101
|
Sheehan K, Schalper KA. Tumor Microenvironment: Immune Effector and Suppressor Imbalance. Lung Cancer 2021. [DOI: 10.1007/978-3-030-74028-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
102
|
Rathi N, McFarland TR, Nussenzveig R, Agarwal N, Swami U. Evolving Role of Immunotherapy in Metastatic Castration Refractory Prostate Cancer. Drugs 2020; 81:191-206. [PMID: 33369720 PMCID: PMC7932934 DOI: 10.1007/s40265-020-01456-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Immunotherapies have shown remarkable success in the treatment of multiple cancer types; however, despite encouraging preclinical activity, registration trials of immunotherapy in prostate cancer have largely been unsuccessful. Sipuleucel-T remains the only approved immunotherapy for the treatment of asymptomatic or minimally symptomatic metastatic castrate-resistant prostate cancer based on modest improvement in overall survival. This immune evasion in the case of prostate cancer has been attributed to tumor-intrinsic factors, an immunosuppressive tumor microenvironment, and host factors, which ultimately make it an inert 'cold' tumor. Recently, multiple approaches have been investigated to turn prostate cancer into a 'hot' tumor. Antibodies directed against programmed cell death protein 1 have a tumor agnostic approval for a small minority of patients with microsatellite instability-high or mismatch repair-deficient metastatic prostate cancer. Herein, we present an overview of the current immunotherapy landscape in metastatic castration-resistant prostate cancer with a focus on immune checkpoint inhibitors. We describe the results of clinical trials of immune checkpoint inhibitors in patients with metastatic castration-resistant prostate cancer; either as single agents or in combination with other checkpoint inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors, tyrosine kinase inhibitors, novel hormonal therapies, chemotherapies, and radioligands. Finally, we review upcoming immunotherapies, including novel monoclonal antibodies, chimeric-antigen receptor (CAR) T cells, Bi-Specific T cell Engagers (BiTEs), therapies targeting the adenosine pathway, and other miscellaneous agents.
Collapse
Affiliation(s)
- Nityam Rathi
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Taylor Ryan McFarland
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Roberto Nussenzveig
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Umang Swami
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
103
|
Gonadotropin-Releasing Hormone Receptors in Prostate Cancer: Molecular Aspects and Biological Functions. Int J Mol Sci 2020; 21:ijms21249511. [PMID: 33327545 PMCID: PMC7765031 DOI: 10.3390/ijms21249511] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Pituitary Gonadotropin-Releasing Hormone receptors (GnRH-R) mediate the activity of the hypothalamic decapeptide GnRH, thus playing a key role in the regulation of the reproductive axis. Early-stage prostate cancer (PCa) is dependent on serum androgen levels, and androgen-deprivation therapy (ADT), based on GnRH agonists and antagonists, represents the standard therapeutic approach for PCa patients. Unfortunately, the tumor often progresses towards the more aggressive castration-resistant prostate cancer (CRPC) stage. GnRH receptors are also expressed in CRPC tissues, where their binding to both GnRH agonists and antagonists is associated with significant antiproliferative/proapoptotic, antimetastatic and antiangiogenic effects, mediated by the Gαi/cAMP signaling cascade. GnRH agonists and antagonists are now considered as an effective therapeutic strategy for CRPC patients with many clinical trials demonstrating that the combined use of these drugs with standard therapies (i.e., docetaxel, enzalutamide, abiraterone) significantly improves disease-free survival. In this context, GnRH-based bioconjugates (cytotoxic drugs covalently linked to a GnRH-based decapeptide) have been recently developed. The rationale of this treatment is that the GnRH peptide selectively binds to its receptors, delivering the cytotoxic drug to CRPC cells while sparing nontumor cells. Some of these compounds have already entered clinical trials.
Collapse
|
104
|
Lund ME, Howard CB, Thurecht KJ, Campbell DH, Mahler SM, Walsh BJ. A bispecific T cell engager targeting Glypican-1 redirects T cell cytolytic activity to kill prostate cancer cells. BMC Cancer 2020; 20:1214. [PMID: 33302918 PMCID: PMC7727117 DOI: 10.1186/s12885-020-07562-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Glypican-1 is a heparan sulfate proteoglycan that is overexpressed in prostate cancer (PCa), and a variety of solid tumors. Importantly, expression is restricted in normal tissue, making it an ideal tumor targeting antigen. Since there is clinical and preclinical evidence of the efficacy of Bispecific T cell Engager (BiTE) therapy in PCa, we sought to produce and test the efficacy of a GPC-1 targeted BiTE construct based on the Miltuximab® sequence. Miltuximab® is a clinical stage anti-GPC-1 antibody that has proven safe in first in human trials. METHODS The single chain variable fragment (scFv) of Miltuximab® and the CD3 binding sequence of Blinatumomab were combined in a standard BiTE format. Binding of the construct to immobilised recombinant CD3 and GPC-1 antigens was assessed by ELISA and BiaCore, and binding to cell surface-expressed antigens was measured by flow cytometry. The ability of MIL-38-CD3 to activate T cells was assessed using in vitro co-culture assays with tumour cell lines of varying GPC-1 expression by measurement of CD69 and CD25 expression, before cytolytic activity was assessed in a similar co-culture. The release of inflammatory cytokines from T cells was measured by ELISA and expression of PD-1 on the T cell surface was measured by flow cytometry. RESULTS Binding activity of MIL-38-CD3 to both cell surface-expressed and immobilised recombinant GPC-1 and CD3 was retained. MIL-38-CD3 was able to mediate the activation of peripheral blood T cells from healthy individuals, resulting in the release of inflammatory cytokines TNF and IFN-g. Activation was reliant on GPC-1 expression as MIL-38-CD3 mediated only low level T cell activation in the presence of C3 cells (constitutively low GPC-1 expression). Activated T cells were redirected to lyse PCa cell lines PC3 and DU-145 (GPC-1 moderate or high expression, respectively) but could not kill GPC-1 negative Raji cells. The expression of PD-1 was up-regulated on the surface of MIL-38-CD3 activated T cells, suggesting potential for synergy with checkpoint inhibition. CONCLUSIONS This study reports preclinical findings into the efficacy of targeting GPC-1 in PCa with BiTE construct MIL-38-CD3. We show the specificity and efficacy of the construct, supporting its further preclinical development.
Collapse
Affiliation(s)
- Maria E Lund
- Glytherix Ltd, Suite 2 Ground Floor, 75 Talavera Road Macquarie Park, Sydney, NSW, 2113, Australia.
| | - Christopher B Howard
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia.,ARC Training Centre for Biopharmaceutical Innovation, The University of Queensland, St Lucia, 4072, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia.,ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, 4072, Australia
| | - Douglas H Campbell
- Glytherix Ltd, Suite 2 Ground Floor, 75 Talavera Road Macquarie Park, Sydney, NSW, 2113, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia.,ARC Training Centre for Biopharmaceutical Innovation, The University of Queensland, St Lucia, 4072, Australia
| | - Bradley J Walsh
- Glytherix Ltd, Suite 2 Ground Floor, 75 Talavera Road Macquarie Park, Sydney, NSW, 2113, Australia.,ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, 4072, Australia
| |
Collapse
|
105
|
Metabolic regulation of prostate cancer heterogeneity and plasticity. Semin Cancer Biol 2020; 82:94-119. [PMID: 33290846 DOI: 10.1016/j.semcancer.2020.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/12/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Metabolic reprogramming is one of the main hallmarks of cancer cells. It refers to the metabolic adaptations of tumor cells in response to nutrient deficiency, microenvironmental insults, and anti-cancer therapies. Metabolic transformation during tumor development plays a critical role in the continued tumor growth and progression and is driven by a complex interplay between the tumor mutational landscape, epigenetic modifications, and microenvironmental influences. Understanding the tumor metabolic vulnerabilities might open novel diagnostic and therapeutic approaches with the potential to improve the efficacy of current tumor treatments. Prostate cancer is a highly heterogeneous disease harboring different mutations and tumor cell phenotypes. While the increase of intra-tumor genetic and epigenetic heterogeneity is associated with tumor progression, less is known about metabolic regulation of prostate cancer cell heterogeneity and plasticity. This review summarizes the central metabolic adaptations in prostate tumors, state-of-the-art technologies for metabolic analysis, and the perspectives for metabolic targeting and diagnostic implications.
Collapse
|
106
|
Flerin NC, Cappellesso F, Pretto S, Mazzone M. Metabolic traits ruling the specificity of the immune response in different cancer types. Curr Opin Biotechnol 2020; 68:124-143. [PMID: 33248423 DOI: 10.1016/j.copbio.2020.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 12/24/2022]
Abstract
Cancer immunotherapy aims to augment the response of the patient's own immune system against cancer cells. Despite effective for some patients and some cancer types, the therapeutic efficacy of this treatment is limited by the composition of the tumor microenvironment (TME), which is not well-suited for the fitness of anti-tumoral immune cells. However, the TME differs between cancer types and tissues, thus complicating the possibility of the development of therapies that would be effective in a large range of patients. A possible scenario is that each type of cancer cell, granted by its own mutations and reminiscent of the functions of the tissue of origin, has a specific metabolism that will impinge on the metabolic composition of the TME, which in turn specifically affects T cell fitness. Therefore, targeting cancer or T cell metabolism could increase the efficacy and specificity of existing immunotherapies, improving disease outcome and minimizing adverse reactions.
Collapse
Affiliation(s)
- Nina C Flerin
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, B3000, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, B3000, Belgium
| | - Federica Cappellesso
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, B3000, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, B3000, Belgium
| | - Samantha Pretto
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, B3000, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, B3000, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, B3000, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, B3000, Belgium.
| |
Collapse
|
107
|
Heidegger I, Necchi A, Pircher A, Tsaur I, Marra G, Kasivisvanathan V, Kretschmer A, Mathieu R, Ceci F, van den Bergh RCN, Thibault C, Tilki D, Valerio M, Surcel C, Gandaglia G. A Systematic Review of the Emerging Role of Immune Checkpoint Inhibitors in Metastatic Castration-resistant Prostate Cancer: Will Combination Strategies Improve Efficacy? Eur Urol Oncol 2020; 4:745-754. [PMID: 33243663 DOI: 10.1016/j.euo.2020.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022]
Abstract
CONTEXT The role of immune checkpoint inhibition (ICI) in the treatment of prostate cancer (PC) still remains elusive. It has been proposed that combination of ICI with other molecules increases the efficacy of immunotherapy in PC. OBJECTIVE To systematically review the literature to assess the potential role of ICI in combination with additional therapies for the management of metastatic castration-resistant PC (mCRPC). EVIDENCE ACQUISITION A systematic review using Medline and scientific meeting records was carried out in September 2020 according to the Preferred Reporting Items for Systematic Review and Meta-analyses guidelines. Ongoing trials of immunotherapy with standard mCRPC therapeutics were identified via a systematic search on ClinicalTrials.gov. EVIDENCE SYNTHESIS A total of five full-text papers, ten congress abstracts, and 15 trials on ClinicalTrials.gov were identified. Preclinical evidence suggests that combinational approaches might be considered to enhance the efficacy of ICI in PC patients. This led to the design of more than 50 immunotherapy-based clinical trials. The majority of the studies focus on ICI combinations with vaccines, androgen deprivation therapy, chemotherapy, PARP inhibition, radiotherapy, and prostate-specific membrane antigen-guided radioligand therapy. Preliminary analyses reported promising findings for the use of ICI in combination with other anticancer therapies. However, no phase 3 trial has yet reported final results, so no level 1 evidence with long-term outcomes currently supports the combination of ICI with mCRPC therapies. CONCLUSIONS Preclinical and clinical trials have demonstrated that combining immunotherapy with standard mCRPC treatment options has the potential to provide a synergistic effect. Nonetheless, a better understanding of the mechanism and of the optimal treatment approach is still needed. PATIENT SUMMARY We reviewed the literature on immunotherapy in combination with standard treatments for patients with metastatic castration-resistant prostate cancer (mCRPC). Current evidence supports the hypothesis that immunotherapeutic drugs might be effective in mCRPC if combined with other treatment options. However, results of ongoing trials are still awaited before this novel treatment approach can be implemented in the daily practice.
Collapse
Affiliation(s)
- Isabel Heidegger
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria.
| | - Andrea Necchi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andreas Pircher
- Department of Internal Medicine V, Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Igor Tsaur
- Department of Urology and Pediatric Urology, Mainz University Medicine, Mainz, Germany
| | - Giancarlo Marra
- Department of Urology, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Veeru Kasivisvanathan
- Division of Surgery and Interventional Science, University College London, London, UK
| | | | | | - Francesco Ceci
- Department of Nuclear Medicine, San Giovanni Battista Hospital, Turin, Italy
| | | | - Constance Thibault
- Department of Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Christian Surcel
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Giorgio Gandaglia
- Division of Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
108
|
Fontana F, Raimondi M, Marzagalli M, Audano M, Beretta G, Procacci P, Sartori P, Mitro N, Limonta P. Mitochondrial functional and structural impairment is involved in the antitumor activity of δ-tocotrienol in prostate cancer cells. Free Radic Biol Med 2020; 160:376-390. [PMID: 32738396 DOI: 10.1016/j.freeradbiomed.2020.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/19/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
The therapeutic options for castration-resistant prostate cancer (CRPC) are still limited. Natural bioactive compounds were shown to possess pro-death properties in different tumors. We previously reported that δ-tocotrienol (δ-TT) induces apoptosis, paraptosis and autophagy in CRPC cells. Here, we investigated whether δ-TT might exert its activity by impairing mitochondrial functions. We demonstrated that, in PC3 and DU145 cells, δ-TT impairs mitochondrial respiration and structural dynamics. In both cell lines, δ-TT triggers mitochondrial Ca2+ and ROS overload. In PC3 cells, both Ca2+ and ROS mediate the δ-TT-related anticancer activities (decrease of cell viability, apoptosis, paraptosis, autophagy and mitophagy). As expected, in autophagy-defective DU145 cells, Ca2+ overload was involved in δ-TT-induced pro-death effects but not in autophagy and mitophagy. In this cell line, we also demonstrated that ROS overload is not involved in the anticancer activities of δ-TT, supporting a low susceptibility of these cells to ROS-related oxidative stress. Taken together, these data demonstrate that, in CRPC cells, δ-TT triggers cell death by inducing mitochondrial functional and structural impairments, providing novel mechanistic insights in its antitumor activity.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Michela Raimondi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Giangiacomo Beretta
- Department of Environmental Science and Policy, Università degli Studi di Milano, Milan, Italy.
| | - Patrizia Procacci
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy.
| | - Patrizia Sartori
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy.
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
109
|
Wu Z, Chen H, Luo W, Zhang H, Li G, Zeng F, Deng F. The Landscape of Immune Cells Infiltrating in Prostate Cancer. Front Oncol 2020; 10:517637. [PMID: 33194581 PMCID: PMC7658630 DOI: 10.3389/fonc.2020.517637] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background This study was to explore the infiltration pattern of immune cells in the prostate cancer (PCa) microenvironment and evaluate the possibility of specific infiltrating immune cells as potential prognostic biomarkers in PCa. Methods Infiltrating percentage of 22 immune cells were extracted from 27 normalized datasets by CIBERSORT algorithm. Samples with CIBERSORT p-value < 0.05 were subsequently merged and divided into normal or tumor groups. The differences of 22 immune cells between normal and tumor tissues were analyzed along with potential infiltrating correlations among 22 immune cells and Gleason grades. SNV data from TCGA was used to calculate the TMB score. A univariate and multivariate regression were used to evaluate the prognostic effects of immune cells in PCa. Results Ten immune cells with significant differences were identified, including seven increased and three decreased infiltrating immune cells from 190 normal prostate tissues and 537 PCa tissues. Among them, the percentage of infiltration of resting NK cells increased the most, whereas the percentage of infiltration of resting mast cells decreased the most. In normal tissues, CD8+ T cells had the strongest infiltrating correlation with monocytes, while activated NK cells and naive B cells were the highest in PCa tissues. Moreover, the infiltration of five immune cells was significantly associated with TMB score and mutations of immune gene change the infiltration of immune cells. The Area Under Curve (AUC) of the multivariate regression model for the five- and 10-year survival prediction of PCa reached 0.796 and 0.862. The validation cohort proved that the model was reproducible. Conclusions This study demonstrated that different infiltrating immune cells in prostate cancer, especially higher infiltrating M1 macrophages and neutrophils in PCa tissue, are associated with patients’ prognosis, suggesting that these two immune cells might be potential targets for PCa diagnosis and prognosis of treatment.
Collapse
Affiliation(s)
- Zhicong Wu
- Department of Clinical Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Clinical Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Hua Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenyang Luo
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hanyun Zhang
- Department of Clinical Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guihuan Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fangyin Zeng
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
110
|
Powers E, Karachaliou GS, Kao C, Harrison MR, Hoimes CJ, George DJ, Armstrong AJ, Zhang T. Novel therapies are changing treatment paradigms in metastatic prostate cancer. J Hematol Oncol 2020; 13:144. [PMID: 33115529 PMCID: PMC7594418 DOI: 10.1186/s13045-020-00978-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/11/2020] [Indexed: 12/11/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains a terminal diagnosis with an aggressive disease course despite currently approved therapeutics. The recent successful development of poly ADP-ribose polymerase (PARP) inhibitors for patients with mCRPC and mutations in DNA damage repair genes has added to the treatment armamentarium and improved personalized treatments for prostate cancer. Other promising therapeutic agents currently in clinical development include the radiotherapeutic 177-lutetium-prostate-specific membrane antigen (PSMA)-617 targeting PSMA-expressing prostate cancer and combinations of immunotherapy with currently effective treatment options for prostate cancer. Herein, we have highlighted the progress in systemic treatments for mCRPC and the promising agents currently in ongoing clinical trials.
Collapse
Affiliation(s)
- Eric Powers
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Georgia Sofia Karachaliou
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Chester Kao
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Michael R Harrison
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Christopher J Hoimes
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA.,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA.,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke University, DUMC 103861, Durham, NC, 27710, USA. .,Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, 27710, USA.
| |
Collapse
|
111
|
Tsaur I, Brandt MP, Juengel E, Manceau C, Ploussard G. Immunotherapy in prostate cancer: new horizon of hurdles and hopes. World J Urol 2020; 39:1387-1403. [PMID: 33106940 PMCID: PMC8514362 DOI: 10.1007/s00345-020-03497-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Prostate cancer (PCa) is the most common malignancy in men and the cause for the second most common cancer-related death in the western world. Despite ongoing development of novel approaches such as second generation androgen receptor targeted therapies, metastatic disease is still fatal. In PCa, immunotherapy (IT) has not reached a therapeutic breakthrough as compared to several other solid tumors yet. We aimed at highlighting the underlying cellular mechanisms crucial for IT in PCa and giving an update of the most essential past and ongoing clinical trials in the field. Methods We searched for relevant publications on molecular and cellular mechanisms involved in the PCa tumor microenvironment and response to IT as well as completed and ongoing IT studies and screened appropriate abstracts of international congresses. Results Tumor progression and patient outcomes depend on complex cellular and molecular interactions of the tumor with the host immune system, driven rather dormant in case of PCa. Sipuleucel-T and pembrolizumab are the only registered immune-oncology drugs to treat this malignancy. A plethora of studies assess combination of immunotherapy with other agents or treatment modalities like radiation therapy which might increase its antineoplastic activity. No robust and clinically relevant prognostic or predictive biomarkers have been established yet. Conclusion Despite immunosuppressive functional status of PCa microenvironment, current evidence, based on cellular and molecular conditions, encourages further research in this field.
Collapse
Affiliation(s)
- Igor Tsaur
- Department of Urology and Pediatric Urology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Maximilian P Brandt
- Department of Urology and Pediatric Urology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Eva Juengel
- Department of Urology and Pediatric Urology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Cécile Manceau
- Department of Urology, CHU-Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Guillaume Ploussard
- Department of Urology, CHU-Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France.,Department of Urology, La Croix du Sud Hospital, Toulouse, France
| |
Collapse
|
112
|
Kerr SC, Morgan MM, Gillette AA, Livingston MK, Lugo-Cintron KM, Favreau PF, Florek L, Johnson BP, Lang JM, Skala MC, Beebe DJ. A bioengineered organotypic prostate model for the study of tumor microenvironment-induced immune cell activation. Integr Biol (Camb) 2020; 12:250-262. [PMID: 33034643 PMCID: PMC7569006 DOI: 10.1093/intbio/zyaa020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/17/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022]
Abstract
The prostate tumor microenvironment (TME) is strongly immunosuppressive; it is largely driven by alteration in cell phenotypes (i.e. tumor-associated macrophages and exhausted cytotoxic T cells) that result in pro-tumorigenic conditions and tumor growth. A greater understanding into how these altered immune cell phenotypes are developed and could potentially be reversed would provide important insights into improved treatment efficacy for prostate cancer. Here, we report a microfluidic model of the prostate TME that mimics prostate ducts across various stages of prostate cancer progression, with associated stroma and immune cells. Using this platform, we exposed immune cells to a benign prostate TME or a metastatic prostate TME and investigated their metabolism, gene and cytokine expression. Immune cells exposed to the metastatic TME showed metabolic differences with a higher redox ratio indicating a switch to a more glycolytic metabolic profile. These cells also increased expression of pro-tumor response cytokines that have been shown to increase cell migration and angiogenesis such as Interleukin-1 (IL-1) a and Granulocyte-macrophage colony-stimulating factor (GM-CSF). Lastly, we observed decreased TLR, STAT signaling and TRAIL expression, suggesting that phenotypes derived from exposure to the metastatic TME could have an impaired anti-tumor response. This platform could provide a valuable tool for studying immune cell phenotypes in in vitro tumor microenvironments.
Collapse
Affiliation(s)
- Sheena C Kerr
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Molly M Morgan
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Amani A Gillette
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Megan K Livingston
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Karina M Lugo-Cintron
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Logan Florek
- Morgridge Institute for Research, Madison, WI, USA
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Melissa C Skala
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
113
|
Montes M, MacKenzie L, McAllister MJ, Roseweir A, McCall P, Hatziieremia S, Underwood MA, Boyd M, Paul A, Plevin R, MacKay SP, Edwards J. Determining the prognostic significance of IKKα in prostate cancer. Prostate 2020; 80:1188-1202. [PMID: 33258506 DOI: 10.1002/pros.24045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/02/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND As the survival of castration-resistant prostate cancer (CRPC) remains poor, and the nuclear factor-κB (NF-κB) pathways play key roles in prostate cancer (PC) progression, several studies have focused on inhibiting the NF-κB pathway through generating inhibitory κB kinase subunit α (IKKα) small molecule inhibitors. However, the identification of prognostic markers able to discriminate which patients could benefit from IKKα inhibitors is urgently required. The present study investigated the prognostic value of IKKα, IKKα phosphorylated at serine 180 (p-IKKα S180) and threonine 23 (p-IKKα T23), and their relationship with the androgen receptor (AR) and Ki67 proliferation index to predict patient outcome. METHODS A cohort of 115 patients with hormone-naïve PC (HNPC) and CRPC specimens available were used to assess tumor cell expression of proteins within both the cytoplasm and the nucleus by immunohistochemistry. The expression levels were dichotomized (low vs high) to determine the associations between IKKα, AR, Ki67, and patients'Isurvival. In addition, an analysis was performed to assess potential IKKα associations with clinicopathological and inflammatory features, and potential IKKα correlations with other cancer pathways essential for CRPC growth. RESULTS High levels of cytoplasmic IKKα were associated with a higher cancer-specific survival in HNPC patients with low AR expression (hazards ratio [HR], 0.33; 95% confidence interval [CI] log-rank, 0.11-0.98; P = .04). Furthermore, nuclear IKKα (HR, 2.60; 95% CI, 1.27-5.33; P = .01) and cytoplasmic p-IKKα S180 (HR, 2.10; 95% CI, 1.17-3.76; P = .01) were associated with a lower time to death from recurrence in patients with CRPC. In addition, high IKKα expression was associated with high levels of T-cells (CD3+ P = .01 and CD8+ P = .03) in HNPC; however, under castration conditions, high IKKα expression was associated with high levels of CD68+ macrophages (P = .04), higher Gleason score (P = .01) and more prostate-specific antigen concentration (P = .03). Finally, we identified crosstalk between IKKα and members of the canonical NF-κB pathway in the nucleus of HNPC. Otherwise, IKKα phosphorylated by noncanonical NF-κB and Akt pathways correlated with members of the canonical NF-κB pathway in CRPC. CONCLUSION The present study reports that patients with CRPC expressing high levels of nuclear IKKα or cytoplasmic p-IKKα S180, which associated with a lower time to death from recurrence, may benefit from IKKα inhibitors.
Collapse
Affiliation(s)
- Melania Montes
- Unit of Gastrointestinal and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Institute of Cancer Science, University of Glasgow, Glasgow, UK
| | - Lewis MacKenzie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Milly J McAllister
- Unit of Gastrointestinal and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Institute of Cancer Science, University of Glasgow, Glasgow, UK
| | - Antonia Roseweir
- Unit of Gastrointestinal and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Institute of Cancer Science, University of Glasgow, Glasgow, UK
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow, UK
| | - Pamela McCall
- Unit of Gastrointestinal and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Institute of Cancer Science, University of Glasgow, Glasgow, UK
| | - Sophia Hatziieremia
- Unit of Gastrointestinal and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Institute of Cancer Science, University of Glasgow, Glasgow, UK
| | - Mark A Underwood
- Department of Urology, Queen Elizabeth University Hospital, Glasgow, UK
| | - Marie Boyd
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Andrew Paul
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Robin Plevin
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Simon P MacKay
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Joanne Edwards
- Unit of Gastrointestinal and Molecular Pathology, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Institute of Cancer Science, University of Glasgow, Glasgow, UK
| |
Collapse
|
114
|
Hua L, Xia H, Zheng W. The Association between Immune Infiltration and Clinical Phenotypes and Prognosis of Prostate Cancer. IRANIAN JOURNAL OF BIOTECHNOLOGY 2020; 18:e2538. [PMID: 34056020 PMCID: PMC8148634 DOI: 10.30498/ijb.2020.2538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Some evidences show that immune infiltration is closely related to the clinical outcomes in cancers such as colorectal cancer. However, previous studies have not explained the diversity of cell types that make up the immune response. In particular, although some studies and reviews have shown that immunotherapy is important for cancer treatment, few studies have elucidated the relationship between prostate cancer (PCa) phenotype and immune infiltration. Objectives: In this study, we analyzed whether different types of tumor-infiltrating immune cells would affect the clinical phenotypes and survival of PCa based on a deconvolution algorithm and annotated gene expression profiles. Materials and Methods: The 22 subsets of immune cells inferred by CIBERSORT and the infiltration abundance of 6 immune cells calculated by TIMER were used to determine the associations between them and the PCa traits and survival response. In addition, the survival tree models were constructed to classify PCa patients into four subtypes, and the traits and prognosis were compared among these subtypes. Results: As a result, we found that some PCa patients with high death risk lacking immune infiltration were related to the poor prognosis. For the cell subsets studied and subtypes analysis, a low proportion of mast resting cells and T-cells follicular helper exhibited the obvious association with poor outcome. Conclusions: In summary, our study suggested the differences in the cellular composition of the immune infiltrate in PCa, and these differences might be important determinants for PCa traits and prognosis.
Collapse
Affiliation(s)
- Lin Hua
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, 100069, China
| | - Hong Xia
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, 100069, China
| | - Weiying Zheng
- School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
115
|
Gamat-Huber M, Jeon D, Johnson LE, Moseman JE, Muralidhar A, Potluri HK, Rastogi I, Wargowski E, Zahm CD, McNeel DG. Treatment Combinations with DNA Vaccines for the Treatment of Metastatic Castration-Resistant Prostate Cancer (mCRPC). Cancers (Basel) 2020; 12:cancers12102831. [PMID: 33008010 PMCID: PMC7601088 DOI: 10.3390/cancers12102831] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 01/04/2023] Open
Abstract
Simple Summary The only vaccine approved by FDA as a treatment for cancer is sipuleucel-T, a therapy for patients with metastatic castration-resistant prostate cancer (mCRPC). Most investigators studying anti-tumor vaccines believe they will be most effective as parts of combination therapies, rather than used alone. Unfortunately, the cost and complexity of sipuleucel-T makes it difficult to feasibly be used in combination with many other agents. In this review article we discuss the use of DNA vaccines as a simpler vaccine approach that has demonstrated efficacy in several animal species. We discuss the use of DNA vaccines in combination with traditional treatments for mCRPC, and other immune-modulating treatments, in preclinical and early clinical trials for patients with mCRPC. Abstract Metastatic castration-resistant prostate cancer (mCRPC) is a challenging disease to treat, with poor outcomes for patients. One antitumor vaccine, sipuleucel-T, has been approved as a treatment for mCRPC. DNA vaccines are another form of immunotherapy under investigation. DNA immunizations elicit antigen-specific T cells that cause tumor cell lysis, which should translate to meaningful clinical responses. They are easily amenable to design alterations, scalable for large-scale manufacturing, and thermo-stable for easy transport and distribution. Hence, they offer advantages over other vaccine formulations. However, clinical trials with DNA vaccines as a monotherapy have shown only modest clinical effects against tumors. Standard therapies for CRPC including androgen-targeted therapies, radiation therapy and chemotherapy all have immunomodulatory effects, which combined with immunotherapies such as DNA vaccines, could potentially improve treatment. In addition, many investigational drugs are being developed which can augment antitumor immunity, and together with DNA vaccines can further enhance antitumor responses in preclinical models. We reviewed the literature available prior to July 2020 exploring the use of DNA vaccines in the treatment of prostate cancer. We also examined various approved and experimental therapies that could be combined with DNA vaccines to potentially improve their antitumor efficacy as treatments for mCRPC.
Collapse
|
116
|
Haughey CM, Mukherjee D, Steele RE, Popple A, Dura-Perez L, Pickard A, Patel M, Jain S, Mullan PB, Williams R, Oliveira P, Buckley NE, Honeychurch J, S. McDade S, Illidge T, Mills IG, Eddie SL. Investigating Radiotherapy Response in a Novel Syngeneic Model of Prostate Cancer. Cancers (Basel) 2020; 12:E2804. [PMID: 33003551 PMCID: PMC7599844 DOI: 10.3390/cancers12102804] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/24/2020] [Indexed: 01/03/2023] Open
Abstract
The prostate cancer (PCa) field lacks clinically relevant, syngeneic mouse models which retain the tumour microenvironment observed in PCa patients. This study establishes a cell line from prostate tumour tissue derived from the Pten-/-/trp53-/- mouse, termed DVL3 which when subcutaneously implanted in immunocompetent C57BL/6 mice, forms tumours with distinct glandular morphology, strong cytokeratin 8 and androgen receptor expression, recapitulating high-risk localised human PCa. Compared to the commonly used TRAMP C1 model, generated with SV40 large T-antigen, DVL3 tumours are immunologically cold, with a lower proportion of CD8+ T-cells, and high proportion of immunosuppressive myeloid derived suppressor cells (MDSCs), thus resembling high-risk PCa. Furthermore, DVL3 tumours are responsive to fractionated RT, a standard treatment for localised and metastatic PCa, compared to the TRAMP C1 model. RNA-sequencing of irradiated DVL3 tumours identified upregulation of type-1 interferon and STING pathways, as well as transcripts associated with MDSCs. Upregulation of STING expression in tumour epithelium and the recruitment of MDSCs following irradiation was confirmed by immunohistochemistry. The DVL3 syngeneic model represents substantial progress in preclinical PCa modelling, displaying pathological, micro-environmental and treatment responses observed in molecular high-risk disease. Our study supports using this model for development and validation of treatments targeting PCa, especially novel immune therapeutic agents.
Collapse
Affiliation(s)
- Charles M. Haughey
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Debayan Mukherjee
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (D.M.); (A.P.); (M.P.); (J.H.)
| | - Rebecca E. Steele
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London SM2 5NG, UK
| | - Amy Popple
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (D.M.); (A.P.); (M.P.); (J.H.)
| | - Lara Dura-Perez
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
| | - Adam Pickard
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
- Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester M13 9PL, UK
| | - Mehjabin Patel
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (D.M.); (A.P.); (M.P.); (J.H.)
| | - Suneil Jain
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
| | - Paul B. Mullan
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
| | - Rich Williams
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
| | - Pedro Oliveira
- The Christie Hospital Foundation Trust, Manchester M20 4BX, UK;
| | - Niamh E. Buckley
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Jamie Honeychurch
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (D.M.); (A.P.); (M.P.); (J.H.)
| | - Simon S. McDade
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
| | - Timothy Illidge
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (D.M.); (A.P.); (M.P.); (J.H.)
- The Christie Hospital Foundation Trust, Manchester M20 4BX, UK;
| | - Ian G. Mills
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Sharon L. Eddie
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (C.M.H.); (R.E.S.); (L.D.-P.); (A.P.); (S.J.); (P.B.M.); (R.W.); (N.E.B.); (S.S.M.)
| |
Collapse
|
117
|
Patel D, McKay R, Parsons JK. Immunotherapy for Localized Prostate Cancer: The Next Frontier? Urol Clin North Am 2020; 47:443-456. [PMID: 33008495 DOI: 10.1016/j.ucl.2020.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer vaccines, cytokines, and checkpoint inhibitors are immunotherapeutic agents that act within the cancer immunity cycle. Prostate cancer has provided unique opportunities for, and challenges to, immunotherapy drug development, including low tumor mutational burdens, limited expression of PD-L1, and minimal T-cell intratumoral infiltrates. Nevertheless, efforts are ongoing to help prime prostate tumors by turning a "cold" prostate cancer "hot" and thus rendering them more susceptible to immunotherapy. Combination treatments, use of molecular biomarkers, and use of new immunotherapeutic agents provide opportunities to enhance the immune response to prostate tumors.
Collapse
Affiliation(s)
- Devin Patel
- Department of Urology, University of California San Diego, 9400 Campus Point Drive, MC7987, La Jolla, CA 92093, USA
| | - Rana McKay
- Division of Hematology-Oncology, Department of Internal Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - J Kellogg Parsons
- Department of Urology, University of California San Diego, 9400 Campus Point Drive, MC7987, La Jolla, CA 92093, USA.
| |
Collapse
|
118
|
Handa S, Hans B, Goel S, Bashorun HO, Dovey Z, Tewari A. Immunotherapy in prostate cancer: current state and future perspectives. Ther Adv Urol 2020; 12:1756287220951404. [PMID: 32952615 PMCID: PMC7476347 DOI: 10.1177/1756287220951404] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023] Open
Abstract
Metastatic castrate resistant prostate cancer (PCa) remains an incurable entity. In the era of immunotherapy, the complex PCa microenvironment poses a unique challenge to the successful application of this class of agents. However, in the last decade, a tremendous effort has been made to explore this field of therapeutics. In this review, the physiology of the cancer immunity cycle is highlighted in the context of the prostate tumor microenvironment, and the current evidence for use of various classes of immunotherapy agents including vaccines (dendritic cell based, viral vector based and DNA/mRNA based), immune checkpoint inhibitors, Chimeric antigen receptor T cell therapy, antibody-mediated radioimmunotherapy, antibody drug conjugates, and bispecific antibodies, is consolidated. Finally, the future directions for combinatorial approaches to combat PCa are discussed.
Collapse
Affiliation(s)
- Shivani Handa
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West Hospital, New York, NY, 10019, USA
| | - Bandhul Hans
- Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Shokhi Goel
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Hafis O Bashorun
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Zach Dovey
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Ashutosh Tewari
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
119
|
Gómez V, Mustapha R, Ng K, Ng T. Radiation therapy and the innate immune response: Clinical implications for immunotherapy approaches. Br J Clin Pharmacol 2020; 86:1726-1735. [PMID: 32388875 PMCID: PMC7444780 DOI: 10.1111/bcp.14351] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is an essential component of cancer care, contributing up to 40% of curative cancer treatment regimens. It creates DNA double-strand breaks causing cell death in highly replicating tumour cells. However, tumours can develop acquired resistance to therapy. The efficiency of radiation treatment has been increased by means of combining it with other approaches such as chemotherapy, molecule-targeted therapies and, in recent years, immunotherapy (IT). Cancer-cell apoptosis after radiation treatment causes an immunological reaction that contributes to eradicating the tumour via antigen presentation and subsequent T-cell activation. By contrast, radiotherapy also contributes to the formation of an immunosuppressive environment that hinders the efficacy of the therapy. Innate immune cells from myeloid and lymphoid origin show a very active role in both acquired resistance and antitumourigenic mechanisms. Therefore, many efforts are being made in order to reach a better understanding of the innate immunity reactions after radiation therapy (RT) and the design of new combinatorial IT strategies focused in these particular populations.
Collapse
Affiliation(s)
- Valentí Gómez
- UCL Cancer InstituteUniversity College LondonLondonUK
- Cancer Research UK City of London CentreUK
| | - Rami Mustapha
- School of Cancer and Pharmaceutical SciencesKing's College LondonLondonUK
- Cancer Research UK King's Health Partners CentreUK
| | - Kenrick Ng
- UCL Cancer InstituteUniversity College LondonLondonUK
- Department of Medical OncologyUniversity College Hospitals NHS Foundation TrustUK
| | - Tony Ng
- UCL Cancer InstituteUniversity College LondonLondonUK
- Cancer Research UK City of London CentreUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonLondonUK
- Cancer Research UK King's Health Partners CentreUK
| |
Collapse
|
120
|
Philippou Y, Sjoberg HT, Murphy E, Alyacoubi S, Jones KI, Gordon-Weeks AN, Phyu S, Parkes EE, Gillies McKenna W, Lamb AD, Gileadi U, Cerundolo V, Scheiblin DA, Lockett SJ, Wink DA, Mills IG, Hamdy FC, Muschel RJ, Bryant RJ. Impacts of combining anti-PD-L1 immunotherapy and radiotherapy on the tumour immune microenvironment in a murine prostate cancer model. Br J Cancer 2020; 123:1089-1100. [PMID: 32641865 PMCID: PMC7525450 DOI: 10.1038/s41416-020-0956-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Radiotherapy enhances innate and adaptive anti-tumour immunity. It is unclear whether this effect may be harnessed by combining immunotherapy with radiotherapy fractions used to treat prostate cancer. We investigated tumour immune microenvironment responses of pre-clinical prostate cancer models to radiotherapy. Having defined this landscape, we tested whether radiotherapy-induced tumour growth delay could be enhanced with anti-PD-L1. METHODS Hypofractionated radiotherapy was delivered to TRAMP-C1 and MyC-CaP flank allografts. Tumour growth delay, tumour immune microenvironment flow-cytometry, and immune gene expression were analysed. TRAMP-C1 allografts were then treated with 3 × 5 Gy ± anti-PD-L1. RESULTS 3 × 5 Gy caused tumour growth delay in TRAMP-C1 and MyC-CaP. Tumour immune microenvironment changes in TRAMP-C1 at 7 days post-radiotherapy included increased tumour-associated macrophages and dendritic cells and upregulation of PD-1/PD-L1, CD8+ T-cell, dendritic cell, and regulatory T-cell genes. At tumour regrowth post-3 × 5 Gy the tumour immune microenvironment flow-cytometry was similar to control tumours, however CD8+, natural killer and dendritic cell gene transcripts were reduced. PD-L1 inhibition plus 3 × 5 Gy in TRAMP-C1 did not enhance tumour growth delay versus monotherapy. CONCLUSION 3 × 5 Gy hypofractionated radiotherapy can result in tumour growth delay and immune cell changes in allograft prostate cancer models. Adjuncts beyond immunomodulation may be necessary to improve the radiotherapy-induced anti-tumour response.
Collapse
Affiliation(s)
| | - Hanna T Sjoberg
- Department of Oncology, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Emma Murphy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Said Alyacoubi
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Keaton I Jones
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Su Phyu
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - David A Scheiblin
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, National Institutes of Health, Frederick, 21702, MD, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, National Institutes of Health, Frederick, 21702, MD, USA
| | - David A Wink
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21702, MD, USA
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Ruth J Muschel
- Department of Oncology, University of Oxford, Oxford, UK
| | - Richard J Bryant
- Department of Oncology, University of Oxford, Oxford, UK.
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
121
|
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: 2.5] [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
|
122
|
Tiwari R, Manzar N, Ateeq B. Dynamics of Cellular Plasticity in Prostate Cancer Progression. Front Mol Biosci 2020; 7:130. [PMID: 32754615 PMCID: PMC7365877 DOI: 10.3389/fmolb.2020.00130] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the current advances in the treatment for prostate cancer, the patients often develop resistance to the conventional therapeutic interventions. Therapy-induced drug resistance and tumor progression have been associated with cellular plasticity acquired due to reprogramming at the molecular and phenotypic levels. The plasticity of the tumor cells is mainly governed by two factors: cell-intrinsic and cell-extrinsic. The cell-intrinsic factors involve alteration in the genetic or epigenetic regulators, while cell-extrinsic factors include microenvironmental cues and drug-induced selective pressure. Epithelial-mesenchymal transition (EMT) and stemness are two important hallmarks that dictate cellular plasticity in multiple cancer types including prostate. Emerging evidence has also pinpointed the role of tumor cell plasticity in driving anti-androgen induced neuroendocrine prostate cancer (NEPC), a lethal and therapy-resistant subtype. In this review, we discuss the role of cellular plasticity manifested due to genetic, epigenetic alterations and cues from the tumor microenvironment, and their role in driving therapy resistant prostate cancer.
Collapse
Affiliation(s)
| | | | - Bushra Ateeq
- Molecular Oncology Laboratory, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
123
|
Claps M, Mennitto A, Guadalupi V, Sepe P, Stellato M, Zattarin E, Gillessen SS, Sternberg CN, Berruti A, De Braud FGM, Verzoni E, Procopio G. Immune-checkpoint inhibitors and metastatic prostate cancer therapy: Learning by making mistakes. Cancer Treat Rev 2020; 88:102057. [PMID: 32574991 DOI: 10.1016/j.ctrv.2020.102057] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022]
Abstract
Despite advances in metastatic prostate cancer therapy, expected survival for patients in the castration-resistant phase of disease is poor. Immune-checkpoints inhibitors significantly prolonged life expectancy in some solid tumors and have been evaluated also in advanced stage prostate cancer. The majority of data available derive from preliminary phase I and II trials evaluating CTLA-4 and PD-1 as monotherapy or in combination with each other, vaccines, radiotherapy or targeted/hormonal therapy, achieving only limited benefits in terms of biochemical and radiologic responses. There are many reasons that may explain why prostate cancer responds poorly to modern immunotherapies, such as its characteristic low tumor mutational burden or immune-suppressive tumor microenvironment. The present review summarizes the results obtained treating advanced prostate cancer patients with immune-checkpoints inhibitors and analyzes potential mechanisms of both resistance and sensitivity, in order to hypothesize possible avenues of special interest for future research.
Collapse
Affiliation(s)
- Mélanie Claps
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Alessia Mennitto
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Valentina Guadalupi
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Pierangela Sepe
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Marco Stellato
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Emma Zattarin
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Sommer Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Faculty of Bio Medical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Division of Cancer Science, University of Manchester, Manchester, UK
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York-Presbyterian, New York, United States
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology Unit, Università degli Studi di Brescia, ASST Spedali Civili, Brescia, Italy
| | - Filippo Guglielmo Maria De Braud
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Elena Verzoni
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Giuseppe Procopio
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| |
Collapse
|
124
|
Coletti R, Leonardelli L, Parolo S, Marchetti L. A QSP model of prostate cancer immunotherapy to identify effective combination therapies. Sci Rep 2020; 10:9063. [PMID: 32493951 PMCID: PMC7270132 DOI: 10.1038/s41598-020-65590-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 05/06/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy, by enhancing the endogenous anti-tumor immune responses, is showing promising results for the treatment of numerous cancers refractory to conventional therapies. However, its effectiveness for advanced castration-resistant prostate cancer remains unsatisfactory and new therapeutic strategies need to be developed. To this end, systems pharmacology modeling provides a quantitative framework to test in silico the efficacy of new treatments and combination therapies. In this paper we present a new Quantitative Systems Pharmacology (QSP) model of prostate cancer immunotherapy, calibrated using data from pre-clinical experiments in prostate cancer mouse models. We developed the model by using Ordinary Differential Equations (ODEs) describing the tumor, key components of the immune system, and seven treatments. Numerous combination therapies were evaluated considering both the degree of tumor inhibition and the predicted synergistic effects, integrated into a decision tree. Our simulations predicted cancer vaccine combined with immune checkpoint blockade as the most effective dual-drug combination immunotherapy for subjects treated with androgen-deprivation therapy that developed resistance. Overall, the model presented here serves as a computational framework to support drug development, by generating hypotheses that can be tested experimentally in pre-clinical models.
Collapse
Affiliation(s)
- Roberta Coletti
- University of Trento, Department of mathematics, Trento, 38123, Italy
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy
| | - Lorena Leonardelli
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy
| | - Silvia Parolo
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy
| | - Luca Marchetti
- Fondazione The Microsoft Research - University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, 38068, Italy.
| |
Collapse
|
125
|
Ihle CL, Owens P. Integrating the immune microenvironment of prostate cancer induced bone disease. Mol Carcinog 2020; 59:822-829. [PMID: 32233011 DOI: 10.1002/mc.23192] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/03/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer for men in the U.S. but does not impede patient survival until the disease is metastatic. Metastatic lesions most frequently occur in the bone, which exhibits a distinct microenvironment of immune and bone cell populations. Advances in the diagnosis and treatment of primary PCa allow for the use of tailored therapeutic approaches based on biomarkers, protein expression, and histopathology. Understanding the molecular and cellular characteristics of primary tumors has advanced therapeutic development and survival for patients with PCa. Personalized medicine has only recently emerged for the treatment of metastatic bone lesions. Tumor induced bone disease (TIBD) in patients with PCa can be classified into lytic, blastic, or mixed pathologies, with most patients exhibiting the blastic phenotype. Progress has been made in treating TIBD, but metastatic PCa has yet to be cured. Immune checkpoint inhibitors have exhibited limited responses in immunosuppressive PCa tumors, but have yet to be assessed in metastatic sites which may be susceptible to an increased inflammatory response. Recent discoveries have uncovered distinct tumor microenvironments (TMEs) of blastic and lytic bone metastases from patients with PCa, identifying actionable targets for therapeutic applications, including immune checkpoint inhibitors and targeted therapeutics. Enrichment for macrophages and T cells in patient samples suggests metastatic sites may be reappraised as immunologically targetable, despite their immunologically "cold" primary tumors. The practice of performing bone biopsies will help identify unique cellular and protein targets in the bone TME that can guide therapy decisions.
Collapse
Affiliation(s)
- Claire L Ihle
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Philip Owens
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Veterans Affairs, Research Service, Eastern Colorado Health Care System, Aurora, Colorado
| |
Collapse
|
126
|
Lobo J, Jerónimo C, Henrique R. Targeting the Immune system and Epigenetic Landscape of Urological Tumors. Int J Mol Sci 2020; 21:E829. [PMID: 32012885 PMCID: PMC7037817 DOI: 10.3390/ijms21030829] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/18/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
In the last years, we have witnessed remarkable advances in targeted therapies for cancer patients. There is a growing effort to either replace or reduce the dose of unspecific, systemic (chemo)therapies, given the associated short- and long-term side effects, by introducing more specific targeted therapies as single or combination agents. Due to the well-known implications of the immune system and epigenetic landscape in modulating cancer development, both have been explored as potential targets in several malignancies, including those affecting the genitourinary tract. As the immune system function is also epigenetically regulated, there is rationale for combining both strategies. However, this is still rather underexplored, namely in urological tumors. We aim to briefly review the use of immune therapies in prostate, kidney, bladder, and testicular cancer, and further describe studies providing supporting evidence on their combination with epigenetic-based therapies.
Collapse
Affiliation(s)
- João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (GEBC CI-IPOP) and Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (GEBC CI-IPOP) and Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Rui Henrique
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (GEBC CI-IPOP) and Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| |
Collapse
|
127
|
Anti-androgen hormonal therapy for cancer and other diseases. Eur J Pharmacol 2020; 866:172783. [DOI: 10.1016/j.ejphar.2019.172783] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/29/2019] [Accepted: 11/07/2019] [Indexed: 01/27/2023]
|
128
|
Ye S, Wang H, He K, Shen H, Peng M, Nian Y, Cui R, Yi L. Gene set based systematic analysis of prostate cancer and its subtypes. Future Oncol 2019; 16:4381-4393. [PMID: 31814446 DOI: 10.2217/fon-2019-0459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: A gene set based systematic analysis strategy is used to investigate prostate tumors and its subclusters with focuses on similarities and differences of biological functions. Results: Dysregulation of methylation status, as well as RAS/RAF/ERK and PI3K-ATK signaling pathways, were found to be the most dramatic changes during prostate cancer tumorigenesis. Besides, neural and inflammation microenvironment is also significantly divergent between tumor and adjacent tissues. Insights of subclasses within prostate tumor cohorts revealed four different clusters with distinct gene expression patterns. We found that samples are mainly clustered by immune environments and proliferation traits. Conclusion: The findings of this article may help to advance the progress of identifying better diagnosis biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Senlin Ye
- Department of Urology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| | - Haohui Wang
- Department of Urology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| | - Kancheng He
- Department of Urology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| | - Hongwei Shen
- Central Lab of the Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| | - Mou Peng
- Department of Urology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| | - Yeqi Nian
- Department of Urology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| | - Rongrong Cui
- Institute of Metabolism & Endocrinology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| | - Lu Yi
- Department of Urology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha 410011, PR China
| |
Collapse
|
129
|
Kacar S, Kar F, Hacioglu C, Kanbak G, Sahinturk V. The effects of L-NAME on DU145 human prostate cancer cell line: A cytotoxicity-based study. Hum Exp Toxicol 2019; 39:182-193. [PMID: 31610702 DOI: 10.1177/0960327119880591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Of all cancer types, prostate cancer is the second most common one with an age-standardized incidence rate of 29.3 per 100,000 men worldwide. Nitric oxide (NO) is both a radical and versatile messenger molecule involved in many physiological activities. NO was documented to be highly secreted and utilized by cancer cells. Nω-nitro-L-arginine methyl ester (L-NAME) is utilized for inhibiting NO synthase. Its worst long-term side effect is reported to be hypertension, hence less cytotoxic than chemotherapeutic agents. Herein, we carried out a cytotoxicity study on how different doses of L-NAME affect DU145 human prostate cancer cells. First, toxic doses of L-NAME were determined. Then, while antioxidant capacity was determined by glutathione and total antioxidant status, oxidative stress was evaluated by quantifying malondialdehyde, NO, and total oxidant status levels. Inflammatory effects of L-NAME were investigated by measuring tumor necrosis factor-α and interleukin-6 (IL-6) levels. Apoptotic effects of L-NAME were evaluated by measuring cytochrome C somatic and caspase 3 levels and by staining Bax protein. Finally, morphological analysis was performed. IC50 of L-NAME against DU145 cells was 12.2 mM. In L-NAME-treated DU145 cells, a dose-dependent increase in oxidative stress, inflammatory, and apoptotic marker proteins and decrease in antioxidant capacity were observed. While at the moderate dose of L-NAME, apoptotic changes were commonly observed, at higher doses, vacuolated and swollen cells were also recorded. We believe that the present study will encourage future studies by providing insights about dose and effects of L-NAME.
Collapse
Affiliation(s)
- S Kacar
- Department of Histology and Embryology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - F Kar
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - C Hacioglu
- Department of Medical Biochemistry, Faculty of Medicine, Duzce University, Duzce, Turkey
| | - G Kanbak
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - V Sahinturk
- Department of Histology and Embryology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
130
|
Immunotherapy in Metastatic Castration-Resistant Prostate Cancer: Past and Future Strategies for Optimization. Curr Urol Rep 2019; 20:64. [PMID: 31482315 DOI: 10.1007/s11934-019-0931-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE OF REVIEW To date, prostate cancer has been poorly responsive to immunotherapy. In the current review, we summarize and discuss the current literature on the use of vaccine therapy and checkpoint inhibitor immunotherapy in metastatic castration-resistant prostate cancer (mCRPC). RECENT FINDINGS Sipuleucel-T currently remains the only FDA-approved immunotherapeutic agent for prostate cancer. Single-agent phase 3 vaccine trials with GVAX and PROSTVAC have failed to demonstrate survival benefit to date. Clinical trials using combination approaches, including combination PROSTVAC along with a neoantigen vaccine and checkpoint inhibitor immunotherapy, are ongoing. Checkpoint inhibitor monotherapy clinical trials have demonstrated limited efficacy in advanced prostate cancer, and combination approaches and molecular patient selection are currently under investigation. The optimal use of vaccine therapy and checkpoint inhibitor immunotherapy in metastatic castration-resistant prostate cancer remains to be determined. Ongoing clinical trials will continue to inform future clinical practice.
Collapse
|
131
|
Sakellariou C, Elhage O, Papaevangelou E, Giustarini G, Esteves AM, Smolarek D, Smith RA, Dasgupta P, Galustian C. Prostate cancer cells enhance interleukin-15-mediated expansion of NK cells. BJU Int 2019; 125:89-102. [PMID: 31392791 DOI: 10.1111/bju.14893] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To identify cytokines that can activate and expand NK cells in the presence of prostate cancer cells in order to determine whether these agents may be useful in future intra-tumoural administration in pre-clinical and clinical prostate cancer trials. MATERIALS AND METHODS Lymphocytes isolated from normal donor blood were set up in co-cultures with either cancer or non-cancerous prostate cell lines, together with each of the cytokines interleukin (IL)-2, IL-12, IL-15, interferon (IFN)-γ or IL-21 for a period of 7 days. Then, expansion of NK cells, NKT cells and CD8 T cells was measured by flow cytometry and compared with the expansion of the same cells in the absence of prostate cells. The cytotoxic activity of NK cells, as measured by perforin and tumour cell killing, was also assessed. NK cell receptors and their corresponding ligands on prostate tumour cells were analysed to determine whether any of these were modulated by co-culture. The role of the tumour-secreted heat shock proteins HSP90 and HSP70 in the expansion of NK cells in the co-cultures was also investigated because of their effects on NK and CD8 T-cell activation. RESULTS We showed that, among a panel of cytokines known to cause NK cell activation and expansion, only IL-15 could actively induce expansion of NK, NKT and CD8 T cells in the presence of prostate cancer cell lines. Furthermore, the expansion of NK cells was far greater (up to 50% greater) in the presence of the cancer cells (LNCaP, PC3) than when lymphocytes were incubated alone. In contrast, non-cancerous cell lines (PNT2 and WPMY-1) did not exert any expansion of NK cells. The cytolytic activity of the NK cells, as measured by perforin, CD107a and killing of tumour cells, was also greatest in co-cultures with IL-15. Examination of NK cell receptors shows that NKG2D is upregulated to a greater degree in the presence of prostate cancer cells, compared with the upregulation with IL-15 in lymphocytes alone. However, blocking of NKG2D does not inhibit the enhanced expansion of NK cells in the presence of tumour cells. CONCLUSIONS Among a panel of NK cell-activating cytokines, IL-15 was the only cytokine that could stimulate expansion of NK cells in the presence of prostate cancer cells; therefore IL-15 may be a good candidate for novel future intra-tumoural therapy of the disease.
Collapse
Affiliation(s)
- Christina Sakellariou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | | | - Efthymia Papaevangelou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Giulio Giustarini
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Ana M Esteves
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Dorota Smolarek
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Richard A Smith
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| | - Prokar Dasgupta
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK.,Urology Centre, Guys Hospital, London, UK
| | - Christine Galustian
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Kings College London, Guys Hospital, London, UK
| |
Collapse
|
132
|
Patel VG, Oh WK. The evolving landscape of immunotherapy in advanced prostate cancer. Immunotherapy 2019; 11:903-912. [PMID: 31161846 DOI: 10.2217/imt-2019-0019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Prostate cancer exists in a clinical continuum of hormone-sensitive to castration-resistant disease. Despite the use of chemotherapy and androgen synthesis inhibitors in the castration-resistant setting, this remains a lethal disease. The advent of immune checkpoint blockade has changed the outlook for cancer treatment and survival for several tumors since its first approval in 2011; however, the clinical benefit in castration-resistant prostate cancer (CRPC) is rather limited. Currently, Sipuleucel-T remains the only immune modality to be approved in CRPC setting. Such immune resistance likely exists due to low immunogenicity of prostate tumor cells and an immunosuppressive tumor microenvironment. In this review, we describe the early experiences of immune checkpoint blockade and therapeutic vaccines in CRPC. We then outline strategies currently being implemented to overcome immune resistance, as well as genomic biomarker investigation to identify patients that may harbor more immunogenic tumors. At last, we preview emerging immunotherapeutic platforms.
Collapse
Affiliation(s)
- Vaibhav G Patel
- Department of Medicine, Division of Hematology & Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10010, USA
| | - William K Oh
- Department of Medicine, Division of Hematology & Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10010, USA
| |
Collapse
|