1
|
Luo M, He N, Xu Q, Wen Z, Wang Z, Zhao J, Liu Y. Roles of prostaglandins in immunosuppression. Clin Immunol 2024; 265:110298. [PMID: 38909972 DOI: 10.1016/j.clim.2024.110298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
Prostaglandins (PGs) play a crucial and multifaceted role in various physiological processes such as intercellular signaling, inflammation regulation, neurotransmission, vasodilation, vasoconstriction, and reproductive functions. The diversity and biological significance of these effects are contingent upon the specific types or subtypes of PGs, with each PG playing a crucial role in distinct physiological and pathological processes. Particularly within the immune system, PGs are essential in modulating the function of immune cells and the magnitude and orientation of immune responses. Hence, a comprehensive comprehension of the functions PG signaling pathways in immunosuppressive regulation holds substantial clinical relevance for disease prevention and treatment strategies. The manuscript provides a review of recent developments in PG signaling in immunosuppressive regulation. Furthermore, the potential clinical applications of PGs in immunosuppression are also discussed. While research into the immunosuppressive effects of PGs required further exploration, targeted therapies against their immunosuppressive pathways might open new avenues for disease prevention and treatment.
Collapse
Affiliation(s)
- Minjie Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Zhongchi Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Ziqin Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| |
Collapse
|
2
|
Efstathiou JA, Morgans AK, Bland CS, Shore ND. Novel hormone therapy and coordination of care in high-risk biochemically recurrent prostate cancer. Cancer Treat Rev 2024; 122:102630. [PMID: 38035646 DOI: 10.1016/j.ctrv.2023.102630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/25/2023] [Indexed: 12/02/2023]
Abstract
Biochemical recurrence (BCR) occurs in 20-50% of patients with prostate cancer (PCa) undergoing primary definitive treatment. Patients with high-risk BCR have an increased risk of metastatic progression and subsequent PCa-specific mortality, and thus could benefit from treatment intensification. Given the increasing complexity of diagnostic and therapeutic modalities, multidisciplinary care (MDC) can play a crucial role in the individualized management of this patient population. This review explores the role for MDC when evaluating the clinical evidence for the evolving definition of high-risk BCR and the emerging therapeutic strategies, especially with novel hormone therapies (NHTs), for patients with either high-risk BCR or oligometastatic PCa. Clinical studies have used different characteristics to define high-risk BCR and there is no consensus regarding the definition of high-risk BCR nor for management strategies. Next-generation imaging and multigene panels offer potential enhanced patient identification and precision-based decision-making, respectively. Treatment intensification with NHTs, either alone or combined with radiotherapy or metastasis-directed therapy, has been promising in clinical trials in patients with high-risk BCR or oligometastases. As novel risk-stratification and treatment options as well as evidence-based literature evolve, it is important to involve a multidisciplinary team to identify patients with high-risk features at an earlier stage, and make informed decisions on the treatments that could optimize their care and long-term outcomes. Nevertheless, MDC data are scarce in the BCR or oligometastatic setting. Efforts to integrate MDC into the standard management of this patient population are needed, and will likely improve outcomes across this heterogeneous PCa patient population.
Collapse
Affiliation(s)
- Jason A Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| | - Alicia K Morgans
- Dana-Farber Cancer Institute, 850 Brookline Ave, Dana 09-930, Boston, MA 02215, USA.
| | - Christopher S Bland
- US Oncology Medical Affairs, Pfizer Inc., 66 Hudson Boulevard, Hudson Yards, Manhattan, New York, NY 10001, USA.
| | - Neal D Shore
- Carolina Urologic Research Center, GenesisCare US, 823 82nd Pkwy, Myrtle Beach, SC, USA.
| |
Collapse
|
3
|
Lee DH, Chung SW, Lee JH, Kim HY, Chung GE, Kim MS, Yang BR, Nam JY, Lee YB, Kim YJ, Yoon JH. Association of Chronic Hepatitis B Infection and Antiviral Treatment With the Development of the Extrahepatic Malignancies: A Nationwide Cohort Study. J Clin Oncol 2022; 40:3394-3405. [PMID: 35561284 DOI: 10.1200/jco.21.01285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Epidemiologic studies suggest that chronic hepatitis B (CHB) is a risk factor for various primary extrahepatic malignancies. Our aim was to evaluate the associations of CHB and nucleos(t)ide analog (NA) treatment with the risk of the development of extrahepatic malignancies. PATIENTS AND METHODS We conducted an 18-month landmark analysis using nationwide claims data from the National Health Insurance Service of South Korea. Patients newly diagnosed with CHB in 2012-2014 (n = 90,944) and matched-controls (n = 685,436) were included. Patients with CHB were further classified as the NA-treated (CHB+/NA+, n = 6,539) or the NA-untreated (CHB+/NA-, n = 84,405) group. Inverse probability of treatment weighting analysis was applied to balance the treatment groups. Time-varying Cox analysis was performed to evaluate time-varying effect of NA treatment. The primary outcome was the development of any primary extrahepatic malignancy. Development of intrahepatic malignancy and death were considered as competing events. RESULTS During the study period (median = 47.4 months), 30,413 patients (3.9%) developed any extrahepatic malignancy. The CHB+/NA- group had a higher overall risk of extrahepatic malignancy than the CHB+/NA+ group (adjusted subdistribution hazard ratio [aSHR] = 1.28; 95% CI, 1.12 to 1.45; P < .001) or controls (aSHR = 1.22; 95% CI, 1.18 to 1.26; P < .001). There was no difference in the risk of extrahepatic malignancy between the CHB+/NA+ group and the controls (CHB+/NA+ v control: aSHR = 0.96; 95% CI, 0.84 to 1.08; P = .48). In time-varying Cox analysis, the CHB+/NA- patients were associated with a higher risk of extrahepatic malignancy than the CHB+/NA+ patients (aSHR = 1.37; 95% CI, 1.23 to 1.52; P < .001). CONCLUSION Patients with CHB have an elevated risk of developing primary extrahepatic malignancy. Long-term NA treatment was associated with a lower risk of extrahepatic malignancy development among patients with CHB.
Collapse
Affiliation(s)
- Dong Hyeon Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Sung Won Chung
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeong-Hoon Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hwi Young Kim
- Department of Internal Medicine, Ewha Womans University Medical Center, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Goh Eun Chung
- Department of Internal Medicine, Healthcare System Gangnam Center Seoul National University Hospital, Seoul, South Korea
| | - Mi-Sook Kim
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, South Korea
| | - Bo Ram Yang
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, South Korea.,College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | - Joon Yeul Nam
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yun Bin Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon Jun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jung-Hwan Yoon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
4
|
van de Merbel AF, van der Horst G, van der Mark MH, Bots STF, van den Wollenberg DJM, de Ridder CMA, Stuurman D, Aalders T, Erkens-Schulz S, van Montfoort N, Karthaus WR, Mehra N, Smits M, Schalken JA, van Weerden WM, Hoeben RC, van der Pluijm G. Reovirus mutant jin-3 exhibits lytic and immune-stimulatory effects in preclinical human prostate cancer models. Cancer Gene Ther 2022; 29:793-802. [PMID: 34135475 PMCID: PMC9209329 DOI: 10.1038/s41417-021-00360-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/08/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Treatment of castration-resistant prostate cancer remains a challenging clinical problem. Despite the promising effects of immunotherapy in other solid cancers, prostate cancer has remained largely unresponsive. Oncolytic viruses represent a promising therapeutic avenue, as oncolytic virus treatment combines tumour cell lysis with activation of the immune system and mounting of effective anti-tumour responses. Mammalian Orthoreoviruses are non-pathogenic human viruses with a preference of lytic replication in human tumour cells. In this study, we evaluated the oncolytic efficacy of the bioselected oncolytic reovirus mutant jin-3 in multiple human prostate cancer models. The jin-3 reovirus displayed efficient infection, replication, and anti-cancer responses in 2D and 3D prostate cancer models, as well as in ex vivo cultured human tumour slices. In addition, the jin-3 reovirus markedly reduced the viability and growth of human cancer cell lines and patient-derived xenografts. The infection induced the expression of mediators of immunogenic cell death, interferon-stimulated genes, and inflammatory cytokines. Taken together, our data demonstrate that the reovirus mutant jin-3 displays tumour tropism, and induces potent oncolytic and immunomodulatory responses in human prostate cancer models. Therefore, jin-3 reovirus represents an attractive candidate for further development as oncolytic agent for treatment of patients with aggressive localised or advanced prostate cancer.
Collapse
Affiliation(s)
- Arjanneke F. van de Merbel
- grid.10419.3d0000000089452978Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| | - Geertje van der Horst
- grid.10419.3d0000000089452978Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maaike H. van der Mark
- grid.10419.3d0000000089452978Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| | - Selas T. F. Bots
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Diana J. M. van den Wollenberg
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Corrina M. A. de Ridder
- grid.5645.2000000040459992XDepartment of Experimental Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Debra Stuurman
- grid.5645.2000000040459992XDepartment of Experimental Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tilly Aalders
- grid.10417.330000 0004 0444 9382Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sigrun Erkens-Schulz
- grid.5645.2000000040459992XDepartment of Experimental Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nadine van Montfoort
- grid.10419.3d0000000089452978Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter R. Karthaus
- grid.51462.340000 0001 2171 9952Human Pathology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Niven Mehra
- grid.10417.330000 0004 0444 9382Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Minke Smits
- grid.10417.330000 0004 0444 9382Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jack A. Schalken
- grid.10417.330000 0004 0444 9382Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wytske M. van Weerden
- grid.5645.2000000040459992XDepartment of Experimental Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Rob C. Hoeben
- grid.10419.3d0000000089452978Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gabri van der Pluijm
- grid.10419.3d0000000089452978Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Bilusic M, Einstein DJ, Karzai FH, Dahut WL, Gulley JL, Aragon-Ching JB, Madan RA. The Potential Role for Immunotherapy in Biochemically Recurrent Prostate Cancer. Urol Clin North Am 2020; 47:457-467. [PMID: 33008496 PMCID: PMC8177734 DOI: 10.1016/j.ucl.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Biochemically recurrent prostate cancer represents a stage of prostate cancer where conventional (continued on next page) computed tomography and technetium Tc 99m bone scan imaging are unable to detect disease after curative intervention despite rising prostate-specific antigen. There is no clear standard of care and no systemic therapy has been shown to improve survival. Immunotherapy-based treatments potentially are attractive options relative to androgen deprivation therapy due to the generally more favorable side-effect profile. Biochemically recurrent prostate cancer patients have a low tumor burden and likely lymph node-based disease, which may make them more likely to respond to immunotherapy.
Collapse
Affiliation(s)
- Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, 13n240b, Bethesda, MD 20892, USA
| | - David J Einstein
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Fatima H Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, 13n240b, Bethesda, MD 20892, USA
| | - William L Dahut
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, 13n240b, Bethesda, MD 20892, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, 13n240b, Bethesda, MD 20892, USA
| | | | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Drive, 13n240b, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Papaevangelou E, Smolarek D, Smith RA, Dasgupta P, Galustian C. Targeting Prostate Cancer Using Intratumoral Cytotopically Modified Interleukin-15 Immunotherapy in a Syngeneic Murine Model. Immunotargets Ther 2020; 9:115-130. [PMID: 32802803 PMCID: PMC7394845 DOI: 10.2147/itt.s257443] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/26/2020] [Indexed: 01/05/2023] Open
Abstract
Background The prostate cancer microenvironment is highly immunosuppressive; immune cells stimulated in the periphery by systemic immunotherapies will be rendered inactive once entering this environment. Immunotherapies for prostate cancer need to break this immune tolerance. We have previously identified interleukin-15 (IL-15) as the only cytokine tested that activates and expands immune cells in the presence of prostate cancer cells. In the current study, we aimed to identify a method of boosting the efficacy of IL-15 in prostate cancer. Methods We engineered, by conjugation to a myristoylated peptide, a membrane-localising form of IL-15 (cyto-IL-15) and the checkpoint inhibitor antibodies cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death ligand 1 (PD-L1) (cyto-abs) to enable them to bind to cell surfaces by non-specific anchoring to the phospholipid bilayer. The efficacy of these agents was investigated by intratumoral administration either alone (cyto-IL-15 or cyto-abs) or in combination (cyto-combo) in subcutaneous TRAMP-C2 prostate tumors in C57BL/6J mice and compared with their non-modified equivalents in vivo. Following the survival endpoint, histological analyses and RNA sequencing were performed on the tumors. Results Intratumoral injection of cyto-IL-15 or cyto-combo delayed tumor growth by 50% and increased median survival to 28 and 25 days, respectively, compared with vehicle (17 days), whereas non-modified IL-15 or antibodies alone had no significant effects on tumor growth or survival. Histological analysis showed that cyto-IL-15 and cyto-combo increased necrosis and infiltration of natural killer (NK) cells and CD8 T cells in the tumors compared with vehicle and non-modified agents. Overall, the efficacy of cyto-combo was not superior to that of cyto-IL-15 alone. Conclusion We have demonstrated that intratumoral injection of cyto-IL-15 leads to prostate cancer growth delay, induces tumor necrosis and increases survival. Hence, cytotopic modification in combination with intratumoral injection appears to be a promising novel approach for prostate cancer immunotherapy.
Collapse
Affiliation(s)
- Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Dorota Smolarek
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Richard A Smith
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK.,Urology Centre, Guy's Hospital, London, UK
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
7
|
Owen KL, Gearing LJ, Zanker DJ, Brockwell NK, Khoo WH, Roden DL, Cmero M, Mangiola S, Hong MK, Spurling AJ, McDonald M, Chan C, Pasam A, Lyons RJ, Duivenvoorden HM, Ryan A, Butler LM, Mariadason JM, Giang Phan T, Hayes VM, Sandhu S, Swarbrick A, Corcoran NM, Hertzog PJ, Croucher PI, Hovens C, Parker BS. Prostate cancer cell-intrinsic interferon signaling regulates dormancy and metastatic outgrowth in bone. EMBO Rep 2020; 21:e50162. [PMID: 32314873 PMCID: PMC7271653 DOI: 10.15252/embr.202050162] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/15/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
The latency associated with bone metastasis emergence in castrate-resistant prostate cancer is attributed to dormancy, a state in which cancer cells persist prior to overt lesion formation. Using single-cell transcriptomics and ex vivo profiling, we have uncovered the critical role of tumor-intrinsic immune signaling in the retention of cancer cell dormancy. We demonstrate that loss of tumor-intrinsic type I IFN occurs in proliferating prostate cancer cells in bone. This loss suppresses tumor immunogenicity and therapeutic response and promotes bone cell activation to drive cancer progression. Restoration of tumor-intrinsic IFN signaling by HDAC inhibition increased tumor cell visibility, promoted long-term antitumor immunity, and blocked cancer growth in bone. Key findings were validated in patients, including loss of tumor-intrinsic IFN signaling and immunogenicity in bone metastases compared to primary tumors. Data herein provide a rationale as to why current immunotherapeutics fail in bone-metastatic prostate cancer, and provide a new therapeutic strategy to overcome the inefficacy of immune-based therapies in solid cancers.
Collapse
|
8
|
Liu Y, Mikrani R, Xie D, Wazir J, Shrestha S, Ullah R, Baig MMFA, Ahmed A, Srivastava PK, Thapa KB, Zhou X. Chronic prostatitis/chronic pelvic pain syndrome and prostate cancer: study of immune cells and cytokines. Fundam Clin Pharmacol 2019; 34:160-172. [DOI: 10.1111/fcp.12517] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/30/2019] [Accepted: 10/22/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Yuqian Liu
- Department of Clinical Pharmacy School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing Jiangsu 211198 China
| | - Reyaj Mikrani
- Department of Clinical Pharmacy School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing Jiangsu 211198 China
| | - Dianyou Xie
- Department of Clinical Pharmacy School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing Jiangsu 211198 China
| | - Junaid Wazir
- Department of Clinical Pharmacy School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing Jiangsu 211198 China
| | - Sajan Shrestha
- Department of Clinical Pharmacy School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing Jiangsu 211198 China
| | - Rahat Ullah
- Department of Clinical Pharmacy School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing Jiangsu 211198 China
| | - Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Sciences School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 China
| | - Abrar Ahmed
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 China
| | | | - Kedar Bahadur Thapa
- Institute of Advanced Materials Nanjing Tech University Nanjing 211816 China
| | - Xiaohui Zhou
- Department of Clinical Pharmacy School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing Jiangsu 211198 China
- Department of Surgery Zhongda Hospital Affiliated to Southeast University Nanjing Jiangsu 210017 China
- Department of Surgery Nanjing Shuiximen Hospital Nanjing Jiangsu 210017 China
| |
Collapse
|
9
|
Becher E, Wang A, Lepor H. Prostate Cancer Screening and Management in Solid Organ Transplant Candidates and Recipients. Rev Urol 2019; 21:85-92. [PMID: 31768135 PMCID: PMC6864909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The number of solid organ transplantations is increasing worldwide. Major medical advances have allowed for incremented survival in this population, which, because approximately 50% of recipients are over age 50 years, makes for an increasingly older population of transplant survivors. This article discusses controversies and current guidelines related to prostate cancer (PCa) screening, detection, and treatment for men in the general population. The relevant literature is reviewed in order to provide insights on how to optimize PCa screening, detection, and treatment pre- and post-solid organ transplantation. There is compelling evidence that immunosuppression does not increase the risk for the development or progression of PCa following solid organ transplantation. Therefore, PCa screening, detection, or treatment should not be influenced by the impact of immunosuppression on the biology of the disease. Prostate-specific antigen (PSA) appears to be as reliable for PCa screening of transplant candidates and recipients as it is for the general population. There is no consensus on how or when it should be implemented. Evidence is also equivocal as to the suggested waiting time between treatment and transplantation. Surgery and radiation therapy appear to be safe and provide good outcomes for managing PCa in solid organ transplant candidates and recipients. However, certain precautions should be taken with this vulnerable population, especially for kidney transplant patients given the pelvic location of the renal graft. Partial gland ablation of PCa should be considered in appropriate candidates.
Collapse
Affiliation(s)
| | - Alex Wang
- Department of Urology, NYU Langone Health New York, NY
| | - Herbert Lepor
- Department of Urology, NYU Langone Health New York, NY
| |
Collapse
|
10
|
Krzastek SC, Goliadze E, Zhou S, Petrossian A, Youniss F, Sundaresan G, Wang L, Zweit J, Guruli G. Dendritic cell trafficking in tumor-bearing mice. Cancer Immunol Immunother 2018; 67:1939-1947. [PMID: 29943070 PMCID: PMC11028156 DOI: 10.1007/s00262-018-2187-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/13/2018] [Indexed: 12/16/2022]
Abstract
Prostate cancer is one of the leading causes of cancer deaths, with no curative treatments once it spreads. Alternative therapies, including immunotherapy, have shown limited efficacy. Dendritic cells (DC) have been widely used in the treatment of various malignancies. DC capture antigens and move to the lymphoid organs where they prime naive T cells. Interaction between DC and T cells are most active in lymph nodes and suppression of DC trafficking to lymph nodes impairs the immune response. In this work, we aimed to study trafficking of DC in vivo via various routes of delivery, to optimize the effectiveness of DC-based therapy. A DC labeling system was developed using 1,1'-dioctadecyltetramethyl indotricarbocyanine Iodine for in vivo fluorescent imaging. DC harvested from C57B/6 mice were matured, labeled, and injected intravenously, subcutaneously, or intratumorally, with or without antigen loading with whole tumor lysate, into C57B/6 mice inoculated with RM-1 murine prostate tumor cells. Signal intensity was measured in vivo and ex vivo. Signal intensity at the tumor site increased over time, suggesting trafficking of DC to the tumor with all modes of injection. Subcutaneous injection showed preferential trafficking to lymph nodes and tumor. Intravenous injection showed trafficking to lungs, intestines, and spleen. Subcutaneous injection of DC pulsed with whole tumor lysate resulted in the highest increase in signal intensity at the tumor site and lymph nodes, suggesting subcutaneous injection of primed DC leads to highest preferential trafficking of DC to the immunocompetent organs.
Collapse
Affiliation(s)
- Sarah C Krzastek
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Ekaterine Goliadze
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Shaoqing Zhou
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Albert Petrossian
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Fatma Youniss
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Gobalakrishnan Sundaresan
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Li Wang
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Jamal Zweit
- Department of Radiology, Center for Molecular Imaging, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Georgi Guruli
- Division of Urology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
11
|
Lacher MD, Bauer G, Fury B, Graeve S, Fledderman EL, Petrie TD, Coleal-Bergum DP, Hackett T, Perotti NH, Kong YY, Kwok WW, Wagner JP, Wiseman CL, Williams WV. SV-BR-1-GM, a Clinically Effective GM-CSF-Secreting Breast Cancer Cell Line, Expresses an Immune Signature and Directly Activates CD4 + T Lymphocytes. Front Immunol 2018; 9:776. [PMID: 29867922 PMCID: PMC5962696 DOI: 10.3389/fimmu.2018.00776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Targeted cancer immunotherapy with irradiated, granulocyte–macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic cancer cell lines has been an effective approach to reduce tumor burden in several patients. It is generally assumed that to be effective, these cell lines need to express immunogenic antigens coexpressed in patient tumor cells, and antigen-presenting cells need to take up such antigens then present them to patient T cells. We have previously reported that, in a phase I pilot study (ClinicalTrials.gov NCT00095862), a subject with stage IV breast cancer experienced substantial regression of breast, lung, and brain lesions following inoculation with clinical formulations of SV-BR-1-GM, a GM-CSF-secreting breast tumor cell line. To identify diagnostic features permitting the prospective identification of patients likely to benefit from SV-BR-1-GM, we conducted a molecular analysis of the SV-BR-1-GM cell line and of patient-derived blood, as well as a tumor specimen. Compared to normal human breast cells, SV-BR-1-GM cells overexpress genes encoding tumor-associated antigens (TAAs) such as PRAME, a cancer/testis antigen. Curiously, despite its presumptive breast epithelial origin, the cell line expresses major histocompatibility complex (MHC) class II genes (HLA-DRA, HLA-DRB3, HLA-DMA, HLA-DMB), in addition to several other factors known to play immunostimulatory roles. These factors include MHC class I components (B2M, HLA-A, HLA-B), ADA (encoding adenosine deaminase), ADGRE5 (CD97), CD58 (LFA3), CD74 (encoding invariant chain and CLIP), CD83, CXCL8 (IL8), CXCL16, HLA-F, IL6, IL18, and KITLG. Moreover, both SV-BR-1-GM cells and the responding study subject carried an HLA-DRB3*02:02 allele, raising the question of whether SV-BR-1-GM cells can directly present endogenous antigens to T cells, thereby inducing a tumor-directed immune response. In support of this, SV-BR-1-GM cells (which also carry the HLA-DRB3*01:01 allele) treated with yellow fever virus (YFV) envelope (Env) 43–59 peptides reactivated YFV-DRB3*01:01-specific CD4+ T cells. Thus, the partial HLA allele match between SV-BR-1-GM and the clinical responder might have enabled patient T lymphocytes to directly recognize SV-BR-1-GM TAAs as presented on SV-BR-1-GM MHCs. Taken together, our findings are consistent with a potentially unique mechanism of action by which SV-BR-1-GM cells can act as APCs for previously primed CD4+ T cells.
Collapse
Affiliation(s)
| | - Gerhard Bauer
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Brian Fury
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Sanne Graeve
- BriaCell Therapeutics Corp., Berkeley, CA, United States
| | - Emily L Fledderman
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tye D Petrie
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Dane P Coleal-Bergum
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tia Hackett
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Nicholas H Perotti
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Ying Y Kong
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | | | | | | |
Collapse
|
12
|
Huang F, Zhao H, Du Z, Jiang H. miR-615 Inhibits Prostate Cancer Cell Proliferation and Invasion by Directly Targeting Cyclin D2. Oncol Res 2018; 27:293-299. [PMID: 29471894 PMCID: PMC7848464 DOI: 10.3727/096504018x15190399381143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Previous studies have reported that miR-615 exerts a tumor suppressor role in some tumors, such as esophageal squamous cell carcinoma and non-small cell lung cancer. However, the role of miR-615 in prostate cancer has not been defined. Here we found that miR-615 was downregulated in prostate cancer tissues and cell lines. Overexpression of miR-615 in PC-3 cells significantly inhibited cellular proliferation, migration, and invasion. Moreover, overexpression of miR-615 delayed tumor growth in vivo. In terms of mechanism, we found that cyclin D2 (CCND2) is a target gene of miR-615 in prostate cancer. We showed that miR-615 could bind to the 3′-UTR region of CCND2 mRNA and inhibit its expression. There was a negative correlation between the expression of miR-615 and CCND2 in prostate cancer tissues. Moreover, restoration of cyclin D2 abolished the inhibitory effects of miR-615 on the proliferation, migration, and invasion of prostate cancer cells. Taken together, our study identified miR-615 as a tumor suppressor by targeting cyclin D2 in prostate cancer.
Collapse
Affiliation(s)
| | - Hongjun Zhao
- Department of Urology, Yantai Municipal Laiyang Central Hospital, Laiyang, Shandong Province, P.R. China
| | - Zhaojin Du
- Reproductive Medical Center, Qingdao Women and Children's Hospital, Qingdao University, Qingdao, Shandong Province, P.R. China
| | - Hong Jiang
- Department of Gastroenterology, Yantai Municipal Laiyang Central Hospital, Laiyang, Shandong Province, P.R. China
| |
Collapse
|
13
|
Guo J, Xiao Z, Yu X, Cao R. miR-20b promotes cellular proliferation and migration by directly regulating phosphatase and tensin homolog in prostate cancer. Oncol Lett 2017; 14:6895-6900. [PMID: 29163708 DOI: 10.3892/ol.2017.7041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 02/17/2017] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs are small non-coding RNAs, which are critical regulators of carcinogenesis and tumor progression. Previous studies have identified that microRNA-20b (miR-20b) acts as an oncogene in numerous cancers. However, the role of miR-20b in prostate cancer remains unclear. The present study aimed to investigate the expression of miR-20b in prostate cancer and to examine whether modulating miR-20b expression impacts prostate cancer cellular proliferation and migration. It was revealed that miR-20b was strongly expressed in prostate cancer tissues compared with adjacent normal prostate tissues (P<0.05). Knockdown of miR-20b expression by miR-20b inhibitor inhibited VCaP and PC-3 cell growth and migration. Through bioinformatics analysis, phosphatase and tensin homolog (PTEN) was predicted as a target gene of miR-20b in prostate cancer cells, which was validated by dual-luciferase reporter assay and western blot analysis. In addition, restoration of PTEN expression levels did not affect endogenous miR-20b expression in prostate cancer cells. In conclusion, the present study indicated that miR-20b promotes cellular proliferation and migration by directly regulating PTEN in prostate cancer.
Collapse
Affiliation(s)
- Ju Guo
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zewen Xiao
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xingwei Yu
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Runfu Cao
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Eckert F, Jelas I, Oehme M, Huber SM, Sonntag K, Welker C, Gillies SD, Strittmatter W, Zips D, Handgretinger R, Schilbach K. Tumor-targeted IL-12 combined with local irradiation leads to systemic tumor control via abscopal effects in vivo. Oncoimmunology 2017; 6:e1323161. [PMID: 28680762 DOI: 10.1080/2162402x.2017.1323161] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/19/2017] [Indexed: 12/11/2022] Open
Abstract
NHS-IL12 is an immunocytokine, a fusion protein of IL12's functional domains and a necrosis-targeting antibody, which has shown significant effects against human rhabdomyosarcoma xenografts in a humanized tumor model, including terminal growth arrest and differentiation of the tumor cells. Here, we locally irradiated the tumors, increasing necrosis and consequently intratumoral immune cytokine availability, and asked whether this effect may surmount efficacy of single treatment modality. Humanized mice bearing bilateral rhabdomyosarcoma xenografts were evaluated for tumor burden and survival after irradiation, systemic NHS-IL12 therapy or a combination of both. Intratumoral immune compartments were characterized by immunohistochemistry and molecular methods. TH1-cytokine dependency of underlying effector mechanisms were investigated in vitro in several human tumor cell lines. NHS-IL12 when combined with irradiation terminally arrested tumor growth and significantly improved survival. Combination treatment induced dense intratumoral T-cell infiltrates, clonal epitope-specific T-cell expansions, expression of cytotoxins, decreased pro-tumorigenic cytokines and induced senescence and differentiation in the cancer cells. Senescence and differentiation were reproduced in vitro and confirmed to be dependent on TH1 cytokines IFNγ and TNF-α. NHS-IL12 and irradiation together induced broad intratumoral TH1 biased NK and T-cell compartments, established antitumoral cytokine profiles and irreversibly growth arrested tumor cells, leading to systemic cancer control and improved survival. For the first time, we describe immune-induced senescence as a novel mechanism resulting from a treatment regimen combining irradiation with immunotherapy.
Collapse
Affiliation(s)
- Franziska Eckert
- Department of Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ivan Jelas
- Department of Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Moritz Oehme
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Katja Sonntag
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christian Welker
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | | | - Daniel Zips
- Department of Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Karin Schilbach
- Department of General Pediatrics, Oncology/Hematology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
15
|
Datta D, Aftabuddin M, Gupta DK, Raha S, Sen P. Human Prostate Cancer Hallmarks Map. Sci Rep 2016; 6:30691. [PMID: 27476486 PMCID: PMC4967902 DOI: 10.1038/srep30691] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022] Open
Abstract
Human prostate cancer is a complex heterogeneous disease that mainly affects elder male population of the western world with a high rate of mortality. Acquisitions of diverse sets of hallmark capabilities along with an aberrant functioning of androgen receptor signaling are the central driving forces behind prostatic tumorigenesis and its transition into metastatic castration resistant disease. These hallmark capabilities arise due to an intense orchestration of several crucial factors, including deregulation of vital cell physiological processes, inactivation of tumor suppressive activity and disruption of prostate gland specific cellular homeostasis. The molecular complexity and redundancy of oncoproteins signaling in prostate cancer demands for concurrent inhibition of multiple hallmark associated pathways. By an extensive manual curation of the published biomedical literature, we have developed Human Prostate Cancer Hallmarks Map (HPCHM), an onco-functional atlas of human prostate cancer associated signaling and events. It explores molecular architecture of prostate cancer signaling at various levels, namely key protein components, molecular connectivity map, oncogenic signaling pathway map, pathway based functional connectivity map etc. Here, we briefly represent the systems level understanding of the molecular mechanisms associated with prostate tumorigenesis by considering each and individual molecular and cell biological events of this disease process.
Collapse
Affiliation(s)
- Dipamoy Datta
- Department of Biotechnology, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Md Aftabuddin
- Maulana Abul Kalam Azad University of Technology, West Bengal, Salt Lake, Sector-I, Kolkata 700064, India
| | - Dinesh Kumar Gupta
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| | - Sanghamitra Raha
- Department of Biotechnology, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Prosenjit Sen
- Biological Chemistry Division, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
16
|
Immune Regulation and Antitumor Effect of TIM-1. J Immunol Res 2016; 2016:8605134. [PMID: 27413764 PMCID: PMC4931049 DOI: 10.1155/2016/8605134] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/10/2016] [Accepted: 04/28/2016] [Indexed: 01/25/2023] Open
Abstract
T cells play an important role in antitumor immunity, and the T cell immunoglobulin domain and the mucin domain protein-1 (TIM-1) on its surface, as a costimulatory molecule, has a strong regulatory effect on T cells. TIM-1 can regulate and enhance type 1 immune response of tumor association. Therefore, TIM-1 costimulatory pathways may be a promising therapeutic target in future tumor immunotherapy. This review describes the immune regulation and antitumor effect of TIM-1.
Collapse
|
17
|
Fu Y, Cao F. MicroRNA-125a-5p regulates cancer cell proliferation and migration through NAIF1 in prostate carcinoma. Onco Targets Ther 2015; 8:3827-35. [PMID: 26719710 PMCID: PMC4689268 DOI: 10.2147/ott.s92314] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background We investigated the functional roles of microRNA-125a-5p in regulating human prostate carcinoma. Methods Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was conducted to evaluate the gene expression levels of miR-125a-5p in eight prostate cancer cell lines and nine biopsy specimens from patients with prostate cancer. miR-125a-5p was genetically knocked down in prostate cancer cell lines, DU145 and VCaP cells by lentiviral transduction. The effects of miR-125a-5p downregulation on prostate cancer cell proliferation and migration were evaluated by MTT assay and transwell assay, respectively. Direct regulation of miR-125a-5p on its downstream targets, NAIF1, and apoptotic gene caspase-3 were evaluated through dual-luciferase reporter assay, qRT-PCR, and Western blot, respectively. NAIF1 was then ectopically overexpressed in DU145 and VCaP cells to modulate prostate cancer cell proliferation and migration. Finally, the effects of miR-125a-5p downregulation or NAIF1 overexpression on the growth of in vivo prostate cancer xenograft were evaluated. Results miR-125a-5p was upregulated in prostate cancer cell lines and human prostate carcinomas. Lentivirus induced miR-125a-5p downregulation in DU145 and VCaP cells inhibited prostate cancer cell proliferation or migration. NAIF1 was the direct target of miR-125a-5p, as both gene and protein expression levels of NAIF1, as well as caspase-3 were upregulated by miR-125a-5p. Forced overexpression of NAIF1 had similar antitumor effects as miR-125a-5p downregulation on prostate cancer cell proliferation and migration. In vivo prostate xenograft assay confirmed the tumor-suppressive effect of miR-125a-5p downregulation or NAIF1 overexpression. Conclusion miR-125a-5p regulates prostate cancer cell proliferation and migration through NAIF1.
Collapse
Affiliation(s)
- Yi Fu
- Department of Urology, The Oilfield General Hospital of Daqing, Daqing, People's Republic of China
| | - Fuhua Cao
- Department of Urology, The Oilfield General Hospital of Daqing, Daqing, People's Republic of China
| |
Collapse
|