1
|
Wang H, Yang Q, Mao Y, Ding H, Huang P, Zhan Z. The efficacy and safety of hydroxychloroquine at different doses and courses for COVID-19 prevention: a systematic review and network meta-analysis. Expert Rev Anti Infect Ther 2024; 22:1209-1220. [PMID: 39365687 DOI: 10.1080/14787210.2024.2413419] [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: 06/21/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND The optimal strategy for using hydroxychloroquine to prevent coronavirus disease 2019 (COVID-19) in patients, either before or after exposure, remains unclear. We evaluated the safety and efficacy of different doses and treatment durations of hydroxychloroquine for COVID-19 prevention. METHOD Databases including PubMed, Web of Science, Cochrane Library, EMBASE, Medline, and ClinicalTrials.gov were systematically searched for randomized controlled trials (RCTs) comparing different doses of hydroxychloroquine for COVID-19 prevention, from their inception to February 2024. RESULTS A total of 20 RCTs involving 12,372 patients were included. Meta-analysis results showed no significant difference between the hydroxychloroquine and control groups in reducing the incidence of syndrome coronavirus type 2 (SARS-CoV-2) positivity (OR = 0.83, 95% CI = 0.67, 1.03). However, the subgroup receiving a daily dose of 200-400 mg (OR = 0.62, 95% CI = 0.51, 0.75) and a treatment duration of 5-8 weeks (OR = 0.52, 95% CI = 0.31, 0.88) showed statistically significant reductions in SARS-CoV-2 positivity. According to the surface under the cumulative ranking curve (SUCRA), the most effective intervention was a 200-400 mg dose for 5-8 weeks. . CONCLUSIONS A hydroxychloroquine dose of 200-400 mg for a duration of 5-8 weeks may moderately reduce the risk of COVID-19 with a relatively low risk of adverse events.
Collapse
Affiliation(s)
- Hang Wang
- Department of Pharmacy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qiyuan Yang
- Department of Pharmacy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yinjun Mao
- Department of Pharmacy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Haibo Ding
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Pinfang Huang
- Department of Pharmacy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhikun Zhan
- Department of Pharmacy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
Liu Y, Meng Y, Zhang J, Gu L, Shen S, Zhu Y, Wang J. Pharmacology Progresses and Applications of Chloroquine in Cancer Therapy. Int J Nanomedicine 2024; 19:6777-6809. [PMID: 38983131 PMCID: PMC11232884 DOI: 10.2147/ijn.s458910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/07/2024] [Indexed: 07/11/2024] Open
Abstract
Chloroquine is a common antimalarial drug and is listed in the World Health Organization Standard List of Essential Medicines because of its safety, low cost and ease of use. Besides its antimalarial property, chloroquine also was used in anti-inflammatory and antivirus, especially in antitumor therapy. A mount of data showed that chloroquine mainly relied on autophagy inhibition to exert its antitumor effects. However, recently, more and more researches have revealed that chloroquine acts through other mechanisms that are autophagy-independent. Nevertheless, the current reviews lacked a comprehensive summary of the antitumor mechanism and combined pharmacotherapy of chloroquine. So here we focused on the antitumor properties of chloroquine, summarized the pharmacological mechanisms of antitumor progression of chloroquine dependent or independent of autophagy inhibition. Moreover, we also discussed the side effects and possible application developments of chloroquine. This review provided a more systematic and cutting-edge knowledge involved in the anti-tumor mechanisms and combined pharmacotherapy of chloroquine in hope of carrying out more in-depth exploration of chloroquine and obtaining more clinical applications.
Collapse
Affiliation(s)
- Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Liwei Gu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Shengnan Shen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
- Department of Pharmacological Sciences, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| |
Collapse
|
3
|
Elshami FI, Shereef HA, El-Mehasseb IM, Shaban SY, van Eldik R. Hydroxychloroquine-Loaded Chitosan Nanoparticles Induce Anticancer Activity in A549 Lung Cancer Cells: Design, BSA Binding, Molecular Docking, Mechanistic, and Biological Evaluation. Int J Mol Sci 2023; 24:14103. [PMID: 37762406 PMCID: PMC10531786 DOI: 10.3390/ijms241814103] [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: 07/30/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
The current study describes the encapsulation of hydroxychloroquine, widely used in traditional medicine due to its diverse pharmacological and medicinal uses, in chitosan nanoparticles (CNPs). This work aims to combine the HCQ drug with CS NPs to generate a novel nanocomposite with improved characteristics and bioavailability. HCQ@CS NPs are roughly shaped like roadways and have a smooth surface with an average size of 159.3 ± 7.1 nm, a PDI of 0.224 ± 0.101, and a zeta potential of +46.6 ± 0.8 mV. To aid in the development of pharmaceutical systems for use in cancer therapy, the binding mechanism and affinity of the interaction between HCQ and HCQ@CS NPs and BSA were examined using stopped-flow and other spectroscopic approaches, supplemented by molecular docking analysis. HCQ and HCQ@CS NPs binding with BSA is driven by a ground-state complex formation that may be accompanied by a non-radiative energy transfer process, and binding constants indicate that HCQ@CS NPs-BSA was more stable than HCQ-BSA. The stopped-flow analysis demonstrated that, in addition to increasing BSA affinity, the nanoformulation HCQ@CS NPS changes the binding process and may open new routes for interaction. Docking experiments verified the development of the HCQ-BSA complex, with HCQ binding to site I on the BSA structure, primarily with the amino acids, Thr 578, Gln 579, Gln 525, Tyr 400, and Asn 404. Furthermore, the nanoformulation HCQ@CS NPS not only increased cytotoxicity against the A549 lung cancer cell line (IC50 = 28.57 ± 1.72 μg/mL) compared to HCQ (102.21 ± 0.67 μg/mL), but also exhibited higher antibacterial activity against both Gram-positive and Gram-negative bacteria when compared to HCQ and chloramphenicol, which is in agreement with the binding constants. The nanoformulation developed in this study may offer a viable therapy option for A549 lung cancer.
Collapse
Affiliation(s)
- Fawzia I. Elshami
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (F.I.E.); (I.M.E.-M.)
| | - Hadeer A. Shereef
- Clinical Pathology Department, University Hospital, Menoufia University, Shebin El-Kom 32512, Egypt;
| | - Ibrahim M. El-Mehasseb
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (F.I.E.); (I.M.E.-M.)
| | - Shaban Y. Shaban
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (F.I.E.); (I.M.E.-M.)
| | - Rudi van Eldik
- Department of Chemistry and Pharmacy, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
- Faculty of Chemistry, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| |
Collapse
|
4
|
Khan R, Panja S, Ding L, Tang S, Tang W, Kapoor E, Bennett RG, Oupický D. Polymeric Chloroquine as an Effective Antimigration Agent in the Treatment of Pancreatic Cancer. Mol Pharm 2022; 19:4631-4643. [PMID: 36346968 DOI: 10.1021/acs.molpharmaceut.2c00596] [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: 11/09/2022]
Abstract
Hydroxychloroquine (HCQ) has been the subject of multiple recent preclinical and clinical studies for its beneficial use in the combination treatments of different types of cancers. Polymeric HCQ (PCQ), a macromolecular multivalent version of HCQ, has been shown to be effective in various cancer models both in vitro and in vivo as an inhibitor of cancer cell migration and experimental lung metastasis. Here, we present detailed in vitro studies that show that low concentrations of PCQ can efficiently inhibit cancer cell migration and colony formation orders of magnitude more effectively compared to HCQ. After intraperitoneal administration of PCQ in vivo, high levels of tumor accumulation and penetration are observed, combined with strong antimetastatic activity in an orthotopic pancreatic cancer model. These studies support the idea that PCQ may be effectively used at low doses as an adjuvant in the therapy of pancreatic cancer. In conjunction with previously published literature, these studies further undergird the potential of PCQ as an anticancer agent.
Collapse
Affiliation(s)
- Rubayat Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Robert G Bennett
- Department of Internal Medicine and Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska 68105, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
5
|
Yamazaki K, Hoshi M, Tezuka H, Morita N, Hirayama M, Sato F, Yoshida S, Saito K. D‑allose enhances the efficacy of hydroxychloroquine against Lewis lung carcinoma cell growth by inducing autophagy. Oncol Rep 2022; 47:117. [PMID: 35543153 PMCID: PMC9115634 DOI: 10.3892/or.2022.8328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/05/2022] [Indexed: 11/05/2022] Open
Abstract
Various cancer cells require massive amounts of glucose as an energy source for their dysregulated growth. Although D‑allose, a rare sugar, inhibits tumor cell growth via inhibition of glucose uptake, a few cells can survive after treatment. However, the mechanism by which D‑allose‑resistant cells are generated remains unclear. Here, we investigated the properties of D‑allose‑resistant cells and evaluated the efficacy of combined treatment with this rare sugar and antitumor drugs. To this end, we established a D‑allose‑resistant tumor cell line and prepared a C57BL/6J mouse tumor xenograft model using Lewis lung carcinoma (LLC) cells. Xenograft‑bearing mice were treated with D‑allose (9 g/kg) and/or hydroxychloroquine (HCQ, 60 mg/kg), an autophagy inhibitor, for two weeks. Although D‑allose inhibited LLC cell growth in a dose‑dependent manner, a few cells survived. The upregulation of LC3‑II, a classical autophagy marker, and the downregulation of mTOR and its downstream molecule Beclin1 were observed in established D‑allose‑resistant LLC cells, which were more sensitive to cell death induced by HCQ. Similarly, in the tumor xenograft model, the tumor volume in mice co‑treated with D‑allose and HCQ was considerably smaller than that in untreated or HCQ‑treated mice. Importantly, the administration of D‑allose induced autophagy selectively at the tumor site of the xenograft‑bearing mice. These results provide a new therapeutic strategy targeting autophagy which is induced in tumor cells by D‑allose administration, and may be used to improve therapies for lung cancer.
Collapse
Affiliation(s)
- Kyoka Yamazaki
- Department of Disease Control and Prevention, Fujita Health University, Aichi 470‑1192, Japan
| | - Masato Hoshi
- Department of Informative Clinical Medicine, Fujita Health University, Aichi 470‑1192, Japan
| | - Hiroyuki Tezuka
- Department of Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Aichi 470‑1192, Japan
| | - Nanaka Morita
- Department of Disease Control and Prevention, Fujita Health University, Aichi 470‑1192, Japan
| | - Masaya Hirayama
- Department of Morphology and Diagnostic Pathology, Fujita Health University, Aichi 470‑1192, Japan
| | - Fumiaki Sato
- Department of Informative Clinical Medicine, Fujita Health University, Aichi 470‑1192, Japan
| | - Sayaka Yoshida
- Department of Informative Clinical Medicine, Fujita Health University, Aichi 470‑1192, Japan
| | - Kuniaki Saito
- Department of Disease Control and Prevention, Fujita Health University, Aichi 470‑1192, Japan
| |
Collapse
|
6
|
Hsieh CH, Huang YW, Tsai TF. Oral Conventional Synthetic Disease-Modifying Antirheumatic Drugs with Antineoplastic Potential: a Review. Dermatol Ther (Heidelb) 2022; 12:835-860. [PMID: 35381976 PMCID: PMC9021342 DOI: 10.1007/s13555-022-00713-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Indexed: 01/17/2023] Open
Abstract
There is an increasing trend of malignancy worldwide. Disease-modifying antirheumatic drugs (DMARDs) are the cornerstones for the treatment of immune-mediated inflammatory diseases (IMIDs), but risk of malignancy is a major concern for patients receiving DMARDs. In addition, many IMIDs already carry higher background risks of neoplasms. Recently, the black box warning of malignancies has been added for Janus kinase inhibitors. Also, the use of biologic DMARDs in patients with established malignancies is usually discouraged owing to exclusion of such patients in pivotal studies and, hence, lack of evidence. In contrast, some conventional synthetic DMARDs (csDMARDs) have been reported to show antineoplastic properties and can be beneficial for patients with cancer. Among the csDMARDs, chloroquine and hydroxychloroquine have been the most extensively studied, and methotrexate is an established chemotherapeutic agent. Even cyclosporine A, a well-known drug associated with cancer risk, can potentiate the effect of some chemotherapeutic agents. We review the possible mechanisms behind and clinical evidence of the antineoplastic activities of csDMARDs, including chloroquine and hydroxychloroquine, cyclosporine, leflunomide, mycophenolate mofetil, mycophenolic acid, methotrexate, sulfasalazine, and thiopurines. This knowledge may guide physicians in the choice of csDMARDs for patients with concurrent IMIDs and malignancies.
Collapse
Affiliation(s)
- Cho-Hsun Hsieh
- Department of Medical Education, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Wei Huang
- Department of Dermatology, National Taiwan University Hospital, 7 Chung Shan S Rd, Taipei, 10048, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital, 7 Chung Shan S Rd, Taipei, 10048, Taiwan. .,Department of Dermatology, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
7
|
Understanding the Role of Autophagy in Cancer Formation and Progression Is a Real Opportunity to Treat and Cure Human Cancers. Cancers (Basel) 2021; 13:cancers13225622. [PMID: 34830777 PMCID: PMC8616104 DOI: 10.3390/cancers13225622] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary The modulation of autophagy represents a potential therapeutic strategy for cancer. More than one hundred clinical trials have been conducted or are ongoing to explore the efficacy of autophagy modulators to reduce the tumor growth and potentiate the anti-cancer effects of conventional therapy. Despite this, the effective role of autophagy during tumor initiation, growth, and metastasis remains not well understood. Depending on the cancer type and stage of cancer, autophagy may have tumor suppressor properties as well as help cancer cells to proliferate and evade cancer therapy. The current review aims to summarize the current knowledge about the autophagy implications in cancer and report the therapeutic opportunities based on the modulation of the autophagy process. Abstract The malignant transformation of a cell produces the accumulation of several cellular adaptions. These changes determine variations in biological processes that are necessary for a cancerous cell to survive during stressful conditions. Autophagy is the main nutrient recycling and metabolic adaptor mechanism in eukaryotic cells, represents a continuous source of energy and biomolecules, and is fundamental to preserve the correct cellular homeostasis during unfavorable conditions. In recent decades, several findings demonstrate a close relationship between autophagy, malignant transformation, and cancer progression. The evidence suggests that autophagy in the cancer context has a bipolar role (it may act as a tumor suppressor and as a mechanism of cell survival for established tumors) and demonstrates that the targeting of autophagy may represent novel therapeutic opportunities. Accordingly, the modulation of autophagy has important clinical benefits in patients affected by diverse cancer types. Currently, about 30 clinical trials are actively investigating the efficacy of autophagy modulators to enhance the efficacy of cytotoxic chemotherapy treatments. A deeper understanding of the molecular pathways regulating autophagy in the cancer context will provide new ways to target autophagy for improving the therapeutic benefits. Herein, we describe how autophagy participates during malignant transformation and cancer progression, and we report the ultimate efforts to translate this knowledge into specific therapeutic approaches to treat and cure human cancers.
Collapse
|
8
|
Zada S, Hwang JS, Ahmed M, Lai TH, Pham TM, Elashkar O, Kim DR. Cross talk between autophagy and oncogenic signaling pathways and implications for cancer therapy. Biochim Biophys Acta Rev Cancer 2021; 1876:188565. [PMID: 33992723 DOI: 10.1016/j.bbcan.2021.188565] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
Autophagy is a highly conserved metabolic process involved in the degradation of intracellular components including proteins and organelles. Consequently, it plays a critical role in recycling metabolic energy for the maintenance of cellular homeostasis in response to various stressors. In cancer, autophagy either suppresses or promotes cancer progression depending on the stage and cancer type. Epithelial-mesenchymal transition (EMT) and cancer metastasis are directly mediated by oncogenic signal proteins including SNAI1, SLUG, ZEB1/2, and NOTCH1, which are functionally correlated with autophagy. In this report, we discuss the crosstalk between oncogenic signaling pathways and autophagy followed by possible strategies for cancer treatment via regulation of autophagy. Although autophagy affects EMT and cancer metastasis, the overall signaling pathways connecting cancer progression and autophagy are still illusive. In general, autophagy plays a critical role in cancer cell survival by providing a minimum level of energy via self-digestion. Thus, cancer cells face nutrient limitations and challenges under stress during EMT and metastasis. Conversely, autophagy acts as a potential cancer suppressor by degrading oncogenic proteins, which are essential for cancer progression, and by removing damaged components such as mitochondria to enhance genomic stability. Therefore, autophagy activators or inhibitors represent possible cancer therapeutics. We further discuss the regulation of autophagy-dependent degradation of oncogenic proteins and its functional correlation with oncogenic signaling pathways, with potential applications in cancer therapy.
Collapse
Affiliation(s)
- Sahib Zada
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Trang Minh Pham
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Omar Elashkar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea.
| |
Collapse
|
9
|
Farhan M, Silva M, Li S, Yan F, Fang J, Peng T, Hu J, Tsao M, Little P, Zheng W. The role of FOXOs and autophagy in cancer and metastasis-Implications in therapeutic development. Med Res Rev 2020; 40:2089-2113. [PMID: 32474970 PMCID: PMC7586888 DOI: 10.1002/med.21695] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 04/21/2020] [Accepted: 05/16/2020] [Indexed: 12/17/2022]
Abstract
Autophagy is a highly conserved intracellular degradation process that plays a crucial role in cell survival and stress reactions as well as in cancer development and metastasis. Autophagy process involves several steps including sequestration, fusion of autophagosomes with lysosomes and degradation. Forkhead box O (FOXO) transcription factors regulate the expression of genes involved in cellular metabolic activity and signaling pathways of cancer growth and metastasis. Recent evidence suggests that FOXO proteins are also involved in autophagy regulation. The relationship among FOXOs, autophagy, and cancer has been drawing attention of many who work in the field. This study summarizes the role of FOXO proteins and autophagy in cancer growth and metastasis and analyzes their potential roles in cancer disease management.
Collapse
Affiliation(s)
- Mohd Farhan
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Marta Silva
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Shuai Li
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Fengxia Yan
- Department of MedicineJinan UniversityGuangzhouChina
| | - Jiankang Fang
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Tangming Peng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Jim Hu
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Ming‐Sound Tsao
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Peter Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Wenhua Zheng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| |
Collapse
|
10
|
Martinez GP, Zabaleta ME, Di Giulio C, Charris JE, Mijares MR. The Role of Chloroquine and Hydroxychloroquine in Immune Regulation and Diseases. Curr Pharm Des 2020; 26:4467-4485. [DOI: 10.2174/1381612826666200707132920] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are derivatives of the heterocyclic aromatic compound
quinoline. These economical compounds have been used as antimalarial agents for many years. Currently,
they are used as monotherapy or in conjunction with other therapies for the treatment of autoimmune diseases
such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS) and antiphospholipid
antibody syndrome (APS). Based on its effects on the modulation of the autophagy process, various
clinical studies suggest that CQ and HCQ could be used in combination with other chemotherapeutics for the
treatment of various types of cancer. Furthermore, the antiviral effects showed against Zika, Chikungunya, and
HIV are due to the annulation of endosomal/lysosomal acidification. Recently, CQ and HCQ were approved for
the U.S. Food and Drug Administration (FDA) for the treatment of infected patients with the coronavirus SARSCoV-
2, causing the disease originated in December 2019, namely COVID-2019. Several mechanisms have been
proposed to explain the pharmacological effects of these drugs: 1) disruption of lysosomal and endosomal pH, 2)
inhibition of protein secretion/expression, 3) inhibition of antigen presentation, 4) decrease of proinflammatory
cytokines, 5) inhibition of autophagy, 6) induction of apoptosis and 7) inhibition of ion channels activation. Thus,
evidence has shown that these structures are leading molecules that can be modified or combined with other
therapeutic agents. In this review, we will discuss the most recent findings in the mechanisms of action of CQ and
HCQ in the immune system, and the use of these antimalarial drugs on diseases.
Collapse
Affiliation(s)
- Gricelis P. Martinez
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Mercedes E. Zabaleta
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Camilo Di Giulio
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Jaime E. Charris
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, 47206, Los Chaguaramos 1041-A, Caracas, Venezuela
| | - Michael R. Mijares
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| |
Collapse
|
11
|
Horne GA, Stobo J, Kelly C, Mukhopadhyay A, Latif AL, Dixon-Hughes J, McMahon L, Cony-Makhoul P, Byrne J, Smith G, Koschmieder S, BrÜmmendorf TH, Schafhausen P, Gallipoli P, Thomson F, Cong W, Clark RE, Milojkovic D, Helgason GV, Foroni L, Nicolini FE, Holyoake TL, Copland M. A randomised phase II trial of hydroxychloroquine and imatinib versus imatinib alone for patients with chronic myeloid leukaemia in major cytogenetic response with residual disease. Leukemia 2020; 34:1775-1786. [PMID: 31925317 PMCID: PMC7224085 DOI: 10.1038/s41375-019-0700-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/19/2022]
Abstract
In chronic-phase chronic myeloid leukaemia (CP-CML), residual BCR-ABL1+ leukaemia stem cells are responsible for disease persistence despite TKI. Based on in vitro data, CHOICES (CHlorOquine and Imatinib Combination to Eliminate Stem cells) was an international, randomised phase II trial designed to study the safety and efficacy of imatinib (IM) and hydroxychloroquine (HCQ) compared with IM alone in CP-CML patients in major cytogenetic remission with residual disease detectable by qPCR. Sixty-two patients were randomly assigned to either arm. Treatment 'successes' was the primary end point, defined as ≥0.5 log reduction in 12-month qPCR level from trial entry. Selected secondary study end points were 24-month treatment 'successes', molecular response and progression at 12 and 24 months, comparison of IM levels, and achievement of blood HCQ levels >2000 ng/ml. At 12 months, there was no difference in 'success' rate (p = 0.58); MMR was achieved in 80% (IM) vs 92% (IM/HCQ) (p = 0.21). At 24 months, the 'success' rate was 20.8% higher with IM/HCQ (p = 0.059). No patients progressed. Seventeen serious adverse events, including four serious adverse reactions, were reported; diarrhoea occurred more frequently with combination. IM/HCQ is tolerable in CP-CML, with modest improvement in qPCR levels at 12 and 24 months, suggesting autophagy inhibition maybe of clinical value in CP-CML.
Collapse
MESH Headings
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cytogenetic Analysis/methods
- Female
- Follow-Up Studies
- Fusion Proteins, bcr-abl/genetics
- Humans
- Hydroxychloroquine/administration & dosage
- Imatinib Mesylate/administration & dosage
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Middle Aged
- Prognosis
- Retrospective Studies
- Survival Rate
Collapse
Affiliation(s)
- G A Horne
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - J Stobo
- Cancer Research UK Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | - C Kelly
- Cancer Research UK Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | - A Mukhopadhyay
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - A L Latif
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - J Dixon-Hughes
- Cancer Research UK Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | - L McMahon
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - P Cony-Makhoul
- Haematology department, CH Annecy-Genevois, Pringy, France
| | - J Byrne
- Department of Haematology, Nottingham City Hospital, Nottingham, UK
| | - G Smith
- Department of Haematology, St James's University Hospital, Leeds, UK
| | - S Koschmieder
- Department of Medicine (Hematology Oncology, Hemostaseology, and Stem Cell Transplantation), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - T H BrÜmmendorf
- Department of Medicine (Hematology Oncology, Hemostaseology, and Stem Cell Transplantation), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - P Schafhausen
- Department of Internal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - P Gallipoli
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - F Thomson
- Experimental therapeutics, Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - W Cong
- Experimental therapeutics, Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - R E Clark
- Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - D Milojkovic
- Department of Haematology, Hammersmith Hospital, London, UK
| | - G V Helgason
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - L Foroni
- Department of Haematology, Imperial College London, London, UK
| | - F E Nicolini
- Hématologie Clinique and INSERM U1052, CRCL, Centre Léon Bérard, Lyon, France
| | - T L Holyoake
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - M Copland
- Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
12
|
Kaleağasıoğlu F, Ali DM, Berger MR. Multiple Facets of Autophagy and the Emerging Role of Alkylphosphocholines as Autophagy Modulators. Front Pharmacol 2020; 11:547. [PMID: 32410999 PMCID: PMC7201076 DOI: 10.3389/fphar.2020.00547] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a highly conserved multistep process and functions as passage for degrading and recycling protein aggregates and defective organelles in eukaryotic cells. Based on the nature of these materials, their size and degradation rate, four types of autophagy have been described, i.e. chaperone mediated autophagy, microautophagy, macroautophagy, and selective autophagy. One of the major regulators of this process is mTOR, which inhibits the downstream pathway of autophagy following the activation of its complex 1 (mTORC1). Alkylphosphocholine (APC) derivatives represent a novel class of antineoplastic agents that inhibit the serine-threonine kinase Akt (i.e. protein kinase B), which mediates cell survival and cause cell cycle arrest. They induce autophagy through inhibition of the Akt/mTOR cascade. They interfere with phospholipid turnover and thus modify signaling chains, which start from the cell membrane and modulate PI3K/Akt/mTOR, Ras-Raf-MAPK/ERK and SAPK/JNK pathways. APCs include miltefosine, perifosine, and erufosine, which represent the first-, second- and third generation of this class, respectively. In a high fraction of human cancers, constitutively active oncoprotein Akt1 suppresses autophagy in vitro and in vivo. mTOR is a down-stream target for Akt, the activation of which suppresses autophagy. However, treatment with APC derivatives will lead to dephosphorylation (hence deactivation) of mTOR and thus induces autophagy. Autophagy is a double-edged sword and may result in chemotherapeutic resistance as well as cancer cell death when apoptotic pathways are inactive. APCs display differential autophagy induction capabilities in different cancer cell types. Therefore, autophagy-dependent cellular responses need to be well understood in order to improve the chemotherapeutic outcome.
Collapse
Affiliation(s)
- Ferda Kaleağasıoğlu
- Department of Pharmacology, Faculty of Medicine, Near East University, Mersin, Turkey
| | - Doaa M. Ali
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pharmacology and Experimental Therapeutics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Martin R. Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
13
|
Brazil L, Swampillai AL, Mak KM, Edwards D, Mesiri P, Clifton-Hadley L, Shaffer R, Lewis J, Watts C, Jeffries S, Gkogkou P, Chalmers AJ, Fersht NL, Hackshaw A, Short SC. Hydroxychloroquine and short-course radiotherapy in elderly patients with newly diagnosed high-grade glioma: a randomized phase II trial. Neurooncol Adv 2020; 2:vdaa046. [PMID: 32642699 PMCID: PMC7236384 DOI: 10.1093/noajnl/vdaa046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Effective treatment for patients at least 70 years with newly diagnosed glioblastoma remains challenging and alternatives to conventional cytotoxics are appealing. Autophagy inhibition has shown promising efficacy and safety in small studies of glioblastoma and other cancers. Methods We conducted a randomized phase II trial to compare radiotherapy with or without hydroxychloroquine (2:1 allocation). Patients aged at least 70 years with newly diagnosed high-grade glioma deemed suitable for short-course radiotherapy with an ECOG performance status of 0-1 were included. Radiotherapy treatment consisted of 30 Gy, delivered as 6 fractions given over 2 weeks (5 Gy per fraction). Hydroxychloroquine was given as 200 mg orally b.d. from 7 days prior to radiotherapy until disease progression. The primary endpoint was 1-year overall survival (OS). Secondary endpoints included progression-free survival (PFS), quality of life, and toxicity. Results Fifty-four patients with a median age of 75 were randomized between May 2013 and October 2016. The trial was stopped early in 2016. One-year OS was 20.3% (95% confidence interval [CI] 8.2-36.0) hydroxychloroquine group, and 41.2% (95% CI 18.6-62.6) radiotherapy alone, with a median survival of 7.9 and 11.5 months, respectively. The corresponding 6-month PFS was 35.3% (95% CI 19.3-51.7) and 29.4% (95% CI 10.7-51.1). The outcome in the control arm was better than expected and the excess of deaths in the hydroxychloroquine group appeared unrelated to cancer. There were more grade 3-5 events in the hydroxychloroquine group (60.0%) versus radiotherapy alone (38.9%) without any clear common causation. Conclusions Hydroxychloroquine with short-course radiotherapy did not improve survival compared to radiotherapy alone in elderly patients with glioblastoma.
Collapse
Affiliation(s)
- Lucy Brazil
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Ka Man Mak
- Cancer Research UK and UCL Cancer Trials Centre, London, UK
| | - Darren Edwards
- Cancer Research UK and UCL Cancer Trials Centre, London, UK
| | | | | | | | - Joanne Lewis
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle Upon Tyne Hospitals Trust, Newcastle, UK
| | - Colin Watts
- University of Birmingham/Queen Elizabeth Hospital, Birmingham, UK
| | - Sarah Jeffries
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | | | - Allan Hackshaw
- Cancer Research UK and UCL Cancer Trials Centre, London, UK
| | | |
Collapse
|
14
|
Mulcahy Levy JM, Thorburn A. Autophagy in cancer: moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ 2020; 27:843-857. [PMID: 31836831 PMCID: PMC7206017 DOI: 10.1038/s41418-019-0474-7] [Citation(s) in RCA: 308] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/12/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy allows for cellular material to be delivered to lysosomes for degradation resulting in basal or stress-induced turnover of cell components that provide energy and macromolecular precursors. These activities are thought to be particularly important in cancer where both tumor-promoting and tumor-inhibiting functions of autophagy have been described. Autophagy has also been intricately linked to apoptosis and programmed cell death, and understanding these interactions is becoming increasingly important in improving cancer therapy and patient outcomes. In this review, we consider how recent discoveries about how autophagy manipulation elicits its effects on cancer cell behavior can be leveraged to improve therapeutic responses.
Collapse
Affiliation(s)
- Jean M Mulcahy Levy
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
15
|
Pérez-Hernández M, Arias A, Martínez-García D, Pérez-Tomás R, Quesada R, Soto-Cerrato V. Targeting Autophagy for Cancer Treatment and Tumor Chemosensitization. Cancers (Basel) 2019; 11:E1599. [PMID: 31635099 PMCID: PMC6826429 DOI: 10.3390/cancers11101599] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy is a tightly regulated catabolic process that facilitates nutrient recycling from damaged organelles and other cellular components through lysosomal degradation. Deregulation of this process has been associated with the development of several pathophysiological processes, such as cancer and neurodegenerative diseases. In cancer, autophagy has opposing roles, being either cytoprotective or cytotoxic. Thus, deciphering the role of autophagy in each tumor context is crucial. Moreover, autophagy has been shown to contribute to chemoresistance in some patients. In this regard, autophagy modulation has recently emerged as a promising therapeutic strategy for the treatment and chemosensitization of tumors, and has already demonstrated positive clinical results in patients. In this review, the dual role of autophagy during carcinogenesis is discussed and current therapeutic strategies aimed at targeting autophagy for the treatment of cancer, both under preclinical and clinical development, are presented. The use of autophagy modulators in combination therapies, in order to overcome drug resistance during cancer treatment, is also discussed as well as the potential challenges and limitations for the use of these novel therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Marta Pérez-Hernández
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| | - Alain Arias
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Department of Integral Adult Dentistry, Research Centre for Dental Sciences (CICO), Universidad de La Frontera, Temuco 4811230, Chile.
- Research Group of Health Sciences, Faculty of Health Sciences, Universidad Adventista de Chile, Chillán 3780000, Chile.
| | - David Martínez-García
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| | - Roberto Quesada
- Department of Chemistry, Universidad de Burgos, 09001 Burgos, Spain.
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08905 Barcelona, Spain.
- Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain.
| |
Collapse
|
16
|
Kranzbühler B, Salemi S, Mortezavi A, Sulser T, Eberli D. Combined N-terminal androgen receptor and autophagy inhibition increases the antitumor effect in enzalutamide sensitive and enzalutamide resistant prostate cancer cells. Prostate 2019; 79:206-214. [PMID: 30345525 DOI: 10.1002/pros.23725] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/25/2018] [Indexed: 11/12/2022]
Abstract
INTRODUCTION AND OBJECTIVES Multiple androgen receptor (AR)-dependent and -independent resistance mechanisms limit the efficacy of current castration-resistant prostate cancer (CRPC) treatment. Novel N-terminal domain (NTD) binding AR-targeting compounds, including EPI-001 (EPI), have the promising ability to block constitutively active splice variants, which represent a major resistance mechanism in CRPC. Autophagy is a conserved lysosomal degradation pathway that acts as survival mechanism in cells exposed to anticancer treatments. We hypothesized, that promising NTD-AR treatment may upregulate autophagy and that a combination of NTD-AR and autophagy inhibition might therefore increase antitumor effects. METHODS AR-expressing prostate cancer cell lines (LNCaP, LNCaP-EnzR) were treated with different concentrations of EPI (10, 25, 50 μM) and in combination with the autophagy inhibitors chloroquine (CHQ, 20 μM) or 3-methyladenine (3-MA, 5 mM). Cell proliferation was assessed by WST-1-assays after 1 and 7 days. Ethidium bromide and Annexin V were used to measure viability and apoptosis on day 7 after treatment. Autophagosome formation was detected by AUTOdot staining. In addition, autophagic activity was monitored by immunocytochemistry and Western blot (WES) for the expression of ATG5, Beclin1, LC3-I/II and p62. RESULTS Treatment with EPI resulted in a dose-dependent reduction of cell growth and increased apoptosis in both cancer cell lines on day 7. In addition, EPI treatment demonstrated an upregulated autophagosome formation in LNCaP and LNCaP-EnzR cells. Assessment of autophagic activity by immunocytochemistry and WES revealed an increase of ATG5 and LC3-II expression and a decreased p62 expression in all EPI-treated cells. A combined treatment of EPI with autophagy inhibitors led to a further significant reduction of cell viability in both cell lines. CONCLUSIONS Our results demonstrate that NTD targeting AR inhibition using EPI leads to an increased autophagic activity in LNCaP and LNCaP-EnzR prostate cancer cells. A combination of NTD AR blockage with simultaneous autophagy inhibition increases the antitumor effect of EPI in prostate cancer cells. Double treatment may offer a promising strategy to overcome resistance mechanisms in advanced prostate cancer.
Collapse
Affiliation(s)
- Benedikt Kranzbühler
- Department of Urology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Souzan Salemi
- Department of Urology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Ashkan Mortezavi
- Department of Urology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| |
Collapse
|
17
|
Yu X, Lin H, Wang Y, Lv W, Zhang S, Qian Y, Deng X, Feng N, Yu H, Qian B. d-limonene exhibits antitumor activity by inducing autophagy and apoptosis in lung cancer. Onco Targets Ther 2018; 11:1833-1847. [PMID: 29670359 PMCID: PMC5894671 DOI: 10.2147/ott.s155716] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Purpose d-limonene is a plant extract with widespread application, and it has been recently reported to have antiproliferative and proapoptotic effects on cancer cells. However, the mechanisms by which d-limonene achieves these effects, especially in lung cancer, are not entirely clear. Therefore, the goal of this study was to examine the effects of d-limonene on lung cancer and explore its mechanisms of action. Methods We examined the therapeutic effects of d-limonene on lung cancer cells and in a xenograft animal model by characterizing its effects on the pathways of apoptosis and autophagy. Cell proliferation was measured using the Cell Counting Kit-8, and apoptosis was determined by flow cytometric analysis. Levels of LC3 puncta, an autophagy marker, were analyzed by laser scanning confocal microscopy. Autophagy and apoptosis-related gene expression were assessed by real-time quantitative polymerase chain reaction and Western blot. Results d-limonene inhibited the growth of lung cancer cells and suppressed the growth of transplanted tumors in nude mice. Expression of apoptosis and autophagy-related genes were increased in tumors after treatment with d-limonene. Furthermore, the use of chloroquine, an autophagy inhibitor, and knockdown of the atg5 gene, suppressed the apoptosis induced by d-limonene. Conclusion d-limonene may have a therapeutic effect on lung cancer as it can induce apoptosis of lung cancer cells by promoting autophagy.
Collapse
Affiliation(s)
- Xiao Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Lin
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenwen Lv
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Qian
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobei Deng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nannan Feng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Biyun Qian
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital & Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Abstract
Mechanistic target of rapamycin (mTOR) is the kinase subunit of two structurally and functionally distinct large multiprotein complexes, referred to as mTOR complex 1 (mTORC1) and mTORC2. mTORC1 and mTORC2 play key physiological roles as they control anabolic and catabolic processes in response to external cues in a variety of tissues and organs. However, mTORC1 and mTORC2 activities are deregulated in widespread human diseases, including cancer. Cancer cells take advantage of mTOR oncogenic signaling to drive their proliferation, survival, metabolic transformation, and metastatic potential. Therefore, mTOR lends itself very well as a therapeutic target for innovative cancer treatment. mTOR was initially identified as the target of the antibiotic rapamycin that displayed remarkable antitumor activity in vitro Promising preclinical studies using rapamycin and its derivatives (rapalogs) demonstrated efficacy in many human cancer types, hence supporting the launch of numerous clinical trials aimed to evaluate the real effectiveness of mTOR-targeted therapies. However, rapamycin and rapalogs have shown very limited activity in most clinical contexts, also when combined with other drugs. Thus, novel classes of mTOR inhibitors with a stronger antineoplastic potency have been developed. Nevertheless, emerging clinical data suggest that also these novel mTOR-targeting drugs may have a weak antitumor activity. Here, we summarize the current status of available mTOR inhibitors and highlight the most relevant results from both preclinical and clinical studies that have provided valuable insights into both their efficacy and failure.
Collapse
|
19
|
Eren M, Kucukevcilioglu M, Durukan AH. Macular sensitivities measured by microperimetry in patients on hydroxychloroquine treatment. Cutan Ocul Toxicol 2018; 37:275-280. [PMID: 29495877 DOI: 10.1080/15569527.2018.1446976] [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: 10/17/2022]
Abstract
PURPOSE To examine retinal sensitivity in patients on hydroxychloroquine (HCQ) with no evidence of retinopathy. MATERIALS AND METHODS Seventy patients on HCQ and 30 healthy control subjects were included prospectively. All subjects underwent complete ophthalmic examination including best corrected visual acuity, tonometry, colour vision testing, biomicroscopy of anterior segment, dilated fundoscopy, 10-2 visual field testing, and spectral domain optical coherence tomography. The patients and control subjects who met the inclusion criteria and had normal tests underwent microperimetry (MP) testing. First, all patients were compared with the control group. Secondly, patients were divided into three sets of groups based on (1) duration of use ≤5 years (DOU≤5) and >5 years (DOU>5), (2) daily dose ≤5 mg/kg/day (DD≤ 5) and >5 mg/kg/day (DD>5), and (3) a cumulative dose ≤1000 gr (CD≤ 1000) and >1000 gr (CD>1000), and these groups were compared to each other and to the control group. A correlation analysis was also performed between MP sensitivity and DOU, DD, and CD. RESULTS Seven patients on HCQ showing visual field abnormality were excluded which yielded 63 patients and 30 control subjects for the final analysis. We observed significant differences only in the central region but not in the paracentral or peripheral regions on MP in HCQ users. The median MP sensitivities in the central region were significantly lower in all the patients [84 (63-100) dB], and in subgroups of DOU >5 [84 (63-99) dB], DD >5 [82 (63-97) dB] and CD >1000 [82 (63-92) dB] when compared to controls [89.7 (83-98) dB]. A statistically significant correlation was found only between DD and MP sensitivity in the central region (r = -0.263; p = 0.02). CONCLUSIONS MP sensitivities in the central macula were significantly lower in patients taking HCQ for more than 5 years, at a daily dose more than 5 mg per day, and with a cumulative dose over 1000 gr. Further research investigating long-term follow-up changes in MP sensitivities is needed to determine cutoff values for early retinal toxicity.
Collapse
Affiliation(s)
- Mustafa Eren
- a Department of Ophthalmology , Marmaris Public Hospital , Mugla , Turkey
| | - Murat Kucukevcilioglu
- b Department of Ophthalmology , Gulhane Training and Research Hospital , Ankara , Turkey
| | - Ali Hakan Durukan
- b Department of Ophthalmology , Gulhane Training and Research Hospital , Ankara , Turkey
| |
Collapse
|
20
|
Chi KH, Wang YS, Huang YC, Chiang HC, Chi MS, Chi CH, Wang HE, Kao SJ. Simultaneous activation and inhibition of autophagy sensitizes cancer cells to chemotherapy. Oncotarget 2018; 7:58075-58088. [PMID: 27486756 PMCID: PMC5295413 DOI: 10.18632/oncotarget.10873] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 07/09/2016] [Indexed: 12/19/2022] Open
Abstract
While combined chemotherapy (CT) with an autophagy inducer and an autophagy inhibitor appears paradoxical, it may provide a more effective perturbation of autophagy pathways. We used two dissimilar cell lines to test the hypothesis that autophagy is the common denominator of cell fate after CT. HA22T cells are characterized by CT-induced apoptosis and use autophagy to prevent cell death, while Huh7.5.1 cells exhibit sustained autophagic morphology after CT. Combined CT and rapamycin treatment resulted in a better combination index (CI) in Huh7.5.1 cells than combined CT and chloroquine, while the reverse was true in HA22T cells. The combination of 3 drugs (triplet drug treatment) had the best CI. After triplet drug treatment, HA22T cells switched from protective autophagy to mitochondrial membrane permeabilization and endoplasmic reticulum stress response-induced apoptosis, while Huh7.5.1 cells intensified autophagic lethality. Most importantly, both cell lines showed activation of Akt after CT, while the triplet combination blocked Akt activation through inhibition of phospholipid lipase D activity. This novel finding warrants further investigation as a broad chemosensitization strategy.
Collapse
Affiliation(s)
- Kwan-Hwa Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Institute of Veterinary Clinical Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Shan Wang
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Department of Research and Development, JohnPro Biotech Inc., Taipei, Taiwan
| | - Yi-Chun Huang
- Department of Research and Development, JohnPro Biotech Inc., Taipei, Taiwan
| | - Hsin-Chien Chiang
- Department of Research and Development, JohnPro Biotech Inc., Taipei, Taiwan
| | - Mau-Shin Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chau-Hwa Chi
- Institute of Veterinary Clinical Science, National Taiwan University, Taipei, Taiwan
| | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Shang-Jyh Kao
- Division of Pulmonary Medicine, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
21
|
Autophagy mediates glucose starvation-induced glioblastoma cell quiescence and chemoresistance through coordinating cell metabolism, cell cycle, and survival. Cell Death Dis 2018; 9:213. [PMID: 29434213 PMCID: PMC5833690 DOI: 10.1038/s41419-017-0242-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/16/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023]
Abstract
Metabolic reprogramming is pivotal to sustain cancer growth and progression. As such dietary restriction therapy represents a promising approach to starve and treat cancers. Nonetheless, tumors are dynamic and heterogeneous populations of cells with metabolic activities modulated by spatial and temporal contexts. Autophagy is a major pathway controlling cell metabolism. It can downregulate cell metabolism, leading to cancer cell quiescence, survival, and chemoresistance. To understand treatment dynamics and provide rationales for better future therapeutic strategies, we investigated whether and how autophagy is involved in the chemo-cytotoxicity and -resistance using two commonly used human glioblastoma (GBM) cell lines U87 and U251 together with primary cancer cells from the GBM patients. Our results suggest that autophagy mediates chemoresistance through reprogramming cancer cell metabolism and promoting quiescence and survival. Further unbiased transcriptome profiling identified a number of clinically relevant pathways and genes, strongly correlated with TCGA data. Our analyses have not only reported many well-known tumor players, but also uncovered a number of genes that were not previously implicated in cancers and/or GBM. The known functions of these genes are highly suggestive. It would be of high interest to investigate their potential involvement in GBM tumorigenesis, progression, and/or drug resistance. Taken together, our results suggest that autophagy inhibition could be a viable approach to aid GBM chemotherapy and combat drug resistance.
Collapse
|
22
|
Hsu SPC, Kuo JS, Chiang HC, Wang HE, Wang YS, Huang CC, Huang YC, Chi MS, Mehta MP, Chi KH. Temozolomide, sirolimus and chloroquine is a new therapeutic combination that synergizes to disrupt lysosomal function and cholesterol homeostasis in GBM cells. Oncotarget 2018; 9:6883-6896. [PMID: 29467937 PMCID: PMC5805523 DOI: 10.18632/oncotarget.23855] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 12/22/2017] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma (GBM) cells are characterized by high phagocytosis, lipogenesis, exocytosis activities, low autophagy capacity and high lysosomal demand are necessary for survival and invasion. The lysosome stands at the cross roads of lipid biosynthesis, transporting, sorting between exogenous and endogenous cholesterol. We hypothesized that three already approved drugs, the autophagy inducer, sirolimus (rapamycin, Rapa), the autophagy inhibitor, chloroquine (CQ), and DNA alkylating chemotherapy, temozolomide (TMZ) could synergize against GBM. This repurposed triple therapy combination induced GBM apoptosis in vitro and inhibited GBM xenograft growth in vivo. Cytotoxicity is caused by induction of lysosomal membrane permeabilization and release of hydrolases, and may be rescued by cholesterol supplementation. Triple treatment inhibits lysosomal function, prevents cholesterol extraction from low density lipoprotein (LDL), and causes clumping of lysosome associated membrane protein-1 (LAMP-1) and lipid droplets (LD) accumulation. Co-treatment of the cell lines with inhibitor of caspases and cathepsin B only partially reverse of cytotoxicities, while N-acetyl cysteine (NAC) can be more effective. A combination of reactive oxygen species (ROS) generation from cholesterol depletion are the early event of underling mechanism. Cholesterol repletion abolished the ROS production and reversed the cytotoxicity from QRT treatment. The shortage of free cholesterol destabilizes lysosomal membranes converting aborted autophagy to apoptosis through either direct mitochondria damage or cathepsin B release. This promising anti-GBM triple therapy combination severely decreases mitochondrial function, induces lysosome-dependent apoptotic cell death, and is now poised for further clinical testing and validation.
Collapse
Affiliation(s)
- Sanford P C Hsu
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming University, Taipei, Taiwan
| | - John S Kuo
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | | | - Hsin-Ell Wang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | - Mau-Shin Chi
- Department of Radiation Therapy and Oncology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | - Kwan-Hwa Chi
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Miami Cancer Institute, Miami, FL, USA
| |
Collapse
|
23
|
Verbaanderd C, Maes H, Schaaf MB, Sukhatme VP, Pantziarka P, Sukhatme V, Agostinis P, Bouche G. Repurposing Drugs in Oncology (ReDO)-chloroquine and hydroxychloroquine as anti-cancer agents. Ecancermedicalscience 2017; 11:781. [PMID: 29225688 PMCID: PMC5718030 DOI: 10.3332/ecancer.2017.781] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Indexed: 12/26/2022] Open
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are well-known 4-aminoquinoline antimalarial agents. Scientific evidence also supports the use of CQ and HCQ in the treatment of cancer. Overall, preclinical studies support CQ and HCQ use in anti-cancer therapy, especially in combination with conventional anti-cancer treatments since they are able to sensitise tumour cells to a variety of drugs, potentiating the therapeutic activity. Thus far, clinical results are mostly in favour of the repurposing of CQ. However, over 30 clinical studies are still evaluating the activity of both CQ and HCQ in different cancer types and in combination with various standard treatments. Interestingly, CQ and HCQ exert effects both on cancer cells and on the tumour microenvironment. In addition to inhibition of the autophagic flux, which is the most studied anti-cancer effect of CQ and HCQ, these drugs affect the Toll-like receptor 9, p53 and CXCR4-CXCL12 pathway in cancer cells. In the tumour stroma, CQ was shown to affect the tumour vasculature, cancer-associated fibroblasts and the immune system. The evidence reviewed in this paper indicates that both CQ and HCQ deserve further clinical investigations in several cancer types. Special attention about the drug (CQ versus HCQ), the dose and the schedule of administration should be taken in the design of new trials.
Collapse
Affiliation(s)
- Ciska Verbaanderd
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium.,Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.,Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Hannelore Maes
- Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Marco B Schaaf
- Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc, Newton, MA 02459, USA.,Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Current address: Emory School of Medicine, Atlanta, GA 30322, USA
| | - Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium.,The George Pantziarka TP53 Trust, London KT1 2JP, UK
| | | | - Patrizia Agostinis
- Cell Death Research and Therapy Lab, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | | |
Collapse
|
24
|
Abstract
Autophagy is a mechanism by which cellular material is delivered to lysosomes for degradation, leading to the basal turnover of cell components and providing energy and macromolecular precursors. Autophagy has opposing, context-dependent roles in cancer, and interventions to both stimulate and inhibit autophagy have been proposed as cancer therapies. This has led to the therapeutic targeting of autophagy in cancer to be sometimes viewed as controversial. In this Review, we suggest a way forwards for the effective targeting of autophagy by understanding the context-dependent roles of autophagy and by capitalizing on modern approaches to clinical trial design.
Collapse
Affiliation(s)
- Jean M Mulcahy Levy
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado 80045, USA
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Christina G Towers
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
25
|
Targeting autophagy in cancer stem cells as an anticancer therapy. Cancer Lett 2017; 393:33-39. [DOI: 10.1016/j.canlet.2017.02.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/18/2022]
|
26
|
Resistance to metronomic chemotherapy and ways to overcome it. Cancer Lett 2017; 400:311-318. [PMID: 28259819 DOI: 10.1016/j.canlet.2017.02.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/20/2017] [Accepted: 02/22/2017] [Indexed: 12/24/2022]
Abstract
Therapeutic resistance is amongst the major determinants of cancer mortality. Contrary to initial expectations, antivascular therapies are equally prone to inherent or acquired resistance as other cancer treatment modalities. However, studies into resistance to vascular endothelial growth factor pathway inhibitors revealed distinct mechanisms of resistance compared to conventional cytotoxic therapy. While some of these novel mechanisms of resistance also appear to be functional regarding metronomic chemotherapy, herein we summarize available evidence for mechanisms of resistance specifically described in the context of metronomic chemotherapy. Numerous preclinically identified molecular targets and pathways represent promising avenues to overcome resistance and enhance the benefits achieved with metronomic chemotherapy eventually. However, there are considerable challenges to clinically translate the preclinical findings.
Collapse
|
27
|
Villar VH, Nguyen TL, Delcroix V, Terés S, Bouchecareilh M, Salin B, Bodineau C, Vacher P, Priault M, Soubeyran P, Durán RV. mTORC1 inhibition in cancer cells protects from glutaminolysis-mediated apoptosis during nutrient limitation. Nat Commun 2017; 8:14124. [PMID: 28112156 PMCID: PMC5264013 DOI: 10.1038/ncomms14124] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 12/01/2016] [Indexed: 12/16/2022] Open
Abstract
A master coordinator of cell growth, mTORC1 is activated by different metabolic inputs, particularly the metabolism of glutamine (glutaminolysis), to control a vast range of cellular processes, including autophagy. As a well-recognized tumour promoter, inhibitors of mTORC1 such as rapamycin have been approved as anti-cancer agents, but their overall outcome in patients is rather poor. Here we show that mTORC1 also presents tumour suppressor features in conditions of nutrient restrictions. Thus, the activation of mTORC1 by glutaminolysis during nutritional imbalance inhibits autophagy and induces apoptosis in cancer cells. Importantly, rapamycin treatment reactivates autophagy and prevents the mTORC1-mediated apoptosis. We also observe that the ability of mTORC1 to activate apoptosis is mediated by the adaptor protein p62. Thus, the mTORC1-mediated upregulation of p62 during nutrient imbalance induces the binding of p62 to caspase 8 and the subsequent activation of the caspase pathway. Our data highlight the role of autophagy as a survival mechanism upon rapamycin treatment.
Collapse
Affiliation(s)
- Victor H. Villar
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Tra Ly Nguyen
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Vanessa Delcroix
- Institut Bergonié, INSERM U1218, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Silvia Terés
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Marion Bouchecareilh
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, 1 Rue Camille Saint-Saëns, Bordeaux 33077, France
| | - Bénédicte Salin
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, 1 Rue Camille Saint-Saëns, Bordeaux 33077, France
| | - Clément Bodineau
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Pierre Vacher
- Institut Bergonié, INSERM U1218, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Muriel Priault
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, 1 Rue Camille Saint-Saëns, Bordeaux 33077, France
| | - Pierre Soubeyran
- Institut Bergonié, INSERM U1218, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Raúl V. Durán
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| |
Collapse
|
28
|
Activating autophagy to potentiate immunogenic chemotherapy and radiation therapy. Nat Rev Clin Oncol 2016; 14:247-258. [PMID: 27845767 DOI: 10.1038/nrclinonc.2016.183] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is fundamental to the maintenance of intracellular homeostasis in virtually all human cells. Accordingly, defective autophagy predisposes healthy cells to undergoing malignant transformation. By contrast, malignant cells are able to harness autophagy to thrive, despite adverse microenvironmental conditions, and to resist therapeutic challenges. Thus, inhibition of autophagy has been proposed as a strategy to kill cancer cells or sensitize them to therapy; however, autophagy is also critical for optimal immune function, and mediates cell-extrinsic homeostatic effects owing to its central role in danger signalling by neoplastic cells responding to immunogenic chemotherapy and/or radiation therapy. In this Perspective, we discuss accumulating preclinical and clinical evidence in support of the all-too-often dismissed possibility that activating autophagy might be a relevant clinical objective that enables an increase in the effectiveness of immunogenic chemotherapy and/or radiation therapy.
Collapse
|
29
|
Gil J, Pesz KA, Sąsiadek MM. May autophagy be a novel biomarker and antitumor target in colorectal cancer? Biomark Med 2016; 10:1081-1094. [PMID: 27626110 DOI: 10.2217/bmm-2016-0083] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a catabolic process associated with intracellular self-digestion of damaged organelles or redundant proteins enabling maintenance of cell homeostasis. It is accepted that impaired autophagy is closely linked to cancer development and has been extensively studied in a variety of malignancies including colorectal cancer (CRC) to elucidate its influence on carcinogenesis, metastasis and antitumor therapy response. CRC remains a great epidemiological problem because of poor 5-year survival and treatment resistance. Many studies concerning autophagy in CRC gave inconsistent and contradictory results, illustrating a multifaceted nature of this process. In this review, we focus on current knowledge of autophagy in CRC development to determinate its role as a potential prognostic and predictive biomarker as well as target in antitumor therapy.
Collapse
Affiliation(s)
- Justyna Gil
- Department of Genetics, Wroclaw Medical University, Wroclaw, Poland
| | - Karolina A Pesz
- Department of Genetics, Wroclaw Medical University, Wroclaw, Poland
| | - Maria M Sąsiadek
- Department of Genetics, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
30
|
Steelman LS, Martelli AM, Cocco L, Libra M, Nicoletti F, Abrams SL, McCubrey JA. The therapeutic potential of mTOR inhibitors in breast cancer. Br J Clin Pharmacol 2016; 82:1189-1212. [PMID: 27059645 DOI: 10.1111/bcp.12958] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/16/2022] Open
Abstract
Rapamycin and modified rapamycins (rapalogs) have been used to prevent allograft rejection after organ transplant for over 15 years. The mechanistic target of rapamycin (mTOR) has been determined to be a key component of the mTORC1 complex which consists of the serine/threonine kinase TOR and at least five other proteins which are involved in regulating its activity. Some of the best characterized substrates of mTORC1 are proteins which are key kinases involved in the regulation of cell growth (e.g., p70S6K) and protein translation (e.g., 4E-BP1). These proteins may in some cases serve as indicators to sensitivity to rapamycin-related therapies. Dysregulation of mTORC1 activity frequently occurs due to mutations at, or amplifications of, upstream growth factor receptors (e.g., human epidermal growth factor receptor-2, HER2) as well as kinases (e.g., PI3K) and phosphatases (e.g., PTEN) critical in the regulation of cell growth. More recently, it has been shown that certain rapalogs may enhance the effectiveness of hormonal-based therapies for breast cancer patients who have become resistant to endocrine therapy. The combined treatment of certain rapalogs (e.g., everolimus) and aromatase inhibitors (e.g., exemestane) has been approved by the United States Food and Drug Administration (US FDA) and other drug regulatory agencies to treat estrogen receptor positive (ER+) breast cancer patients who have become resistant to hormonal-based therapies and have progressed. This review will summarize recent basic and clinical research in the area and evaluate potential novel therapeutic approaches.
Collapse
Affiliation(s)
- Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology & Functional Genomics, Section of Pathology & Oncology, University of Catania, Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology & Functional Genomics, Section of Pathology & Oncology, University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA.
| |
Collapse
|