1
|
Lee S, Kang S, Kim WJ. Targeted Protein Degradation in Cancer Therapy via Hydrophobic Polymer-Tagged Nanoparticles. ACS NANO 2025; 19:7742-7754. [PMID: 39982901 DOI: 10.1021/acsnano.4c12747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Targeted protein degradation (TPD) strategies offer a significant advantage over traditional small molecule inhibitors by selectively degrading disease-causing proteins. While small molecules can lead to recurrence and resistance due to compensatory pathway activation, TPD addresses this limitation by promoting protein degradation, thereby reducing the likelihood of recurrence and resistance over the long-term. Despite these benefits, bifunctional TPD molecules face challenges such as low solubility, poor bioavailability, and limited tumor specificity. In this study, we developed polymer-based nanoparticles that combine TPD strategies with nanotechnology through a hydrophobic tagging method. Hydrophobic polymer-tagged nanoparticles facilitate targeted protein degradation by incorporating hydrophobic polymers that mimic hydrophobic residues in misfolded proteins. This system combines degradation and delivery capabilities within a polymer-based platform, inducing protein degradation while improving solubility, stability, and tumor targeting. These nanoparticles consist of a block copolymer composed of an androgen receptor ligand (ARL)-conjugated hydrophobic polylactic acid (PLA) and a hydrophilic polyethylene glycol (PEG), connected by a GSH-cleavable disulfide bond. In aqueous solutions, this block copolymer (ARL-PLA-SS-PEG) forms micelles that degrade in reducible cellular environments. The micelles demonstrated significant in vitro degradation of the target androgen receptor (AR). Furthermore, they achieved substantial tumor accumulation and significantly inhibited tumor growth in a tumor-bearing mouse model. A mechanistic study revealed that the micelle-mediated TPD follows a dual pathway involving both proteasome and autophagosome. This approach has the potential to serve as a universal platform for protein degradation, eliminating the need to develop disease-specific TPD molecules.
Collapse
Affiliation(s)
- Seohee Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Seonwoo Kang
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- OmniaMed Co, Ltd, Pohang 37673, Republic of Korea
| |
Collapse
|
2
|
Zhang Y, Du D, Fang C, Yu X, Fang Y, Liu X, Ou D, Yin H, Liu H, Wang T, Lu L, Li X, Zhang K. Epigenetics disruptions enabled by porphyrin-derived metal-organic frameworks disarm resistances to sonocatalytic ROS anti-tumor actions. FUNDAMENTAL RESEARCH 2025; 5:296-306. [PMID: 40166102 PMCID: PMC11955030 DOI: 10.1016/j.fmre.2022.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 12/07/2022] Open
Abstract
Post-transcriptional epigenetic modifications provide numerous implications for tumor progression, metastasis and recurrence, which also pose resistances to reactive oxygen species (ROS)-based anti-tumor. Herein, we proposed an epigenetic deubiquitination disruption strategy to disarm the ubiquitination-deubiquitination balance-induced resistances to ROS production and ROS-based anti-tumor action for potentiating sonodynamic treatment (SDT) efficiency. To end it, porphyrin-derived metal-organic framework (MOF) sonocatalytic nanoplatforms were developed to load deubiquitination inhibitors (i.e., Auranofin). Ultrasound-triggered Auranofin release from PCN224@Au has been validated to blockade the deubiquitinating process and drive proteasome-mediated target protein degradation. The epigenetic deubiquitination disruption not only synergized with MOF-mediated sonocatalytic ROS production, but also inactivate deubiquitinating enzymes, blockade the deubiquitination process and further remove these resistances, both of which mutually behaved as reciprocal impetuses to significantly magnify SDT outcomes against liver cancers. Such a deubiquitination-engineered disruption approach finds an unprecedented pathway to disarm deubiquitination-induced resistances to SDT and other ROS-based anti-tumor means, which also enlightens us to establish other post-transcriptional epigenetic modification disruption strategies to re-program the tumor microenvironment and elevate the anti-tumor efficiency of various treatment methods (e.g., immunotherapy).
Collapse
Affiliation(s)
- Yan Zhang
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Dou Du
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Chao Fang
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Xin Yu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Tongji University, Shanghai 200433, China
| | - Yujia Fang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Tongji University, Shanghai 200433, China
| | - Xinyu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Tongji University, Shanghai 200433, China
| | - Di Ou
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Haohao Yin
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Hui Liu
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Taixia Wang
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Lu Lu
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Xiaolong Li
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| | - Kun Zhang
- Central Laboratory, Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
3
|
Ke X, Hu H, Peng Q, Ying H, Chu X. USP33 promotes nonalcoholic fatty acid disease-associated fibrosis in gerbils via the c-myc signaling. Biochem Biophys Res Commun 2023; 669:68-76. [PMID: 37267862 DOI: 10.1016/j.bbrc.2023.05.100] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Nonalcoholic fatty acid disease (NAFLD) is a common complication of obesity associated with liver fibrosis. The underlying molecular mechanisms involved in the progression from normal to fibrosis remain unclear. Liver tissues from the liver fibrosis model identified the USP33 gene as a key gene in NAFLD-associated fibrosis. USP33 knockdown inhibited hepatic stellate cell activation and glycolysis in gerbils with NAFLD-associated fibrosis. Conversely, overexpression of USP33 caused a contrast function on hepatic stellate cell activation and glycolysis activation, which was inhibited by c-Myc inhibitor 10058-F4. The copy number of short-chain fatty acids-producing bacterium Alistipes sp. AL-1, Mucispirillum schaedleri, Helicobacter hepaticus in the feces, and the total bile acid level in serum were higher in gerbils with NAFLD-associated fibrosis. Bile acid promoted USP33 expression and inhibiting its receptor reversed hepatic stellate cell activation in gerbils with NAFLD-associated fibrosis. These results suggest that the expression of USP33, an important deubiquitinating enzyme, is increased in NAFLD fibrosis. These data also point to hepatic stellate cells as a key cell type that may respond to liver fibrosis via USP33-induced cell activation and glycolysis.
Collapse
Affiliation(s)
- Xianfu Ke
- Hangzhou Medical College, Zhejiang, China.
| | - Huiying Hu
- Hangzhou Medical College, Zhejiang, China.
| | | | | | | |
Collapse
|
4
|
Drug Resistance and Novel Therapies in Cancers in 2020. Cancers (Basel) 2023; 15:cancers15030717. [PMID: 36765674 PMCID: PMC9913530 DOI: 10.3390/cancers15030717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
After a very successful year in 2019 with 34 publications, our Topic collection "Drug Resistance and Novel Therapies in Cancers" guaranteed another productive year with the publication of 17 research articles and 4 review articles in 2020 [...].
Collapse
|
5
|
Pozas J, Álvarez Rodríguez S, Fernández VA, Burgos J, Santoni M, Manneh Kopp R, Molina-Cerrillo J, Alonso-Gordoa T. Androgen Receptor Signaling Inhibition in Advanced Castration Resistance Prostate Cancer: What Is Expected for the Near Future? Cancers (Basel) 2022; 14:6071. [PMID: 36551557 PMCID: PMC9776956 DOI: 10.3390/cancers14246071] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The androgen signaling pathway is the cornerstone in the treatment of high risk or advanced prostate cancer patients. However, in recent years, different mechanisms of resistance have been defined in this field, limiting the efficacy of the currently approved antiandrogen drugs. Different therapeutic approaches are under research to assess the role of combination therapies against escape signaling pathways or the development of novel antiandrogen drugs to try to solve the primary or acquired resistance against androgen dependent or independent pathways. The present review aims to summarize the current state of androgen inhibition in the therapeutic algorithm of patients with advanced prostate cancer and the mechanisms of resistance to those available drugs. In addition, this review conducted a comprehensive overview of the main present and future research approaches in the field of androgen receptor inhibition to overcome these resistances and the potential new drugs under research coming into this setting.
Collapse
Affiliation(s)
- Javier Pozas
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Sara Álvarez Rodríguez
- Urology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- The Ramon y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain
- Medicine School, Alcalá University, 28805 Madrid, Spain
| | | | - Javier Burgos
- Urology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- The Ramon y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain
- Medicine School, Alcalá University, 28805 Madrid, Spain
| | - Matteo Santoni
- Medical Oncology Department, Mazerata Hospital, 62100 Macerata, Italy
| | - Ray Manneh Kopp
- Sociedad de Oncología y Hematología del Cesar, Valledupar 200001, Colombia
| | - Javier Molina-Cerrillo
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- The Ramon y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain
- Medicine School, Alcalá University, 28805 Madrid, Spain
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- The Ramon y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain
- Medicine School, Alcalá University, 28805 Madrid, Spain
| |
Collapse
|
6
|
Westaby D, Jimenez-Vacas JM, Padilha A, Varkaris A, Balk SP, de Bono JS, Sharp A. Targeting the Intrinsic Apoptosis Pathway: A Window of Opportunity for Prostate Cancer. Cancers (Basel) 2021; 14:51. [PMID: 35008216 PMCID: PMC8750516 DOI: 10.3390/cancers14010051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
Despite major improvements in the management of advanced prostate cancer over the last 20 years, the disease remains invariably fatal, and new effective therapies are required. The development of novel hormonal agents and taxane chemotherapy has improved outcomes, although primary and acquired resistance remains problematic. Inducing cancer cell death via apoptosis has long been an attractive goal in the treatment of cancer. Apoptosis, a form of regulated cell death, is a highly controlled process, split into two main pathways (intrinsic and extrinsic), and is stimulated by a multitude of factors, including cellular and genotoxic stress. Numerous therapeutic strategies targeting the intrinsic apoptosis pathway are in clinical development, and BH3 mimetics have shown promising efficacy for hematological malignancies. Utilizing these agents for solid malignancies has proved more challenging, though efforts are ongoing. Molecular characterization and the development of predictive biomarkers is likely to be critical for patient selection, by identifying tumors with a vulnerability in the intrinsic apoptosis pathway. This review provides an up-to-date overview of cell death and apoptosis, specifically focusing on the intrinsic pathway. It summarizes the latest approaches for targeting the intrinsic apoptosis pathway with BH3 mimetics and discusses how these strategies may be leveraged to treat prostate cancer.
Collapse
Affiliation(s)
- Daniel Westaby
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| | - Juan M. Jimenez-Vacas
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
| | - Ana Padilha
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
| | - Andreas Varkaris
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (A.V.); (S.P.B.)
| | - Steven P. Balk
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (A.V.); (S.P.B.)
| | - Johann S. de Bono
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| | - Adam Sharp
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| |
Collapse
|
7
|
Han X, Zhao L, Xiang W, Qin C, Miao B, McEachern D, Wang Y, Metwally H, Wang L, Matvekas A, Wen B, Sun D, Wang S. Strategies toward Discovery of Potent and Orally Bioavailable Proteolysis Targeting Chimera Degraders of Androgen Receptor for the Treatment of Prostate Cancer. J Med Chem 2021; 64:12831-12854. [PMID: 34431670 DOI: 10.1021/acs.jmedchem.1c00882] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteolysis targeting chimera (PROTAC) small-molecule degraders have emerged as a promising new type of therapeutic agents, but the design of PROTAC degraders with excellent oral pharmacokinetics is a major challenge. In this study, we present our strategies toward the discovery of highly potent PROTAC degraders of androgen receptor (AR) with excellent oral pharmacokinetics. Employing thalidomide to recruit cereblon/cullin 4A E3 ligase and through the rigidification of the linker, we discovered highly potent AR degraders with good oral pharmacokinetic properties in mice with ARD-2128 being the best compound. ARD-2128 achieves 67% oral bioavailability in mice, effectively reduces AR protein and suppresses AR-regulated genes in tumor tissues with oral administration, leading to the effective inhibition of tumor growth in mice without signs of toxicity. This study supports the development of an orally active PROTAC AR degrader for the treatment of prostate cancer and provides insights and guidance into the design of orally active PROTAC degraders.
Collapse
Affiliation(s)
- Xin Han
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lijie Zhao
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Weiguo Xiang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chong Qin
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bukeyan Miao
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hoda Metwally
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aleksas Matvekas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
8
|
Evasion of cell death: A contributory factor in prostate cancer development and treatment resistance. Cancer Lett 2021; 520:213-221. [PMID: 34343635 DOI: 10.1016/j.canlet.2021.07.045] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/17/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022]
Abstract
Cell death is a natural process in organismal development, homeostasis and response to disease or infection that eliminates unnecessary or potentially dangerous cells and acts as an innate barrier to oncogenesis. Inactivation of cell death is a key step in tumour development and also impedes effective response to cancer therapy. Precise execution of unwanted cells is achieved through regulated cell death processes including the intrinsic apoptotic pathway that is governed by the BCL-2 (B-cell lymphoma 2) protein family. There is compelling evidence that intrinsic apoptosis is defective in prostate cancer, particularly in metastatic and castration resistant advanced disease, currently a lethal diagnosis. New therapeutics have been developed to target pro-survival BCL-2 proteins (including BCL-2, BCL-XL and MCL-1) and show promise in reinstating apoptosis to destroy tumour cells in haematological cancers. Here we discuss perturbation of cell death in prostate cancer and how new therapeutics could improve treatment outcome in prostate cancer.
Collapse
|
9
|
Wang Y, Chen J, Wu Z, Ding W, Gao S, Gao Y, Xu C. Mechanisms of enzalutamide resistance in castration-resistant prostate cancer and therapeutic strategies to overcome it. Br J Pharmacol 2020; 178:239-261. [PMID: 33150960 DOI: 10.1111/bph.15300] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer is the second most common malignancy in men and androgen deprivation therapy is the first-line therapy. However, most cases will eventually develop castration-resistant prostate cancer after androgen deprivation therapy treatment. Enzalutamide is a second-generation androgen receptor antagonist approved by the Food and Drug Administration to treat patients with castration-resistant prostate cancer. Unfortunately, patients receiving enzalutamide treatment will ultimately develop resistance via various complicated mechanisms. This review examines the emerging information on these resistance mechanisms, including androgen receptor-related signalling pathways, glucocorticoid receptor-related pathways and metabolic effects. Notably, lineage plasticity and phenotype switching, gene polymorphisms and the relationship between microRNAs and drug resistance are addressed. Furthermore, potential therapeutic strategies for enzalutamide-resistant castration-resistant prostate cancer treatment are suggested, which can help discover more effective and specific regimens to overcome enzalutamide resistance.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiyuan Chen
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Zhengjie Wu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shen Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Gao
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
10
|
Han C, Wang Z, Xu Y, Chen S, Han Y, Li L, Wang M, Jin X. Roles of Reactive Oxygen Species in Biological Behaviors of Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1269624. [PMID: 33062666 PMCID: PMC7538255 DOI: 10.1155/2020/1269624] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa), known as a heterogenous disease, has a high incidence and mortality rate around the world and seriously threatens public health. As an inevitable by-product of cellular metabolism, reactive oxygen species (ROS) exhibit beneficial effects by regulating signaling cascades and homeostasis. More and more evidence highlights that PCa is closely associated with age, and high levels of ROS are driven through activation of several signaling pathways with age, which facilitate the initiation, development, and progression of PCa. Nevertheless, excessive amounts of ROS result in harmful effects, such as genotoxicity and cell death. On the other hand, PCa cells adaptively upregulate antioxidant genes to detoxify from ROS, suggesting that a subtle balance of intracellular ROS levels is required for cancer cell functions. The current review discusses the generation and biological roles of ROS in PCa and provides new strategies based on the regulation of ROS for the treatment of PCa.
Collapse
Affiliation(s)
- Chenglin Han
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zilong Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yingkun Xu
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Shuxiao Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yuqing Han
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Lin Li
- Department of Orthopedics, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Muwen Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xunbo Jin
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| |
Collapse
|
11
|
Carpenter VJ, Patel BB, Autorino R, Smith SC, Gewirtz DA, Saleh T. Senescence and castration resistance in prostate cancer: A review of experimental evidence and clinical implications. Biochim Biophys Acta Rev Cancer 2020; 1874:188424. [PMID: 32956765 DOI: 10.1016/j.bbcan.2020.188424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/10/2023]
Abstract
The development of Castration-Resistant Prostate Cancer (CRPC) remains a major challenge in the treatment of this disease. While Androgen Deprivation Therapy (ADT) can result in tumor shrinkage, a primary response of Prostate Cancer (PCa) cells to ADT is a senescent growth arrest. As a response to cancer therapies, senescence has often been considered as a beneficial outcome due to its association with stable growth abrogation, as well as the potential for immune system activation via the Senescence-Associated Secretory Phenotype (SASP). However, there is increasing evidence that not only can senescent cells regain proliferative capacity, but that senescence contributes to deleterious effects of cancer chemotherapy, including disease recurrence. Notably, the preponderance of work investigating the consequences of therapy-induced senescence on tumor progression has been performed in non-PCa models. Here, we summarize the evidence that ADT promotes a senescent response in PCa and postulate mechanisms by which senescence may contribute to the development of castration-resistance. Primarily, we suggest that ADT-induced senescence may support CRPC development via escape from senescence, by cell autonomous-reprogramming, and by the formation of a pro-tumorigenic SASP. However, due to the scarcity of direct evidence from PCa models, the consequences of ADT-induced senescence outlined here remain speculative until the relationship between senescence and CRPC can be experimentally defined.
Collapse
Affiliation(s)
- Valerie J Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Bhaumik B Patel
- Department of Internal Medicine, Division of Hematology, Oncology & Palliative Care, VCU Health, Richmond, VA, USA
| | - Riccardo Autorino
- Department of Surgery, Division of Urology, VCU Health, Richmond, VA, USA
| | | | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tareq Saleh
- The Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan.
| |
Collapse
|
12
|
Marczyk M, Patwardhan GA, Zhao J, Qu R, Li X, Wali VB, Gupta AK, Pillai MM, Kluger Y, Yan Q, Hatzis C, Pusztai L, Gunasekharan V. Multi-Omics Investigation of Innate Navitoclax Resistance in Triple-Negative Breast Cancer Cells. Cancers (Basel) 2020; 12:E2551. [PMID: 32911681 PMCID: PMC7563413 DOI: 10.3390/cancers12092551] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer cells employ various defense mechanisms against drug-induced cell death. Investigating multi-omics landscapes of cancer cells before and after treatment can reveal resistance mechanisms and inform new therapeutic strategies. We assessed the effects of navitoclax, a BCL2 family inhibitor, on the transcriptome, methylome, chromatin structure, and copy number variations of MDA-MB-231 triple-negative breast cancer (TNBC) cells. Cells were sampled before treatment, at 72 h of exposure, and after 10-day drug-free recovery from treatment. We observed transient alterations in the expression of stress response genes that were accompanied by corresponding changes in chromatin accessibility. Most of these changes returned to baseline after the recovery period. We also detected lasting alterations in methylation states and genome structure that suggest permanent changes in cell population composition. Using single-cell analyses, we identified 2350 genes significantly upregulated in navitoclax-resistant cells and derived an 18-gene navitoclax resistance signature. We assessed the navitoclax-response-predictive function of this signature in four additional TNBC cell lines in vitro and in silico in 619 cell lines treated with 251 different drugs. We observed a drug-specific predictive value in both experiments, suggesting that this signature could help guiding clinical biomarker studies involving navitoclax.
Collapse
Affiliation(s)
- Michal Marczyk
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
- Department of Data Science and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Gauri A. Patwardhan
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
| | - Jun Zhao
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT 06511, USA; (J.Z.); (R.Q.); (Y.K.)
| | - Rihao Qu
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT 06511, USA; (J.Z.); (R.Q.); (Y.K.)
| | - Xiaotong Li
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
| | - Vikram B. Wali
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
| | - Abhishek K. Gupta
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
| | - Manoj M. Pillai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA;
| | - Yuval Kluger
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT 06511, USA; (J.Z.); (R.Q.); (Y.K.)
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA;
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA;
| | - Christos Hatzis
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
| | - Vignesh Gunasekharan
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; (M.M.); (G.A.P.); (X.L.); (V.B.W.); (A.K.G.); (M.M.P.); (C.H.); (V.G.)
| |
Collapse
|