1
|
Sun Z, Wei S, Guo Q, Ouyang H, Mao Z, Wang W, Tong Z, Ding Y. V9302-loaded copper-polyphenol hydrogel for enhancing the anti-tumor effect of disulfiram. J Colloid Interface Sci 2025; 678:866-877. [PMID: 39270387 DOI: 10.1016/j.jcis.2024.08.240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
Disulfiram (DSF) is a safe drug with negligible toxicity and Cu-dependent anti-tumor efficacy. However, the accumulation and combination of DSF and Cu in non-tumor tissues leads to systemic toxicity owing to the formation of highly poisonous diethyldithiocarbamate (CuET). In addition, CuET-mediated tumor-killing reactive oxygen species may be weakened by intra-tumoral glutathione (GSH). Herein, a synergistic treatment was developed that utilized the oral delivery of DSF and an injectable polyphenol-copper (PA-Cu) hydrogel loaded with the glutamine uptake inhibitor 2-amino-4-bis(phenoxymethyl)aminobutane (V9302). The injectable hydrogels were synthesized by the Schiff base reaction of hydroxypropyl chitosan (HPCS) with a PA-Cu reversible cross-linking agent. Because of the dynamic coordination between PA and Cu, the PA-Cu/HPCS hydrogel gradually releases Cu2+, forming CuET with DSF. The released V9302 inhibits glutamine uptake, thereby suppressing GSH synthesis and enhancing the therapeutic efficacy of the in situ formed CuET. The synergistic effect of PA-Cu/HPCS/V9302 and DSF in eliminating intracellular GSH and killing tumor cells was validated by in vitro cell experiments. Animal experiments further confirmed that PA-Cu/HPCS/V9302 and DSF have an inhibitory effect on tumor growth while maintaining the biosafety of main organs.
Collapse
Affiliation(s)
- Zhongquan Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, PR China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Shenyu Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, PR China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Quanshi Guo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, PR China
| | - Hanxiang Ouyang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, PR China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, PR China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, PR China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Zongrui Tong
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, PR China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China; National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang 310009, PR China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
2
|
Ohno K, Murakami H, Ogo N, Asai A. Imaging phenotype reveals that disulfirams induce protein insolubility in the mitochondrial matrix. Sci Rep 2024; 14:31401. [PMID: 39733149 DOI: 10.1038/s41598-024-82939-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
The cell painting assay is useful for understanding cellular phenotypic changes and drug effects. To identify other aspects of well-known chemicals, we screened 258 compounds with the cell painting assay and focused on a mitochondrial punctate phenotype seen with disulfiram. To elucidate the reason for this punctate phenotype, we looked for clues by examining staining steps and gene knockdown as well as examining protein solubility and comparing cell lines. From these results, we found that the punctate phenotype was caused by protein insolubility in the mitochondrial matrix. Interestingly, the punctate phenotype of disulfiram was sensitive to the relative expression of LonP1, a protease in the mitochondrial matrix that regulates proteostasis, suggesting that the punctate phenotype manifests when the protein quality control capacity in the mitochondrial matrix is exceeded. Moreover, we discovered that disulfiram and its derivatives, which have all been reported to increase acetaldehyde in the blood after the in vivo intake of alcohol, induced a punctate phenotype as well. The investigated punctate phenotype not only provides a novel clue for elucidating the common mechanism of action among disulfiram derivatives but is also a novel example of chemical perturbation of proteostasis in the mitochondrial matrix.
Collapse
Affiliation(s)
- Ken Ohno
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan
- Discovery Technology Laboratories, Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Muraoka-Higashi, Fujisawa, 251-8555, Kanagawa, Japan
| | - Hisashi Murakami
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan.
| |
Collapse
|
3
|
Huang D, Yao Y, Lou Y, Kou L, Yao Q, Chen R. Disulfiram and cancer immunotherapy: Advanced nano-delivery systems and potential therapeutic strategies. Int J Pharm X 2024; 8:100307. [PMID: 39678262 PMCID: PMC11638648 DOI: 10.1016/j.ijpx.2024.100307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The initial focus of the clinical application of disulfiram was its efficacy in treating alcoholism. However, recent research has revealed its potential as an anti-tumor agent and even as an enhancer of cancer immunotherapy. Disulfiram has received safety approval from the FDA, indicating its safety advantages over other substances used for disease treatment. Although clinical trials have been conducted on strategies involving disulfiram or its combination with other anti-tumor drugs, the treatment outcomes have not yielded satisfactory results, thereby emphasizing the significance of addressing drug delivery as a crucial challenge to be resolved. The need to explore advanced nano-delivery systems and the potential immunotherapy enhancement effect of disulfiram in cancer treatment has increased. This review highlights various ways in which disulfiram can combat cancer and importantly, activate immune-related mechanisms. It also discusses obstacles related to delivering disulfiram and provides existing solutions in terms of drug delivery. These drug delivery strategies offer solutions to address various challenges encountered in diverse delivery methods and aim to achieve enhanced therapeutic effects. The focus is on recent advancements in disulfiram delivery strategies and the future potential of disulfiram in immune regulation.
Collapse
Affiliation(s)
- Di Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yinsha Yao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yifei Lou
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
4
|
Cordeiro S, Oliveira BB, Valente R, Ferreira D, Luz A, Baptista PV, Fernandes AR. Breaking the mold: 3D cell cultures reshaping the future of cancer research. Front Cell Dev Biol 2024; 12:1507388. [PMID: 39659521 PMCID: PMC11628512 DOI: 10.3389/fcell.2024.1507388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Despite extensive efforts to unravel tumor behavior and develop anticancer therapies, most treatments fail when advanced to clinical trials. The main challenge in cancer research has been the absence of predictive cancer models, accurately mimicking the tumoral processes and response to treatments. The tumor microenvironment (TME) shows several human-specific physical and chemical properties, which cannot be fully recapitulated by the conventional 2D cell cultures or the in vivo animal models. These limitations have driven the development of novel in vitro cancer models, that get one step closer to the typical features of in vivo systems while showing better species relevance. This review introduces the main considerations required for developing and exploiting tumor spheroids and organoids as cancer models. We also detailed their applications in drug screening and personalized medicine. Further, we show the transition of these models into novel microfluidic platforms, for improved control over physiological parameters and high-throughput screening. 3D culture models have provided key insights into tumor biology, more closely resembling the in vivo TME and tumor characteristics, while enabling the development of more reliable and precise anticancer therapies.
Collapse
Affiliation(s)
- Sandra Cordeiro
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Beatriz B. Oliveira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ruben Valente
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Daniela Ferreira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - André Luz
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V. Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R. Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
5
|
Luo J, Zeng Y, Chen Z, Luo Y, Shi L, Zhou X. Safety assessment of disulfiram: real-world adverse event analysis based on FAERS database. Front Psychiatry 2024; 15:1498204. [PMID: 39628493 PMCID: PMC11611860 DOI: 10.3389/fpsyt.2024.1498204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/30/2024] [Indexed: 12/06/2024] Open
Abstract
Objective Disulfiram, an FDA-approved medication for AUD, has shown significant potential as a repurposed drug in therapeutic areas including oncology and infectious diseases. The purpose of study is to analyze adverse events (AEs) associated with disulfiram by examining the FAERS database, with a focus on understanding its safety profile in both traditional and emerging applications. Methods AE reports concerning disulfiram in the FAERS database from the fourth quarter of 2002 to the third quarter of 2023 were extracted. Various signal detection methods, including ROR, PRR, BCPNN, and MGPS, were used to detect and categorize adverse events. Results The study collected 52,159,321 AE reports, with 508 reports primarily suspecting disulfiram, identifying 104 Preferred Terms (PTs) across 25 System Organ Classes (SOCs). Major categories of AEs included off label use, psychiatric symptom, liver transplant, and polyneuropathy, with off label use being notably the most reported issue. Strong and new potential AEs were identified, including neurological and psychiatric issues like hypomania, delirium, and vocal cord paralysis; cardiac issues such as electrocardiogram st segment depression; and off label use-related issues like Jarisch-Herxheimer reaction. Conclusion Disulfiram poses risks of various adverse reactions while having promise as a "repurposed" agent. In clinical applications, practitioners should closely monitor occurrences of hepatobiliary disorders, psychiatric disorders, and nervous system disorders.
Collapse
Affiliation(s)
- Jing Luo
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, The Second People's Hospital of Hunan Province (Brain Hospital of Hunan Province), Changsha, Hunan, China
| | - Yaqi Zeng
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, The Second People's Hospital of Hunan Province (Brain Hospital of Hunan Province), Changsha, Hunan, China
| | - Zhe Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaan Luo
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, The Second People's Hospital of Hunan Province (Brain Hospital of Hunan Province), Changsha, Hunan, China
| | - Li Shi
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, The Second People's Hospital of Hunan Province (Brain Hospital of Hunan Province), Changsha, Hunan, China
| | - Xuhui Zhou
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, The Second People's Hospital of Hunan Province (Brain Hospital of Hunan Province), Changsha, Hunan, China
| |
Collapse
|
6
|
Merrill NM, Kaffenberger SD, Bao L, Vandecan N, Goo L, Apfel A, Cheng X, Qin Z, Liu CJ, Bankhead A, Wang Y, Kathawate V, Tudrick L, Serhan HA, Farah Z, Ellimoottil C, Hafez KS, Herrel LA, Montgomery JS, Morgan TM, Salami SS, Weizer AZ, Ulintz PJ, Day ML, Soellner MB, Palmbos PL, Merajver SD, Udager AM. Integrative Drug Screening and Multiomic Characterization of Patient-derived Bladder Cancer Organoids Reveal Novel Molecular Correlates of Gemcitabine Response. Eur Urol 2024; 86:434-444. [PMID: 39155193 DOI: 10.1016/j.eururo.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND AND OBJECTIVE Predicting response to therapy for each patient's tumor is critical to improving long-term outcomes for muscle-invasive bladder cancer. This study aims to establish ex vivo bladder cancer patient-derived organoid (PDO) models that are representative of patients' tumors and determine the potential efficacy of standard of care and curated experimental therapies. METHODS Tumor material was collected prospectively from consented bladder cancer patients to generate short-term PDO models, which were screened against a panel of clinically relevant drugs in ex vivo three-dimensional culture. Multiomic profiling was utilized to validate the PDO models, establish the molecular characteristics of each tumor, and identify potential biomarkers of drug response. Gene expression (GEX) patterns between paired primary tissue and PDO samples were assessed using Spearman's rank correlation coefficients. Molecular correlates of therapy response were identified using Pearson correlation coefficients and Kruskal-Wallis tests with Dunn's post hoc pairwise comparison testing. KEY FINDINGS AND LIMITATIONS A total of 106 tumors were collected from 97 patients, with 65 samples yielding sufficient material for complete multiomic molecular characterization and PDO screening with six to 32 drugs/combinations. Short-term PDOs faithfully represent the tumor molecular characteristics, maintain diverse cell types, and avoid shifts in GEX-based subtyping that accompany long-term PDO cultures. Utilizing an integrative approach, novel correlations between ex vivo drug responses and genomic alterations, GEX, and protein expression were identified, including a multiomic signature of gemcitabine response. The positive predictive value of ex vivo drug responses and the novel multiomic gemcitabine response signature need to be validated in future studies. CONCLUSIONS AND CLINICAL IMPLICATIONS Short-term PDO cultures retain the molecular characteristics of tumor tissue and avoid shifts in expression-based subtyping that have plagued long-term cultures. Integration of multiomic profiling and ex vivo drug screening data identifies potential predictive biomarkers, including a novel signature of gemcitabine response. PATIENT SUMMARY Better models are needed to predict patient response to therapy in bladder cancer. We developed a platform that uses short-term culture to best mimic each patient's tumor and assess potential sensitivity to therapeutics.
Collapse
Affiliation(s)
- Nathan M Merrill
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Liwei Bao
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Laura Goo
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Athena Apfel
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xu Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhaoping Qin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chia-Jen Liu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Armand Bankhead
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yin Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Varun Kathawate
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lila Tudrick
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Habib A Serhan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zackariah Farah
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chad Ellimoottil
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Khaled S Hafez
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Lindsey A Herrel
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey S Montgomery
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Todd M Morgan
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Simpa S Salami
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Alon Z Weizer
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Peter J Ulintz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mark L Day
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | | | - Phillip L Palmbos
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sofia D Merajver
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Aaron M Udager
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Zhang C, Huang T, Li L. Targeting cuproptosis for cancer therapy: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:68. [PMID: 39152464 PMCID: PMC11328505 DOI: 10.1186/s13045-024-01589-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
Cuproptosis is a newly identified form of cell death induced by excessive copper (Cu) accumulation within cells. Mechanistically, cuproptosis results from Cu-induced aggregation of dihydrolipoamide S-acetyltransferase, correlated with the mitochondrial tricarboxylic acid cycle and the loss of iron-sulfur cluster proteins, ultimately resulting in proteotoxic stress and triggering cell death. Recently, cuproptosis has garnered significant interest in tumor research due to its potential as a crucial therapeutic strategy against cancer. In this review, we summarized the cellular and molecular mechanisms of cuproptosis and its relationship with other types of cell death. Additionally, we reviewed the current drugs or strategies available to induce cuproptosis in tumor cells, including Cu ionophores, small compounds, and nanomedicine. Furthermore, we targeted cell metabolism and specific regulatory genes in cancer therapy to enhance tumor sensitivity to cuproptosis. Finally, we discussed the feasibility of targeting cuproptosis to overcome tumor chemotherapy and immunotherapy resistance and suggested future research directions. This study suggested that targeting cuproptosis could open new avenues for developing tumor therapy.
Collapse
Affiliation(s)
- Chenliang Zhang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Tingting Huang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Liping Li
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
8
|
Wu J, He J, Liu Z, Zhu X, Li Z, Chen A, Lu J. Cuproptosis: Mechanism, role, and advances in urological malignancies. Med Res Rev 2024; 44:1662-1682. [PMID: 38299968 DOI: 10.1002/med.22025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 02/02/2024]
Abstract
Prostate, bladder, and kidney cancers are the most common malignancies of the urinary system. Chemotherapeutic drugs are generally used as adjuvant treatment in the middle, late, or recurrence stages after surgery for urologic cancers. However, traditional chemotherapy is plagued by problems such as poor efficacy, severe side effects, and complications. Copper-containing nanomedicines are promising novel cancer treatment modalities that can potentially overcome these disadvantages. Copper homeostasis and cuproptosis play crucial roles in the development, adaptability, and therapeutic sensitivity of urological malignancies. Cuproptosis refers to the direct binding of copper ions to lipoylated components of the tricarboxylic acid cycle, leading to protein oligomerization, loss of iron-sulfur proteins, proteotoxic stress, and cell death. This review focuses on copper homeostasis and cuproptosis as well as recent findings on copper and cuproptosis in urological malignancies. Furthermore, we highlight the potential therapeutic applications of copper- and cuproptosis-targeted therapies to better understand cuproptosis-based drugs for the treatment of urological tumors in the future.
Collapse
Affiliation(s)
- Jialong Wu
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Jide He
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Zenan Liu
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Xuehua Zhu
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Ziang Li
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
9
|
Mohapatra D, Senapati PC, Senapati S, Pandey V, Dubey PK, Singh S, Sahu AN. Quality-by-design-based microemulsion of disulfiram for repurposing in melanoma and breast cancer therapy. Ther Deliv 2024; 15:521-544. [PMID: 38949622 PMCID: PMC11412148 DOI: 10.1080/20415990.2024.2363136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 05/30/2024] [Indexed: 07/02/2024] Open
Abstract
Aim: The current study aims to develop and optimize microemulsions (ME) through Quality-by-Design (QbD) approach to improve the aqueous solubility and dissolution of poorly water-soluble drug disulfiram (DSF) for repurposing in melanoma and breast cancer therapy.Materials & methods: The ME was formulated using Cinnamon oil & Tween® 80, statistically optimized using a D-optimal mixture design-based QbD approach to develop the best ME with low vesicular size (Zavg) and polydispersity index (PDI).Results: The DSF-loaded optimized stable ME showed enhanced dissolution, in-vitro cytotoxicity and improved cellular uptake in B16F10 and MCF-7 cell lines compared with their unformulated free DSF.Conclusion: Our investigations suggested the potential of the statistically designed DSF-loaded optimized ME for repurposing melanoma and breast cancer therapy.
Collapse
Affiliation(s)
- Debadatta Mohapatra
- Phytomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Varanasi- 221005, Uttar Pradesh, India
| | | | - Shantibhusan Senapati
- Tumor Microenvironment & Animal Models Laboratory, Institute of Life Sciences, Bhubaneswar- 751023, Odisha, India
| | - Vivek Pandey
- Centre for Genetics Disorders, Institute of Science (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Pawan K Dubey
- Centre for Genetics Disorders, Institute of Science (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Sanjay Singh
- Nanomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Varanasi- 221005, Uttar Pradesh, India
| | - Alakh N Sahu
- Phytomedicine Research Laboratory, Department of Pharmaceutical Engineering & Technology, IIT (BHU), Varanasi- 221005, Uttar Pradesh, India
| |
Collapse
|
10
|
Li L, Zhou H, Zhang C. Cuproptosis in cancer: biological implications and therapeutic opportunities. Cell Mol Biol Lett 2024; 29:91. [PMID: 38918694 PMCID: PMC11201306 DOI: 10.1186/s11658-024-00608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Cuproptosis, a newly identified copper (Cu)-dependent form of cell death, stands out due to its distinct mechanism that sets it apart from other known cell death pathways. The molecular underpinnings of cuproptosis involve the binding of Cu to lipoylated enzymes in the tricarboxylic acid cycle. This interaction triggers enzyme aggregation and proteotoxic stress, culminating in cell death. The specific mechanism of cuproptosis has yet to be fully elucidated. This newly recognized form of cell death has sparked numerous investigations into its role in tumorigenesis and cancer therapy. In this review, we summarized the current knowledge on Cu metabolism and its link to cancer. Furthermore, we delineated the molecular mechanisms of cuproptosis and summarized the roles of cuproptosis-related genes in cancer. Finally, we offered a comprehensive discussion of the most recent advancements in Cu ionophores and nanoparticle delivery systems that utilize cuproptosis as a cutting-edge strategy for cancer treatment.
Collapse
Affiliation(s)
- Liping Li
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Houfeng Zhou
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Chenliang Zhang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
11
|
Zeng M, Wu B, Wei W, Jiang Z, Li P, Quan Y, Hu X. Disulfiram: A novel repurposed drug for cancer therapy. Chin Med J (Engl) 2024; 137:1389-1398. [PMID: 38275022 PMCID: PMC11188872 DOI: 10.1097/cm9.0000000000002909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Indexed: 01/27/2024] Open
Abstract
ABSTRACT Cancer is a major global health issue. Effective therapeutic strategies can prolong patients' survival and reduce the costs of treatment. Drug repurposing, which identifies new therapeutic uses for approved drugs, is a promising approach with the advantages of reducing research costs, shortening development time, and increasing efficiency and safety. Disulfiram (DSF), a Food and Drug Administration (FDA)-approved drug used to treat chronic alcoholism, has a great potential as an anticancer drug by targeting diverse human malignancies. Several studies show the antitumor effects of DSF, particularly the combination of DSF and copper (DSF/Cu), on a wide range of cancers such as glioblastoma (GBM), breast cancer, liver cancer, pancreatic cancer, and melanoma. In this review, we summarize the antitumor mechanisms of DSF/Cu, including induction of intracellular reactive oxygen species (ROS) and various cell death signaling pathways, and inhibition of proteasome activity, as well as inhibition of nuclear factor-kappa B (NF-κB) signaling. Furthermore, we highlight the ability of DSF/Cu to target cancer stem cells (CSCs), which provides a new approach to prevent tumor recurrence and metastasis. Strikingly, DSF/Cu inhibits several molecular targets associated with drug resistance, and therefore it is becoming a novel option to increase the sensitivity of chemo-resistant and radio-resistant patients. Studies of DSF/Cu may shed light on its improved application to clinical tumor treatment.
Collapse
Affiliation(s)
- Min Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Baibei Wu
- The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wenjie Wei
- Institute of Biochemistry of Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zihan Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Peiqiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yuanting Quan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaobo Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
12
|
Qu S, Xu R, Yi G, Li Z, Zhang H, Qi S, Huang G. Patient-derived organoids in human cancer: a platform for fundamental research and precision medicine. MOLECULAR BIOMEDICINE 2024; 5:6. [PMID: 38342791 PMCID: PMC10859360 DOI: 10.1186/s43556-023-00165-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 12/08/2023] [Indexed: 02/13/2024] Open
Abstract
Cancer is associated with a high degree of heterogeneity, encompassing both inter- and intra-tumor heterogeneity, along with considerable variability in clinical response to common treatments across patients. Conventional models for tumor research, such as in vitro cell cultures and in vivo animal models, demonstrate significant limitations that fall short of satisfying the research requisites. Patient-derived tumor organoids, which recapitulate the structures, specific functions, molecular characteristics, genomics alterations and expression profiles of primary tumors. They have been efficaciously implemented in illness portrayal, mechanism exploration, high-throughput drug screening and assessment, discovery of innovative therapeutic targets and potential compounds, and customized treatment regimen for cancer patients. In contrast to conventional models, tumor organoids offer an intuitive, dependable, and efficient in vitro research model by conserving the phenotypic, genetic diversity, and mutational attributes of the originating tumor. Nevertheless, the organoid technology also confronts the bottlenecks and challenges, such as how to comprehensively reflect intra-tumor heterogeneity, tumor microenvironment, tumor angiogenesis, reduce research costs, and establish standardized construction processes while retaining reliability. This review extensively examines the use of tumor organoid techniques in fundamental research and precision medicine. It emphasizes the importance of patient-derived tumor organoid biobanks for drug development, screening, safety evaluation, and personalized medicine. Additionally, it evaluates the application of organoid technology as an experimental tumor model to better understand the molecular mechanisms of tumor. The intent of this review is to explicate the significance of tumor organoids in cancer research and to present new avenues for the future of tumor research.
Collapse
Affiliation(s)
- Shanqiang Qu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
| | - Rongyang Xu
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- The First Clinical Medical College of Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
| | - Huayang Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China.
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China.
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
13
|
Wang Y, Chen Y, Zhang J, Yang Y, Fleishman JS, Wang Y, Wang J, Chen J, Li Y, Wang H. Cuproptosis: A novel therapeutic target for overcoming cancer drug resistance. Drug Resist Updat 2024; 72:101018. [PMID: 37979442 DOI: 10.1016/j.drup.2023.101018] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023]
Abstract
Cuproptosis is a newly identified form of cell death driven by copper. Recently, the role of copper and copper triggered cell death in the pathogenesis of cancers have attracted attentions. Cuproptosis has garnered enormous interest in cancer research communities because of its great potential for cancer therapy. Copper-based treatment exerts an inhibiting role in tumor growth and may open the door for the treatment of chemotherapy-insensitive tumors. In this review, we provide a critical analysis on copper homeostasis and the role of copper dysregulation in the development and progression of cancers. Then the core molecular mechanisms of cuproptosis and its role in cancer is discussed, followed by summarizing the current understanding of copper-based agents (copper chelators, copper ionophores, and copper complexes-based dynamic therapy) for cancer treatment. Additionally, we summarize the emerging data on copper complexes-based agents and copper ionophores to subdue tumor chemotherapy resistance in different types of cancers. We also review the small-molecule compounds and nanoparticles (NPs) that may kill cancer cells by inducing cuproptosis, which will shed new light on the development of anticancer drugs through inducing cuproptosis in the future. Finally, the important concepts and pressing questions of cuproptosis in future research that should be focused on were discussed. This review article suggests that targeting cuproptosis could be a novel antitumor therapy and treatment strategy to overcome cancer drug resistance.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China.
| | - Yongming Chen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Junjing Zhang
- Department of Hepato-Biliary Surgery, Department of Surgery, Huhhot First Hospital, Huhhot 010030, PR China
| | - Yihui Yang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yan Wang
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China
| | - Yuanfang Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, PR China.
| | - Hongquan Wang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| |
Collapse
|
14
|
Nsairat H, Alshaer W, Lafi Z, Ahmad S, Al-Sanabrah A, El-Tanani M. Development and validation of reversed-phase-HPLC method for simultaneous quantification of fulvestrant and disulfiram in liposomes. Bioanalysis 2023; 15:1393-1405. [PMID: 37847056 DOI: 10.4155/bio-2023-0137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
This study aims to develop and validate an HPLC technique for the determination of fulvestrant and disulfiram in liposomes. Encapsulation of both drugs into liposomes may improve their anticancer potential. Validation was performed following the International Conference on Harmonization guidelines for specificity, linearity, limit of detection, limit of quantification, precision, accuracy and robustness. Method specificity displayed no interference and linearity over 25-200 and 12.5-100 μg/ml for fulvestrant and disulfiram, respectively. Precision and accuracy exhibited a low relative standard deviation (<1.70%) and appropriate recovery. The validated method could be designated as a proper method for the simultaneous determination of fulvestrant and disulfiram in liposomes. The liposomes displayed 148.5 ± 5.1 nm size. The encapsulation efficiencies were 73.52 and 50.50% for fulvestrant and disulfiram, respectively.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological & Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, the University of Jordan, Amman, 11942, Jordan
| | - Zainab Lafi
- Pharmacological & Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Somaya Ahmad
- Pharmacological & Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Alaa Al-Sanabrah
- Pharmacological & Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Mohamed El-Tanani
- Pharmacological & Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| |
Collapse
|
15
|
Lin F, Ke ZB, Xue YT, Chen JY, Cai H, Lin YZ, Li XD, Wei Y, Xue XY, Xu N. A novel CD8 + T cell-related gene signature for predicting the prognosis and immunotherapy efficacy in bladder cancer. Inflamm Res 2023; 72:1665-1687. [PMID: 37578544 DOI: 10.1007/s00011-023-01772-6] [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: 05/25/2023] [Revised: 05/25/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
OBJECTIVE To identify CD8+ T cell-related molecular clusters and establish a novel gene signature for predicting the prognosis and efficacy of immunotherapy in bladder cancer (BCa). METHODS Transcriptome and clinical data of BCa samples were obtained from the Cancer Genome Atlas (TCGA) and GEO databases. The CD8+ T cell-related genes were screened through the CIBERSORT algorithm and correlation analysis. Consensus clustering analysis was utilized to identified CD8+ T cell-related molecular clusters. A novel CD8+ T cell-related prognostic model was developed using univariate Cox regression analysis and Lasso regression analysis. Internal and external validations were performed and the validity of the model was validated in a real-world cohort. Finally, preliminary experimental verifications were carried out to verify the biological functions of SH2D2A in bladder cancer. RESULTS A total of 52 CD8+ T cell-related prognostic genes were screened and two molecular clusters with notably diverse immune cell infiltration, prognosis and clinical features were developed. Then, a novel CD8+ T cell-related prognostic model was constructed. The patients with high-risk scores exhibited a significantly worse overall survival in training, test, whole TCGA and validating cohort. The AUC was 0.766, 0.725, 0.739 and 0.658 in the four cohorts sequentially. Subgroup analysis suggested that the novel prognostic model has a robust clinical application for selecting high-risk patients. Finally, we confirmed that patients in the low-risk group might benefit more from immunotherapy or chemotherapy, and validated the prognostic model in a real-world immunotherapy cohort. Preliminary experiment showed that SH2D2A was capable of attenuating proliferation, migration and invasion of BCa cells. CONCLUSIONS CD8+ T cell-related molecular clusters were successfully identified. Besides, a novel CD8+ T cell-related prognostic model with an excellent predictive performance in predicting survival rates and immunotherapy efficacy of BCa was developed.
Collapse
Affiliation(s)
- Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhi-Bin Ke
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jia-Yin Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Hai Cai
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yun-Zhi Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiao-Dong Li
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
16
|
Xiao P, Tao X, Wang H, Liu H, Feng Y, Zhu Y, Jiang Z, Yin T, Zhang Y, He H, Gou J, Tang X. Enzyme/pH dual stimuli-responsive nanoplatform co-deliver disulfiram and doxorubicin for effective treatment of breast cancer lung metastasis. Expert Opin Drug Deliv 2023; 20:1015-1031. [PMID: 37452715 DOI: 10.1080/17425247.2023.2237888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/20/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVES Metastasis is still one of the main obstacles in the treatment of breast cancer. This study aimed to develop disulfiram (DSF) and doxorubicin (DOX) co-loaded nanoparticles (DSF-DOX NPs) with enzyme/pH dual stimuli-responsive characteristics to inhibit breast cancer metastasis. METHODS DSF-DOX NPs were prepared using the amphiphilic poly(ε-caprolactone)-b-poly(L-glutamic acid)-g-methoxy poly(ethylene glycol) (PCL-b-PGlu-g-mPEG) copolymer by a classical dialysis method. In vitro release tests, in vitro cytotoxicity assay, and anti-metastasis studies were conducted to evaluate pH/enzyme sensitivity and therapeutic effect of DSF-DOX NPs. RESULTS The specific pH and enzyme stimuli-responsiveness of DSF-DO NPs can be attributed to the transformation of secondary structure and the degradation of amide bonds in the PGlu segment, respectively. This accelerated drug release significantly increased the cytotoxicity to 4T1 cells. Compared with the control group, the DSF-DOX NPs showed a strong inhibition of in vitro metastasis with a wound healing rate of 36.50% and a migration rate of 18.39%. Impressively, in vivo anti-metastasis results indicated that the metastasis of 4T1 cells was almost completely suppressed by DSF-DOX NPs. CONCLUSION DSF-DOX NPs with controllable tumor site delivery of DOX and DSF were a prospectively potential strategy for metastatic breast cancer treatment.
Collapse
Affiliation(s)
- Peifu Xiao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoguang Tao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Hanxun Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, China
| | - Hongbing Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yupeng Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yueqi Zhu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhengzhen Jiang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
17
|
Zhang S, Zong Y, Chen L, Li Q, Li Z, Meng R. The immunomodulatory function and antitumor effect of disulfiram: paving the way for novel cancer therapeutics. Discov Oncol 2023; 14:103. [PMID: 37326784 DOI: 10.1007/s12672-023-00729-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/14/2023] [Indexed: 06/17/2023] Open
Abstract
More than 60 years ago, disulfiram (DSF) was employed for the management of alcohol addiction. This promising cancer therapeutic agent inhibits proliferation, migration, and invasion of malignant tumor cells. Furthermore, divalent copper ions can enhance the antitumor effects of DSF. Molecular structure, pharmacokinetics, signaling pathways, mechanisms of action and current clinical results of DSF are summarized here. Additionally, our attention is directed towards the immunomodulatory properties of DSF and we explore novel administration methods that may address the limitations associated with antitumor treatments based on DSF. Despite the promising potential of these various delivery methods for utilizing DSF as an effective anticancer agent, further investigation is essential in order to extensively evaluate the safety and efficacy of these delivery systems.
Collapse
Affiliation(s)
- Sijia Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Zong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Leichong Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qianwen Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhenyu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
18
|
Yuan M, Wu Q, Zhang M, Lai M, Chen W, Yang J, Jiang L, Cao J. Disulfiram enhances the antitumor activity of cisplatin by inhibiting the Fanconi anemia repair pathway. J Zhejiang Univ Sci B 2023; 24:207-220. [PMID: 36915997 PMCID: PMC10014319 DOI: 10.1631/jzus.b2200405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
A series of chemotherapeutic drugs that induce DNA damage, such as cisplatin (DDP), are standard clinical treatments for ovarian cancer, testicular cancer, and other diseases that lack effective targeted drug therapy. Drug resistance is one of the main factors limiting their application. Sensitizers can overcome the drug resistance of tumor cells, thereby enhancing the antitumor activity of chemotherapeutic drugs. In this study, we aimed to identify marketable drugs that could be potential chemotherapy sensitizers and explore the underlying mechanisms. We found that the alcohol withdrawal drug disulfiram (DSF) could significantly enhance the antitumor activity of DDP. JC-1 staining, propidium iodide (PI) staining, and western blotting confirmed that the combination of DSF and DDP could enhance the apoptosis of tumor cells. Subsequent RNA sequencing combined with Gene Set Enrichment Analysis (GSEA) pathway enrichment analysis and cell biology studies such as immunofluorescence suggested an underlying mechanism: DSF makes cells more vulnerable to DNA damage by inhibiting the Fanconi anemia (FA) repair pathway, exerting a sensitizing effect to DNA damaging agents including platinum chemotherapy drugs. Thus, our study illustrated the potential mechanism of action of DSF in enhancing the antitumor effect of DDP. This might provide an effective and safe solution for combating DDP resistance in clinical treatment.
Collapse
Affiliation(s)
- Meng Yuan
- Laboratory of Fruit Quality Biology / the State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Fruit Science Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Qian Wu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingyang Zhang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minshan Lai
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Polytechnic Institute, Zhejiang University, Hangzhou 310015, China
| | - Wenbo Chen
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Polytechnic Institute, Zhejiang University, Hangzhou 310015, China
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310006, China
| | - Li Jiang
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China.
| | - Ji Cao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China. .,The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China. .,Cancer Center of Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
19
|
Yuan XX, Duan YF, Luo C, Li L, Yang MJ, Liu TY, Cao ZR, Huang W, Bu X, Yue X, Liu RY. Disulfiram enhances cisplatin cytotoxicity by forming a novel platinum chelate Pt(DDTC) 3. Biochem Pharmacol 2023; 211:115498. [PMID: 36913990 DOI: 10.1016/j.bcp.2023.115498] [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: 11/21/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/15/2023]
Abstract
Despite the use of targeted therapy in non-small cell lung cancer (NSCLC) patients, cisplatin (DDP)-based chemotherapy is still the main option. However, DDP resistance is the major factor contributing to the failure of chemotherapy. In this study, we tried to screen DDP sensitizers from an FDA-approved drug library containing 1374 small-molecule drugs to overcome DDP resistance in NSCLC. As a result, disulfiram (DSF) was identified as a DDP sensitizer: DSF and DDP had synergistic anti-NSCLC effects, which are mainly reflected in inhibiting tumor cell proliferation, plate colony formation and 3D spheroidogenesis and inducing apoptosis in vitro, as well as the growth of NSCLC xenografts in mice. Although DSF has recently been reported to promote the antitumor effect of DDP by inhibiting ALDH activity or modulating some important factors or pathways, unexpectedly, we found that DSF reacted with DDP to form a new platinum chelate, Pt(DDTC)3+, which might be one of the important mechanisms for their synergistic effect. Moreover, Pt(DDTC)3+ has a stronger anti-NSCLC effect than DDP, and its antitumor activity is broad-spectrum. These findings reveal a novel mechanism underlying the synergistic antitumor effect of DDP and DSF, and provide a drug candidate or a lead compound for the development of a new antitumor drug.
Collapse
Affiliation(s)
- Xue-Xia Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - You-Fa Duan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chunxiang Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Lu Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Meng-Jie Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Ting-Yu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhi-Rui Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenlin Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Tumor Targeted Drugs & Guangzhou Enterprise Key Laboratory of Gene Medicine, Guangzhou DoublleBioproduct Co., Ltd., Guangzhou 510535, China
| | - Xianzhang Bu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xin Yue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Institute of Precision Medicine, The First Affiliated Hospital, SunYat-sen University, Guangzhou 510080, China.
| | - Ran-Yi Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
20
|
Selim MS, Kassem AB, El-Bassiouny NA, Salahuddin A, Abu El-Ela RY, Hamza MS. Polymorphic renal transporters and cisplatin's toxicity in urinary bladder cancer patients: current perspectives and future directions. Med Oncol 2023; 40:80. [PMID: 36650399 PMCID: PMC9845168 DOI: 10.1007/s12032-022-01928-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/10/2022] [Indexed: 01/19/2023]
Abstract
Urinary bladder cancer (UBC) holds a potentially profound social burden and affects over 573,278 new cases annually. The disease's primary risk factors include occupational tobacco smoke exposure and inherited genetic susceptibility. Over the past 30 years, a number of treatment modalities have emerged, including cisplatin, a platinum molecule that has demonstrated effectiveness against UBC. Nevertheless, it has severe dose-limiting side effects, such as nephrotoxicity, among others. Since intracellular accumulation of platinum anticancer drugs is necessary for cytotoxicity, decreased uptake or enhanced efflux are the root causes of platinum resistance and response failure. Evidence suggests that genetic variations in any transporter involved in the entry or efflux of platinum drugs alter their kinetics and, to a significant extent, determine patients' responses to them. This review aims to consolidate and describe the major transporters and their polymorphic variants in relation to cisplatin-induced toxicities and resistance in UBC patients. We concluded that the efflux transporters ABCB1, ABCC2, SLC25A21, ATP7A, and the uptake transporter OCT2, as well as the organic anion uptake transporters OAT1 and OAT2, are linked to cisplatin accumulation, toxicity, and resistance in urinary bladder cancer patients. While suppressing the CTR1 gene's expression reduced cisplatin-induced nephrotoxicity and ototoxicity, inhibiting the expression of the MATE1 and MATE2-K genes has been shown to increase cisplatin's nephrotoxicity and resistance. The roles of ABCC5, ABCA8, ABCC10, ABCB10, ABCG1, ATP7B, ABCG2, and mitochondrial SLC25A10 in platinum-receiving urinary bladder cancer patients should be the subject of further investigation.
Collapse
Affiliation(s)
- Mohamed S Selim
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt.
| | - Amira B Kassem
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Noha A El-Bassiouny
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Ahmad Salahuddin
- Biochemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
- Biochemistry Department, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Raghda Y Abu El-Ela
- Medical Oncology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Marwa Samir Hamza
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
21
|
Nunes M, Duarte D, Vale N, Ricardo S. The Antineoplastic Effect of Carboplatin Is Potentiated by Combination with Pitavastatin or Metformin in a Chemoresistant High-Grade Serous Carcinoma Cell Line. Int J Mol Sci 2022; 24:ijms24010097. [PMID: 36613537 PMCID: PMC9820586 DOI: 10.3390/ijms24010097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The combination of Carboplatin with Paclitaxel is the mainstay treatment for high-grade serous carcinoma; however, many patients with advanced disease undergo relapse due to chemoresistance. Drug repurposing coupled with a combination of two or more compounds with independent mechanisms of action has the potential to increase the success rate of the antineoplastic treatment. The purpose of this study was to explore whether the combination of Carboplatin with repurposed drugs led to a therapeutic benefit. Hence, we assessed the cytotoxic effects of Carboplatin alone and in combination with several repurposed drugs (Pitavastatin, Metformin, Ivermectin, Itraconazole and Alendronate) in two tumoral models, i.e., Carboplatin (OVCAR8) and Carboplatin-Paclitaxel (OVCAR8 PTX R P) chemoresistant cell lines and in a non-tumoral (HOSE6.3) cell line. Cellular viability was measured using the Presto Blue assay, and the synergistic interactions were evaluated using the Chou-Talalay, Bliss Independence and Highest Single Agent reference models. Combining Carboplatin with Pitavastatin or Metformin displayed the highest cytotoxic effect and the strongest synergism among all combinations for OVCAR8 PTX R P cells, resulting in a chemotherapeutic effect superior to Carboplatin as a single agent. Concerning HOSE6.3 cells, combining Carboplatin with almost all the repurposed drugs demonstrated a safe pharmacological profile. Overall, we propose that Pitavastatin or Metformin could act synergistically in combination with Carboplatin for the management of high-grade serous carcinoma patients with a Carboplatin plus Paclitaxel resistance profile.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Diana Duarte
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal
- Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal
- Toxicology Research Unit (TOXRUN), University Institute of Health Sciences, Polytechnic and University Cooperative (CESPU), 4585-116 Gandra, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- Correspondence:
| |
Collapse
|
22
|
Zhong S, Shengyu Liu, Xin Shi, Zhang X, Li K, Liu G, Li L, Tao S, Zheng B, Sheng W, Ye Z, Xing Q, Zhai Q, Ren L, Wu Y, Bao Y. Disulfiram in glioma: Literature review of drug repurposing. Front Pharmacol 2022; 13:933655. [PMID: 36091753 PMCID: PMC9448899 DOI: 10.3389/fphar.2022.933655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Gliomas are the most common malignant brain tumors. High-grade gliomas, represented by glioblastoma multiforme (GBM), have a poor prognosis and are prone to recurrence. The standard treatment strategy is tumor removal combined with radiotherapy and chemotherapy, such as temozolomide (TMZ). However, even after conventional treatment, they still have a high recurrence rate, resulting in an increasing demand for effective anti-glioma drugs. Drug repurposing is a method of reusing drugs that have already been widely approved for new indication. It has the advantages of reduced research cost, safety, and increased efficiency. Disulfiram (DSF), originally approved for alcohol dependence, has been repurposed for adjuvant chemotherapy in glioma. This article reviews the drug repurposing method and the progress of research on disulfiram reuse for glioma treatment.
Collapse
|
23
|
Disulfiram: A Food and Drug Administration-approved multifunctional role in synergistically drug delivery systems for tumor treatment. Int J Pharm 2022; 626:122130. [PMID: 36007849 DOI: 10.1016/j.ijpharm.2022.122130] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/27/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Disulfiram (DSF), a Food and Drug Administration (FDA)-approved drug for the treatment of alcoholism, has been found to have antitumor activity. DSF showed better antitumor efficiency when it was used in combination with certain antitumor drugs. DSF plays an important role in cancer treatment. It has been used as multidrug resistance (MDR) modulator to reverse MDR and can also combine with copper ions (Cu2+), which will produce copper diethyldithiocarbamate (Cu[DDC]2) complex with antitumor activity. The synergistic targeted drug delivery for cancer treatment based on DSF, especially the combination with exogenous Cu2+ and its forms of administration, has attracted extensive attention in the biomedical field. In this review, we summarize these synergistic delivery systems, in the hope that they will contribute to the continuous optimization and development of more advanced drug delivery systems. Furthermore, we discuss the current limitation and future directions of DSF-based drug delivery systems in the field of tumor therapy. Hopefully, our work may inspire further innovation of DSF-based antitumor drug delivery systems.
Collapse
|
24
|
Solovieva M, Shatalin Y, Odinokova I, Krestinina O, Baburina Y, Mishukov A, Lomovskaya Y, Pavlik L, Mikheeva I, Holmuhamedov E, Akatov V. Disulfiram oxy-derivatives induce entosis or paraptosis-like death in breast cancer MCF-7 cells depending on the duration of treatment. Biochim Biophys Acta Gen Subj 2022; 1866:130184. [PMID: 35660414 DOI: 10.1016/j.bbagen.2022.130184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/26/2022] [Accepted: 05/28/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Dithiocarbamates and derivatives (including disulfiram, DSF) are currently investigated as antineoplastic agents. We have revealed earlier the ability of hydroxocobalamin (vitamin В12b) combined with diethyldithiocarbamate (DDC) to catalyze the formation of highly cytotoxic oxidized derivatives of DSF (DSFoxy, sulfones and sulfoxides). METHODS Electron and fluorescent confocal microscopy, molecular biology and conventional biochemical techniques were used to study the morphological and functional responses of MCF-7 human breast cancer cells to treatment with DDC and B12b alone or in combination. RESULTS DDC induces unfolded protein response in MCF-7 cells. The combined use of DDC and B12b causes MCF-7 cell death. Electron microscopy revealed the separation of ER and nuclear membranes, leading to the formation of both cytoplasmic and perinuclear vacuoles, with many fibers inside. The process of vacuolization coincided with the appearance of ER stress markers, a marked damage to mitochondria, a significant inhibition of 20S proteasome, and actin depolimerization at later stages. Specific inhibitors of apoptosis, necroptosis, autophagy, and ferroptosis did not prevent cell death. A short- time (6-h) exposure to DSFoxy caused a significant increase in the number of entotic cells. CONCLUSIONS These observations indicate that MCF-7 cells treated with a mixture of DDC and B12b die by the mechanism of paraptosis. A short- time exposure to DSFoxy caused, along with paraptosis, a significant activation of the entosis and its final stage, lysosomal cell death. GENERAL SIGNIFICANCE The results obtained open up opportunities for the development of new approaches to induce non-apoptotic death of cancer cells by dithiocarbamates.
Collapse
Affiliation(s)
- Marina Solovieva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Yuri Shatalin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia.
| | - Irina Odinokova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Olga Krestinina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Yulia Baburina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Artem Mishukov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia; Laboratory of Biorheology and Biomechanics, Center for Theoretical Problems of Physicochemical Pharmacology RAS, Moscow 109029, Russian Federation
| | - Yana Lomovskaya
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Liubov Pavlik
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Irina Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| | - Ekhson Holmuhamedov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia; Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Vladimir Akatov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region 142290, Russia
| |
Collapse
|
25
|
Sano T, Fukui T, Makita N, Shimizu K, Kono J, Masui K, Sato T, Goto T, Sawada A, Fujimoto M, Kojima F, Torishima M, Wada T, Furuya M, Ogawa O, Kobayashi T, Akamatsu S. A novel missense mutation in the folliculin gene associated with the renal tumor-only phenotype of Birt-Hogg-Dubé syndrome. Cancer Genet 2022; 266-267:28-32. [DOI: 10.1016/j.cancergen.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/30/2022] [Accepted: 06/04/2022] [Indexed: 11/29/2022]
|
26
|
Tang XY, Wu S, Wang D, Chu C, Hong Y, Tao M, Hu H, Xu M, Guo X, Liu Y. Human organoids in basic research and clinical applications. Signal Transduct Target Ther 2022; 7:168. [PMID: 35610212 PMCID: PMC9127490 DOI: 10.1038/s41392-022-01024-9] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Organoids are three-dimensional (3D) miniature structures cultured in vitro produced from either human pluripotent stem cells (hPSCs) or adult stem cells (AdSCs) derived from healthy individuals or patients that recapitulate the cellular heterogeneity, structure, and functions of human organs. The advent of human 3D organoid systems is now possible to allow remarkably detailed observation of stem cell morphogens, maintenance and differentiation resemble primary tissues, enhancing the potential to study both human physiology and developmental stage. As they are similar to their original organs and carry human genetic information, organoids derived from patient hold great promise for biomedical research and preclinical drug testing and is currently used for personalized, regenerative medicine, gene repair and transplantation therapy. In recent decades, researchers have succeeded in generating various types of organoids mimicking in vivo organs. Herein, we provide an update on current in vitro differentiation technologies of brain, retinal, kidney, liver, lung, gastrointestinal, cardiac, vascularized and multi-lineage organoids, discuss the differences between PSC- and AdSC-derived organoids, summarize the potential applications of stem cell-derived organoids systems in the laboratory and clinic, and outline the current challenges for the application of organoids, which would deepen the understanding of mechanisms of human development and enhance further utility of organoids in basic research and clinical studies.
Collapse
Affiliation(s)
- Xiao-Yan Tang
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Shanshan Wu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Da Wang
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Chu Chu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Yuan Hong
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Mengdan Tao
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Hao Hu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Min Xu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Xing Guo
- Department of Neurobiology, School of Basic Medical Sciences; Nanjing Medical University, Nanjing, China.
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China.
| |
Collapse
|
27
|
Medle B, Sjödahl G, Eriksson P, Liedberg F, Höglund M, Bernardo C. Patient-Derived Bladder Cancer Organoid Models in Tumor Biology and Drug Testing: A Systematic Review. Cancers (Basel) 2022; 14:cancers14092062. [PMID: 35565191 PMCID: PMC9104249 DOI: 10.3390/cancers14092062] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/07/2022] [Accepted: 04/15/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Primary culture of cancer cells from patient tumors in a physiologically relevant system can provide information about tumor biology, disentangle the role of different cell types within the tumors, and give information about drug sensitivity for the development of cancer-targeted therapies and precision medicine. This requires the use of well-characterized and easily expandable tumor models. This review focuses on 3D models developed from primary human tissue including normal urothelium or bladder cancer samples, the characteristics of the models, and to what extent the organoids represent the diversity observed among human tumors. Abstract Bladder cancer is a common and highly heterogeneous malignancy with a relatively poor outcome. Patient-derived tumor organoid cultures have emerged as a preclinical model with improved biomimicity. However, the impact of the different methods being used in the composition and dynamics of the models remains unknown. This study aims to systematically review the literature regarding patient-derived organoid models for normal and cancer tissue of the bladder, and their current and potential future applications for tumor biology studies and drug testing. A PRISMA-compliant systematic review of the PubMED, Embase, Web of Sciences, and Scopus databases was performed. The results were analyzed based on the methodologies, comparison with primary tumors, functional analysis, and chemotherapy and immunotherapy testing. The literature search identified 536 articles, 24 of which met the inclusion criteria. Bladder cancer organoid models have been increasingly used for tumor biology studies and drug screening. Despite the heterogeneity between methods, organoids and primary tissues showed high genetic and phenotypic concordance. Organoid sensitivity to chemotherapy matched the response in patient-derived xenograft (PDX) models and predicted response based on clinical and mutation data. Advances in bioengineering technology, such as microfluidic devices, bioprinters, and imaging, are likely to further standardize and expand the use of organoids.
Collapse
Affiliation(s)
- Benjamin Medle
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden; (B.M.); (P.E.); (M.H.)
| | - Gottfrid Sjödahl
- Division of Clinical and Experimental Urothelial Carcinoma Research, Department of Translational Medicine, Lund University, Malmö and Department of Urology, Skåne University Hospital, Jan Waldenströms Gata 5, 205 02 Malmö, Sweden; (G.S.); (F.L.)
| | - Pontus Eriksson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden; (B.M.); (P.E.); (M.H.)
| | - Fredrik Liedberg
- Division of Clinical and Experimental Urothelial Carcinoma Research, Department of Translational Medicine, Lund University, Malmö and Department of Urology, Skåne University Hospital, Jan Waldenströms Gata 5, 205 02 Malmö, Sweden; (G.S.); (F.L.)
| | - Mattias Höglund
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden; (B.M.); (P.E.); (M.H.)
| | - Carina Bernardo
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden; (B.M.); (P.E.); (M.H.)
- Correspondence: ; Tel.: +46-73-032-48-78
| |
Collapse
|
28
|
Sun F, Wang H, Nie J, Hong B. Repurposing disulfiram as a chemo-therapeutic sensitizer: molecular targets and mechanisms. Anticancer Agents Med Chem 2022; 22:2920-2926. [PMID: 35430981 DOI: 10.2174/1871520621666220415102553] [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: 10/14/2021] [Revised: 12/20/2021] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
Currently, chemo-therapy is still the main strategy for cancer treatment. However, chemo-therapy resistance remains its main challenge. Disulfiram [DSF] is a drug approved by FDA for the treatment of alcohol addiction, but it is later discovered that it has the anticancer activity. Importantly, there have been many literatures reporting that DSF can be used as a chemo-therapeutic sensitizer to enhance the anticancer activity of chemo-drugs in a variety of cancers. Furthermore, the combinations of DSF and chemo-drugs have been tested in clinic trials. In the review, we summarized the possible molecular targets and mechanisms of DSF to reverse chemo-resistance. We also further discussed the opportunities and challenges of DSF as a chemo-therapeutic sensitizer. In conclusion, DSF could be a potential repurposed drug to sensitize cancer cells to chemo-therapy in clinic.
Collapse
Affiliation(s)
- Feilong Sun
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Hefei, Anhui, China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jinfu Nie
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Bo Hong
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, China
| |
Collapse
|
29
|
Abedi Z, MotieGhader H, Hosseini SS, Sheikh Beig Goharrizi MA, Masoudi-Nejad A. mRNA-miRNA bipartite networks reconstruction in different tissues of bladder cancer based on gene co-expression network analysis. Sci Rep 2022; 12:5885. [PMID: 35393513 PMCID: PMC8991185 DOI: 10.1038/s41598-022-09920-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/24/2022] [Indexed: 12/14/2022] Open
Abstract
Bladder cancer (BC) is one of the most important cancers worldwide, and if it is diagnosed early, its progression in humans can be prevented and long-term survival will be achieved accordingly. This study aimed to identify novel micro-RNA (miRNA) and gene-based biomarkers for diagnosing BC. The microarray dataset of BC tissues (GSE13507) listed in the GEO database was analyzed for this purpose. The gene expression data from three BC tissues including 165 primary bladder cancer (PBC), 58 normal looking-bladder mucosae surrounding cancer (NBMSC), and 23 recurrent non-muscle invasive tumor tissues (RNIT) were used to reconstruct gene co-expression networks. After preprocessing and normalization, deferentially expressed genes (DEGs) were obtained and used to construct the weighted gene co-expression network (WGCNA). Gene co-expression modules and low-preserved modules were extracted among BC tissues using network clustering. Next, the experimentally validated mRNA-miRNA interaction information were used to reconstruct three mRNA-miRNA bipartite networks. Reactome pathway database and Gene ontology (GO) was subsequently performed for the extracted genes of three bipartite networks and miRNAs, respectively. To further analyze the data, ten hub miRNAs (miRNAs with the highest degree) were selected in each bipartite network to reconstruct three bipartite subnetworks. Finally, the obtained biomarkers were comprehensively investigated and discussed in authentic studies. The obtained results from our study indicated a group of genes including PPARD, CST4, CSNK1E, PTPN14, ETV6, and ADRM1 as well as novel miRNAs (e.g., miR-16-5p, miR-335-5p, miR-124-3p, and let-7b-5p) which might be potentially associated with BC and could be a potential biomarker. Afterward, three drug-gene interaction networks were reconstructed to explore candidate drugs for the treatment of BC. The hub miRNAs in the mRNA-miRNA bipartite network played a fundamental role in BC progression; however, these findings need further investigation.
Collapse
Affiliation(s)
- Zahra Abedi
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Habib MotieGhader
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | - Sahar Sadat Hosseini
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
30
|
Wang W, Liu X, Zhang C, Sheng F, Song S, Li P, Dai S, Wang B, Lu D, Zhang L, Yang X, Zhang Z, Liu S, Zhang A, Liu Q, Jiang G. Identification of two-dimensional copper signatures in human blood for bladder cancer with machine learning. Chem Sci 2022; 13:1648-1656. [PMID: 35282611 PMCID: PMC8826767 DOI: 10.1039/d1sc06156a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/11/2022] [Indexed: 12/26/2022] Open
Abstract
Currently, almost all available cancer biomarkers are based on concentrations of compounds, often suffering from low sensitivity, poor specificity, and false positive or negative results. The stable isotopic composition of elements provides a different dimension from the concentration and has been widely used as a tracer in geochemistry. In health research, stable isotopic analysis has also shown potential as a new diagnostic/prognostic tool, which is still in the nascent stage. Here we discovered that bladder cancer (BCa) could induce a significant variation in the ratio of natural copper isotopes (65Cu/63Cu) in the blood of patients relative to benign and healthy controls. Such inherent copper isotopic signatures permitted new insights into molecular mechanisms of copper imbalance underlying the carcinogenic process. More importantly, to enhance the diagnostic capability, a machine learning model was developed to classify BCa and non-BCa subjects based on two-dimensional copper signatures (copper isotopic composition and concentration in plasma and red blood cells) with a high sensitivity, high true negative rate, and low false positive rate. Our results demonstrated the promise of blood copper signatures combined with machine learning as a versatile tool for cancer research and potential clinical application. The blood Cu concentration and isotopic composition enable new insights into Cu imbalance and diagnosis of bladder cancer with machine learning.![]()
Collapse
Affiliation(s)
- Weichao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Xian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China
| | - Changwen Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin 300211 China
| | - Fei Sheng
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin 300211 China
| | - Shanjun Song
- National Institute of Metrology Beijing 100029 China
| | - Penghui Li
- Tianjin University of Technology Tianjin 300384 China
| | - Shaoqing Dai
- Faculty of Geo-Information Science and Earth Observation (ITC), University of Twente P.O. Box 217 7500AE Enschede The Netherlands
| | - Bin Wang
- Institute of Reproductive and Child Health, National Health Commission's Key Laboratory of Reproductive Health, Peking University Beijing 100191 China
| | - Dawei Lu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Luyao Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Xuezhi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Zhihong Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin 300211 China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China
| | - Aiqian Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China .,University of Chinese Academy of Sciences Beijing 100049 China
| | - Qian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China .,Institute of Environment and Health, Jianghan University Wuhan 430056 China.,University of Chinese Academy of Sciences Beijing 100049 China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences Beijing 100085 China .,University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
31
|
Leveraging disulfiram to treat cancer: Mechanisms of action, delivery strategies, and treatment regimens. Biomaterials 2021; 281:121335. [PMID: 34979419 DOI: 10.1016/j.biomaterials.2021.121335] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/07/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023]
Abstract
Disulfiram (DSF) has been used as an alcoholism drug for 70 years. Recently, it has attracted increasing attention owing to the distinguished anticancer activity, which can be further potentiated by the supplementation of Cu2+. Although encouraging anticancer results are obtained in lab, the clinical outcomes of oral DSF are not satisfactory, which urges an in-depth understanding of the underlying mechanisms, bottlenecks, and proposal of potential methods to address the dilemma. In this review, a critical summarization of various molecular biological anticancer mechanisms of DSF/Cu2+ is provided and the predicament of orally delivering DSF in clinical oncotherapy is explained by the metabolic barriers. We highlight the recent advances in the DSF/Cu2+ delivery strategies and the emerging treatment regimens for cancer treatment. Last but not the least, we summarize the clinical trials regarding DSF and make a prospect of DSF/Cu-based cancer therapy.
Collapse
|
32
|
Zhao S, Li P, Wu W, Wang Q, Qian B, Li X, Shen M. Roles of ferroptosis in urologic malignancies. Cancer Cell Int 2021; 21:676. [PMID: 34922551 PMCID: PMC8684233 DOI: 10.1186/s12935-021-02264-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic cell death, is believed to strongly contribute to the pathogenesis of multiple cancers. Recently, the positive association between ferroptosis and urologic malignancies has drawn considerable attention, while a comprehensive review focused on this issue is absent. Based on this review, ferroptosis has been implicated in the development and therapeutic responses of prostate cancer, kidney cancer, and bladder cancer. Mechanistically, a large number of biomolecules and tumor-associated signaling pathways, including DECR1, PANX2, HSPB1, ACOT8, SUV39H1, NCOA4, PI3K-AKT-mTOR signaling, VHL/HIF-2α pathway, and Hippo/TAZ signaling pathway, have been reported to regulate ferroptosis in urologic cancers. Ferroptosis inducers, such as erastin, ART, CPNPs, and quinazolinyl-arylurea derivatives, exert potential therapeutic effects per se and/or enhance the anticancer response of other anticancer drugs in urologic oncology. A better understanding of ferroptosis may provide a promising way to treat therapy-resistant urologic cancers.
Collapse
Affiliation(s)
- Shankun Zhao
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China
| | - Peng Li
- Department of Urology, Qingdao Women and Children's Hospital, Qingdao, 266000, Shandong, China
| | - Weizhou Wu
- Department of Urology, Maoming People's Hospital, Maoming, 525000, Guangdong, China
| | - Qinzhang Wang
- Department of Urology, The First Affiliated Hospital of Shihezi University Medical School, Shihezi, China
| | - Biao Qian
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Xin Li
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China.
| | - Maolei Shen
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China.
| |
Collapse
|
33
|
Liu S, Chen X, Lin T. Emerging strategies for the improvement of chemotherapy in bladder cancer: Current knowledge and future perspectives. J Adv Res 2021; 39:187-202. [PMID: 35777908 PMCID: PMC9263750 DOI: 10.1016/j.jare.2021.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 11/24/2022] Open
Abstract
The response of chemotherapy and prognosis in bladder cancer is unsatisfied. Immunotherapy, targeted therapy, and ADC improve the efficacy of chemotherapy. Emerging targets in cancer cells and TME spawned novel preclinical agents. Novel drug delivery, such as nanotechnology, enhances effects of chemotherapeutics. The organoid and PDX model are promising to screen and evaluate the target therapy.
Background Chemotherapy is a first-line treatment for advanced and metastatic bladder cancer, but the unsatisfactory objective response rate to this treatment yields poor 5-year patient survival. Only PD-1/PD-L1-based immune checkpoint inhibitors, FGFR3 inhibitors and antibody-drug conjugates are approved by the FDA to be used in bladder cancer, mainly for platinum-refractory or platinum-ineligible locally advanced or metastatic urothelial carcinoma. Emerging studies indicate that the combination of targeted therapy and chemotherapy shows better efficacy than targeted therapy or chemotherapy alone. Newly identified targets in cancer cells and various functions of the tumour microenvironment have spawned novel agents and regimens, which give impetus to sensitizing chemotherapy in the bladder cancer setting. Aim of Review This review aims to present the current evidence for potentiating the efficacy of chemotherapy in bladder cancer. We focus on combining chemotherapy with other treatments as follows: targeted therapy, including immunotherapy and antibody-drug conjugates in clinic; novel targeted drugs and nanoparticles in preclinical models and potential targets that may contribute to chemosensitivity in future clinical practice. The prospect of precision therapy is also discussed in bladder cancer. Key Scientific Concepts of Review Combining chemotherapy drugs with immune checkpoint inhibitors, antibody-drug conjugates and VEGF inhibitors potentially elevates the response rate and survival. Novel targets, including cancer stem cells, DNA damage repair, antiapoptosis, drug metabolism and the tumour microenvironment, contribute to chemosensitization. Gene alteration-based drug selection and patient-derived xenograft- and organoid-based drug validation are the future for precision therapy.
Collapse
|
34
|
Li Z, Xie X, Tan G, Xie F, Liu N, Li W, Sun X. Disulfiram Synergizes with SRC Inhibitors to Suppress the Growth of Pancreatic Ductal Adenocarcinoma Cells in Vitro and in Vivo. Biol Pharm Bull 2021; 44:1323-1331. [PMID: 34471060 DOI: 10.1248/bpb.b21-00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Disulfiram (DSF), an old anti-alcoholism drug, has emerged as a candidate for drug repurposing in oncology. In exploratory studies on its therapeutic effects, we unexpectedly discovered that DSF increased the phosphorylation of SRC, a proto-oncogene tyrosine-protein kinase elevated in 70% of pancreatic ductal adenocarcinoma (PDAC) cases. This serendipitous and novel finding led to our hypothesis for the current study which proposes DSF may synergize with SRC inhibitors in suppressing PDAC. Human PDAC PANC-1 and BXPC-3 cells were incubated with DSF chelated with copper (Cu2+), SRC inhibitors (PP2 and dasatinib), or transfected with lentiviral short hairpin RNA (shRNA), and their proliferation and apoptosis were analyzed. A xenograft model was employed to verify the in vitro results. The expression of key molecules was detected. DSF significantly inhibited cell proliferation and induced cell apoptosis by increasing the cleavage of poly ADP ribose polymerase (PARP), downregulating Bcl-2 and upregulating p27 in concentration- and time-dependent manners. DSF had little effect on signal transducer and activator of transcription 3 (STAT3) expression but inhibited its phosphorylation. DSF did not alter SRC expression but significantly increased its phosphorylation through upregulating actin filament associated protein 1 like 2 (AFAP1L2). DSF exhibited a synergistic effect, as analyzed by drug coefficient interactions, with either PP2, or dasatinib, or SRC depletion in suppressing PDAC cells in vitro and/or in vivo. The present results indicate DSF is a potential therapeutic drug, particularly when it is combined with SRC inhibitors, and warrant further studies on the pharmacological utility of DSF as a promising adjunct therapy for the treatment of PDAC.
Collapse
Affiliation(s)
- Ziyi Li
- Hepatosplenic Surgery Center, the First Affiliated Hospital of Harbin Medical University
| | - Xiangjun Xie
- Department of Gastroenterology, Qingdao Municipal Hospital Affiliated to Qingdao University
| | - Gang Tan
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University
| | - Fangyu Xie
- Department of Cardiology, Qingdao Municipal Hospital Affiliated to Qingdao University
| | - Nianjiao Liu
- Department of Endocrinology, the First Hospital Affiliated of Harbin Medical University
| | - Weidong Li
- Hepatosplenic Surgery Center, the First Affiliated Hospital of Harbin Medical University.,Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University
| | - Xueying Sun
- Hepatosplenic Surgery Center, the First Affiliated Hospital of Harbin Medical University
| |
Collapse
|
35
|
Babak MV, Ahn D. Modulation of Intracellular Copper Levels as the Mechanism of Action of Anticancer Copper Complexes: Clinical Relevance. Biomedicines 2021; 9:biomedicines9080852. [PMID: 34440056 PMCID: PMC8389626 DOI: 10.3390/biomedicines9080852] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
Copper (Cu) is a vital element required for cellular growth and development; however, even slight changes in its homeostasis might lead to severe toxicity and deleterious medical conditions. Cancer patients are typically associated with higher Cu content in serum and tumor tissues, indicating increased demand of cancer cells for this micronutrient. Cu is known to readily cycle between the +1 and +2 oxidation state in biological systems. The mechanism of action of Cu complexes is typically based on their redox activity and induction of reactive oxygen species (ROS), leading to deadly oxidative stress. However, there are a number of other biomolecular mechanisms beyond ROS generation that contribute to the activity of anticancer Cu drug candidates. In this review, we discuss how interfering with intracellular Cu balance via either diet modification or addition of inorganic Cu supplements or Cu-modulating compounds affects tumor development, progression, and sensitivity to treatment modalities. We aim to provide the rationale for the use of Cu-depleting and Cu-overloading conditions to generate the best possible patient outcome with minimal toxicity. We also discuss the advantages of the use of pre-formed Cu complexes, such as Cu-(bis)thiosemicarbazones or Cu-N-heterocyclic thiosemicarbazones, in comparison with the in situ formed Cu complexes with metal-binding ligands. In this review, we summarize available clinical and mechanistic data on clinically relevant anticancer drug candidates, including Cu supplements, Cu chelators, Cu ionophores, and Cu complexes.
Collapse
|
36
|
Murakami K, Kita Y, Sakatani T, Hamada A, Mizuno K, Nakamura K, Takada H, Matsumoto K, Sano T, Goto T, Akamatsu S, Saito R, Tsuruyama T, Ogawa O, Kobayashi T. Antitumor effect of WEE1 blockade as monotherapy or in combination with cisplatin in urothelial cancer. Cancer Sci 2021; 112:3669-3681. [PMID: 34212455 PMCID: PMC8409401 DOI: 10.1111/cas.15051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Overcoming cisplatin (CDDP) resistance is a major issue in urothelial cancer (UC), in which CDDP‐based chemotherapy is the first‐line treatment. WEE1, a G2/M checkpoint kinase, confers chemoresistance in response to genotoxic agents. However, the efficacy of WEE1 blockade in UC has not been reported. MK‐1775, a WEE1 inhibitor also known as AZD‐1775, blocked proliferation of UC cell lines in a dose‐dependent manner irrespective of TP53 status. MK‐1775 synergized with CDDP to block proliferation, inducing apoptosis and mitotic catastrophe in TP53‐mutant UC cells but not in TP53‐WT cells. Knocking down TP53 in TP53‐WT cells induced synergism of MK‐1775 and CDDP. In UMUC3 cell xenografts and two patient‐derived xenograft lines with MDM2 overexpression, in which the p53/cell cycle pathway was inactivated, AZD‐1775 combined with CDDP suppressed tumor growth inducing both M‐phase entry and apoptosis, whereas AZD‐1775 alone was as effective as the combination in RT4 cell xenografts. Drug susceptibility assay using an ex vivo cancer tissue‐originated spheroid system showed correlations with the in vivo efficacy of AZD‐1775 alone or combined with CDDP. We determined the feasibility of the drug susceptibility assay using spheroids established from UC surgical specimens obtained by transurethral resection. In conclusion, WEE1 is a promising therapeutic target in the treatment of UC, and a highly specific small molecule inhibitor is currently in early phase clinical trials for cancer. Differential antitumor efficacy of WEE1 blockade alone or combined with CDDP could exist according to p53/cell cycle pathway activity, which might be predictable using an ex vivo 3D primary culture system.
Collapse
Affiliation(s)
- Kaoru Murakami
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Kita
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toru Sakatani
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihiro Hamada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kei Mizuno
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Nakamura
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Takada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Keiyu Matsumoto
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Sano
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayuki Goto
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shusuke Akamatsu
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryoichi Saito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tatsuaki Tsuruyama
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
37
|
Kronemberger GS, Miranda GASC, Tavares RSN, Montenegro B, Kopke ÚDA, Baptista LS. Recapitulating Tumorigenesis in vitro: Opportunities and Challenges of 3D Bioprinting. Front Bioeng Biotechnol 2021; 9:682498. [PMID: 34239860 PMCID: PMC8258101 DOI: 10.3389/fbioe.2021.682498] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is considered one of the most predominant diseases in the world and one of the principal causes of mortality per year. The cellular and molecular mechanisms involved in the development and establishment of solid tumors can be defined as tumorigenesis. Recent technological advances in the 3D cell culture field have enabled the recapitulation of tumorigenesis in vitro, including the complexity of stromal microenvironment. The establishment of these 3D solid tumor models has a crucial role in personalized medicine and drug discovery. Recently, spheroids and organoids are being largely explored as 3D solid tumor models for recreating tumorigenesis in vitro. In spheroids, the solid tumor can be recreated from cancer cells, cancer stem cells, stromal and immune cell lineages. Organoids must be derived from tumor biopsies, including cancer and cancer stem cells. Both models are considered as a suitable model for drug assessment and high-throughput screening. The main advantages of 3D bioprinting are its ability to engineer complex and controllable 3D tissue models in a higher resolution. Although 3D bioprinting represents a promising technology, main challenges need to be addressed to improve the results in cancer research. The aim of this review is to explore (1) the principal cell components and extracellular matrix composition of solid tumor microenvironment; (2) the recapitulation of tumorigenesis in vitro using spheroids and organoids as 3D culture models; and (3) the opportunities, challenges, and applications of 3D bioprinting in this area.
Collapse
Affiliation(s)
- Gabriela S. Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
| | - Guilherme A. S. C. Miranda
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Renata S. N. Tavares
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Bianca Montenegro
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
| | - Úrsula de A. Kopke
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Leandra S. Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro Xerém, Duque de Caxias, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
- Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| |
Collapse
|
38
|
Kuo MT, Huang YF, Chou CY, Chen HHW. Targeting the Copper Transport System to Improve Treatment Efficacies of Platinum-Containing Drugs in Cancer Chemotherapy. Pharmaceuticals (Basel) 2021; 14:ph14060549. [PMID: 34201235 PMCID: PMC8227247 DOI: 10.3390/ph14060549] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022] Open
Abstract
The platinum (Pt)-containing antitumor drugs including cisplatin (cis-diamminedichloroplatinum II, cDDP), carboplatin, and oxaliplatin, have been the mainstay of cancer chemotherapy. These drugs are effective in treating many human malignancies. The major cell-killing target of Pt drugs is DNA. Recent findings underscored the important roles of Pt drug transport system in cancer therapy. While many mechanisms have been proposed for Pt-drug transport, the high-affinity copper transporter (hCtr1), Cu chaperone (Atox1), and Cu exporters (ATP7A and ATP7B) are also involved in cDDP transport, highlighting Cu homeostasis regulation in Pt-based cancer therapy. It was demonstrated that by reducing cellular Cu bioavailable levels by Cu chelators, hCtr1 is transcriptionally upregulated by transcription factor Sp1, which binds the promoters of Sp1 and hCtr1. In contrast, elevated Cu poisons Sp1, resulting in suppression of hCtr1 and Sp1, constituting the Cu-Sp1-hCtr1 mutually regulatory loop. Clinical investigations using copper chelator (trientine) in carboplatin treatment have been conducted for overcoming Pt drug resistance due in part to defective transport. While results are encouraging, future development may include targeting multiple steps in Cu transport system for improving the efficacies of Pt-based cancer chemotherapy. The focus of this review is to delineate the mechanistic interrelationships between Cu homeostasis regulation and antitumor efficacy of Pt drugs.
Collapse
Affiliation(s)
- Macus Tien Kuo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yu-Fang Huang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Cheng-Yang Chou
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Correspondence: (C.-Y.C.); (H.H.W.C.)
| | - Helen H. W. Chen
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (C.-Y.C.); (H.H.W.C.)
| |
Collapse
|
39
|
Mokou M, Lygirou V, Angelioudaki I, Paschalidis N, Stroggilos R, Frantzi M, Latosinska A, Bamias A, Hoffmann MJ, Mischak H, Vlahou A. A Novel Pipeline for Drug Repurposing for Bladder Cancer Based on Patients' Omics Signatures. Cancers (Basel) 2020; 12:E3519. [PMID: 33255925 PMCID: PMC7759896 DOI: 10.3390/cancers12123519] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Multi-omics signatures of patients with bladder cancer (BC) can guide the identification of known de-risked therapeutic compounds through drug repurposing, an approach not extensively explored yet. In this study, we target drug repurposing in the context of BC, driven by tissue omics signatures. To identify compounds that can reverse aggressive high-risk Non-Muscle Invasive BC (NMIBC) to less aggressive low-risk molecular subtypes, the next generation Connectivity Map (CMap) was employed using as input previously published proteomics and transcriptomics respective signatures. Among the identified compounds, the ATP-competitive inhibitor of mTOR, WYE-354, showed a consistently very high score for reversing the aggressive BC molecular signatures. WYE-354 impact was assessed in a panel of eight multi-origin BC cell lines and included impaired colony growth and proliferation rate without any impact on apoptosis. Overall, with this study we introduce a promising pipeline for the repurposing of drugs for BC treatment, based on patients' omics signatures.
Collapse
Affiliation(s)
- Marika Mokou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (M.F.); (A.L.); (H.M.)
| | - Vasiliki Lygirou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| | - Ioanna Angelioudaki
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| | - Nikolaos Paschalidis
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Rafael Stroggilos
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| | - Maria Frantzi
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (M.F.); (A.L.); (H.M.)
| | | | - Aristotelis Bamias
- Haematology-Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Michèle J. Hoffmann
- Department of Urology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (M.F.); (A.L.); (H.M.)
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8QQ, UK
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| |
Collapse
|
40
|
Rieber M. Cancer Pro-oxidant Therapy Through Copper Redox Cycling: Repurposing Disulfiram and Tetrathiomolybdate. Curr Pharm Des 2020; 26:4461-4466. [DOI: 10.2174/1381612826666200628022113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Background:
Copper (Cu) is a transition metal active in Fenton redox cycling from reduced Cu+ and
H2O2, to oxidized Cu2+ and the hydroxyl radical (·OH) highly reactive oxygen species (ROS). At homeostatic Cu
levels, ROS promote cell proliferation, migration, angiogenesis, and wound repair. To limit ROS toxicity, cells
use Cu-dependent chaperone proteins, Cu-binding ceruloplasmin, and Cu-modulated enzymes like superoxide
dismutases (SOD) like SOD1 and SOD3 to scavenge excess superoxide anions which favour Cu+ reduction, and
mitochondrial cytochrome c oxidase, important in aerobic energy production. Because Cu helps drive tumor cell
proliferation by promoting growth factor-independent receptor tyrosine kinase signaling, and Cu-dependent
MEK1 involved in oncogenic BRAF-V600E signaling, further augmenting bioavailable Cu may promote ROS overproduction,
cancer progression and eventually tumor cell death. For these reasons, the following clinically approved
copper chelators are being repurposed as anti-cancer agents: a) ammonium tetrathiomolybdate (TTM)
used to treat Wilson’s disease (copper overload) and Menkes disease (copper deficiency); b) Disulfiram (DSF),
used against alcoholism, since it inhibits Aldehyde Dehydrogenase (ALDH1) enzyme, important in ethanol detoxification,
and a key target against cancer stem cells. Moreover, TTM and DSF are also relevant in cancer clinical
trials, because they increase the uptake of both Cu and Platinum (Pt)-containing anti-cancer drugs, since Pt
and Cu share the same CTR1 copper transporter.
Purpose:
The majority of reports on Cu chelators dealt separately with either TTM, DSF or others. Here, we
compare in parallel, the anti-cancer efficacy of low doses of TTM and DSF, asking whether they can be synergistic
or antagonistic. The relevance of their unequal ROS inducing abilities and their different behavior as ionophores
is also addressed.
Significance:
The potential of Cu chelators as repurposed anti-cancer drugs, should be greater in patients with
higher endogenous Cu levels. Since platinum and Cu share uptake receptors, the synergism by drugs containing
these metals should not be under-estimated. The potential of disulfiram or its metabolically active Cu-containing
form, to inhibit ALDH1-positive tumor cells is therapeutically very important.
Collapse
Affiliation(s)
- Manuel Rieber
- IVIC, Cancer Cell Biology Laboratory, CMBC, Caracas 1020A, Venezuela
| |
Collapse
|
41
|
Drug screening model meets cancer organoid technology. Transl Oncol 2020; 13:100840. [PMID: 32822897 PMCID: PMC7451679 DOI: 10.1016/j.tranon.2020.100840] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/12/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor organoids inherit the genomic and molecular characteristics of the donor tumor, which not only bridge the gap between genome and phenotype but also circumvent the disadvantages such as genetic information change by using 2D cell lines and the mouse-specific tumor evolution in patient-derived xenograft (PDX). So, cancer organoid has been widely applied to preclinical drug evaluation, biomarker identification, biological research, and individualized therapy. Besides, cancer organoid can be preserved, resuscitated, passed infinitely, and mechanically cultured on a chip for drug screening; it has become one of the partial models for low/high-throughput drug screening in the preclinical trial in vitro. Therefore, this review presents the recent developments of tumor organoids for drug screening, which will introduce from four aspects, including the stability/credibility, types, application, deficiency and prospect of the tumor organoids model for drug screening.
Collapse
|
42
|
Kita Y, Saito R, Inoue T, Kim WY, Ogawa O, Kobayashi T. Patient-Derived Urothelial Cancer Xenograft Models: A Systematic Review and Future Perspectives. Bladder Cancer 2020. [DOI: 10.3233/blc-200281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Lack of appropriate models that recapitulate the diversity, heterogeneity, and tumor microenvironment of urothelial cancer (UC) is a limitation to preclinical models. Patient-derived xenograft (PDX) models are a promising tool to overcome some of these issues, and thus we present an up-to-date and comprehensive overview of UC PDX models to aid in their future use. OBJECTIVE: To provide an overview on methodology, applications and limitations as well as future perspectives on bladder cancer PDX models. METHODS: Literature searches using PubMed and Web of Science databases were performed for relevant articles according to the following MeSH terms: “urothelial carcinoma(s)” OR “urothelial cancer” OR “urothelial tumor” OR “bladder cancer(s)” OR “bladder carcinoma(s)” OR “transitional cell carcinoma(s)” AND “xenograft(s)” OR “xenotransplant” at December 6th, 2019. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS: Of the 49 studies extracted, 41 studies after the year 2000 were finally analyzed. Published studies show that (1) UC PDX platforms retained the histology and genomic characteristics of the corresponding patient tumors. (2) UC PDX can be applied to ask various questions including to study the mechanisms of disease progression and treatment resistance, to develop novel drugs and biomarkers, as well as to potentially realize personalized drug selection. Recent topics of research using PDX have included the development of humanized mice as well as the use of 3D culture to complement some of the limitations of PDX models. CONCLUSIONS: UC PDX models serve as tools for understanding cancer biology, drug development and empowering precision medicine. The improvement of experimental systems using humanized mice to recapitulate the immune microenvironment of tumors will optimize UC PDX to study future questions in the field of immunotherapy.
Collapse
Affiliation(s)
- Yuki Kita
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ryoichi Saito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahiro Inoue
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - William Y. Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|