Irvine, CA 92617, USA
| |
Collapse
|
195
|
Wang XH, Wang ZQ, Mu ZY, Zhu LP, Zhong CF, Guo S. The efficacy and safety of immune checkpoint inhibitors in metastatic castration-resistant prostate cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e29715. [PMID: 35945714 PMCID: PMC9351907 DOI: 10.1097/md.0000000000029715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND We aim to assess the efficacy and safety profiles of immune checkpoint inhibitors in patients with metastatic castration-resistant prostate cancer using a meta-analysis. METHODS We extracted and examined data from phase I, II and III clinical trials from PubMed, Embase, Web of Science, and Cochrane Library, which included patients with metastatic castration-resistant prostate cancer who were treated with immune checkpoint inhibitors. We performed a meta-analysis to investigate several indexes of efficacy and safety, including the objective response rate, 1-year overall survival (OS) rate, prostate-specific antigen response rate, and adverse event rate of immune checkpoint inhibitors. The material data were calculated and pooled using The R Project for Statistical Computing and STATA 12.0 software. RESULTS We identified 12 clinical trials in our study. We assessed the pooled frequencies of all-grade AEs and grade ≥ 3 AEs first and showed 0.82 (95% CI: 0.74-0.91, I2 = 94%, P < .01) and 0.42 (95% CI: 0.33-0.54, I2 = 96%, P < .01), respectively. The objective response rate was 0.10 (95% CI: 0.04-0.19, I2 = 70%, P < .01), and the 1-year OS and prostate-specific antigen response rate were 0.55 (95% CI: 0.45-0.67, I2 = 93%, P < .01) and 0.18 (95% CI: 0.16-0.20, I2 = 43%, P = .03), respectively. CONCLUSION The immune checkpoint inhibitors therapy was well tolerated and showed potential to improve tumor responses in patients with metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Xing-Hui Wang
- Department of Pathology, People’s hospital of Shouguang, Shandong, China
| | - Zhi-Qiang Wang
- Department of Urology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong, China
| | - Zhen-Yu Mu
- Department of Neurology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong, China
| | - Li-Ping Zhu
- Department of Oncology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong, China
| | - Chong-Fu Zhong
- Department of Andrology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, Shandong, China
| | - Shanchun Guo
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA
- *Correspondence: Shanchun Guo, PhD, RCMI Cancer Research Center, Xavier University of Louisiana, 1 Drexel Dr, New Orleans, LA 70125 (e-mail: )
| |
Collapse
|
196
|
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
|
197
|
Kageyama T, Soga N, Sekito S, Kato S, Ogura Y, Kojima T, Kanai M, Inoue T. Dramatic response to pembrolizumab after pseudoprogression in a patient with advanced metastatic castration‐resistant prostate cancer. IJU Case Rep 2022; 5:442-445. [DOI: 10.1002/iju5.12508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Takumi Kageyama
- Department of Nephro‐Urologic Surgery and Andrology Mie University Graduate School of Medicine Tsu Mie Japan
- Department of Urology Aichi Cancer Center Hospital Nagoya Aichi Japan
- Department of Urology Suzuka General Hospital Suzuka Mie Japan
| | - Norihito Soga
- Department of Urology Aichi Cancer Center Hospital Nagoya Aichi Japan
| | - Sho Sekito
- Department of Urology Aichi Cancer Center Hospital Nagoya Aichi Japan
| | - Seiichi Kato
- Department of Pathology and Molecular Diagnostics Aichi Cancer Center Hospital Nagoya Aichi Japan
| | - Yuji Ogura
- Department of Urology Aichi Cancer Center Hospital Nagoya Aichi Japan
| | - Takahiro Kojima
- Department of Urology Aichi Cancer Center Hospital Nagoya Aichi Japan
| | - Masahiro Kanai
- Department of Urology Suzuka General Hospital Suzuka Mie Japan
| | - Takahiro Inoue
- Department of Nephro‐Urologic Surgery and Andrology Mie University Graduate School of Medicine Tsu Mie Japan
| |
Collapse
|
198
|
The Role of Histology-Agnostic Drugs in the Treatment of Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2022; 23:ijms23158535. [PMID: 35955671 PMCID: PMC9369092 DOI: 10.3390/ijms23158535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023] Open
Abstract
Precision medicine has opened up a new era in the development of anti-cancer agents that is focused on identifying biomarkers predictive of treatment response regardless of tumor histology. Since 2017, the Food and Drug Administration has approved six drugs with histology-agnostic indications: pembrolizumab (both for tumors with the mismatch-repair deficiency (dMMR)/high microsatellite instability (MSI-H) phenotype and for those with the high tumor mutational burden (TMB-H) phenotype), dostarlimab (for dMMR tumors), larotrectinib and entrectinib (for tumors harboring neurotrophic tyrosine receptor kinase (NTRK) fusions), and the combination of dabrafenib plus trametinib (for BRAF V600E-mutated tumors). The genomic alterations targeted by these antineoplastic agents are rare in metastatic castration-resistant prostate cancer (mCRPC). Furthermore, only a small number of mCRPC patients were enrolled in the clinical trials that led to the approval of the above-mentioned drugs. Therefore, we critically reviewed the literature on the efficacy of histology-agnostic drugs in mCRPC patients. Although the available evidence derives from retrospective studies and case reports, our results confirmed the efficacy of pembrolizumab in dMMR/MSI-H mCRPC. In contrast, few data are available for dostarlimab, larotrectinib, entrectinib, and dabrafenib-trametinib in this subset of patients. Large, multi-institutional registries aimed at collecting real-world data are needed to better comprehend the role of tissue-agnostic drugs in mCRPC patients.
Collapse
|
199
|
Graham LS, Schweizer MT. Mismatch repair deficiency and clinical implications in prostate cancer. Prostate 2022; 82 Suppl 1:S37-S44. [PMID: 35358351 DOI: 10.1002/pros.24343] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 12/11/2022]
Abstract
Despite recent therapeutic advances, castration-resistant prostate cancer (CRPC) remains a lethal disease and novel therapies are needed. Precision oncology provides an avenue for developing effective tailored approaches for treating malignancies based on a tumor's molecular profile. Indeed, the presence of mismatch repair deficiency (MMRd) has proven to be an important predictive biomarker for response to immune checkpoint blockade across multiple tumor types, including prostate cancer, and represents a major precision oncology success story. The mismatch repair (MMR) system is integral to maintaining genomic fidelity during cellular replication. Cancers with deficiencies in this system accumulate high numbers of mutations and express many neoantigens that may be recognized by the immune system. The checkpoint inhibitor pembrolizumab has recently been approved for all cancers that are MMR deficient, and several retrospective series have specifically shown that pembrolizumab is effective in MMRd prostate cancer. Although the prevalence of MMRd in CRPC is low (approximately 3%-5% of cases), this is an important subset of men that require a unique therapeutic approach. This review will focus on MMRd in prostate cancer, highlighting the clinical implications, role of immunotherapy, and areas of future research.
Collapse
Affiliation(s)
- Laura S Graham
- Division of Medical Oncology, University of Colorado, Aurora, Colorado, USA
| | - Michael T Schweizer
- Division of Medical Oncology, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
200
|
Peiffer LB, White JR, Jones CB, Slottke RE, Ernst SE, Moran AE, Graff JN, Sfanos KS. Composition of gastrointestinal microbiota in association with treatment response in individuals with metastatic castrate resistant prostate cancer progressing on enzalutamide and initiating treatment with anti-PD-1 (pembrolizumab). Neoplasia 2022; 32:100822. [PMID: 35908379 PMCID: PMC9340532 DOI: 10.1016/j.neo.2022.100822] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/10/2022] [Accepted: 07/11/2022] [Indexed: 01/04/2023]
Abstract
Recent studies in cancer patients and animal models demonstrate that intestinal microbiota influence the therapeutic efficacy of cancer treatments, including immune checkpoint inhibition. However, no studies to-date have investigated relationships between gastrointestinal microbiota composition and response to checkpoint inhibition in advanced metastatic castrate resistant prostate cancer (mCRPC). We performed 16S rRNA gene sequencing of fecal DNA from 23 individuals with mCRPC progressing on enzalutamide and just prior to treatment with anti-PD-1 (pembrolizumab) to determine whether certain features of the microbiome are associated with treatment response (defined as serum PSA decrease >50% at any time on treatment or radiographic response per RECIST V.1.1). Global bacterial composition was similar between responders and non-responders, as assessed by multiple alpha and beta diversity metrics. However, certain bacterial taxa identified by sequencing across multiple 16S rRNA hypervariable regions were consistently associated with response, including the archetypal oral bacterium Streptococcus salivarius. Quantitative PCR (qPCR) of DNA extracts from fecal samples confirmed increased Streptococcus salivarius fecal levels in responders, whereas qPCR of oral swish DNA extracts showed no relationship between oral Streptococcus salivarius levels and response status. Contrary to previous reports in other cancer types, Akkermansia muciniphila levels were reduced in responder samples as assessed by both 16S rRNA sequencing and qPCR. We further analyzed our data in the context of a previously published “integrated index” describing bacteria associated with response and non-response to checkpoint inhibition. We found that the index was not reflective of response status in our cohort. Lastly, we demonstrate little change in the microbiome over time, and with pembrolizumab treatment. Our results suggest that the association between fecal microbiota and treatment response to immunotherapy may be unique to cancer type and/or previous treatment history.
Collapse
Affiliation(s)
- Lauren B Peiffer
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Carli B Jones
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rachel E Slottke
- Division of Hematology & Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Sarah E Ernst
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy E Moran
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Julie N Graff
- Division of Hematology & Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA; Portland VA Health Care System, Portland, OR, USA.
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Departments of Oncology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|