1
|
Paredes-Ruiz D, Martin-Iglesias D, Amo L, Ruiz-Irastorza G. Elucidating the mechanisms and efficacy of antimalarial drugs in systemic lupus erythematosus. Expert Opin Pharmacother 2024; 25:2047-2060. [PMID: 39354741 DOI: 10.1080/14656566.2024.2412252] [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: 06/29/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/03/2024]
Abstract
INTRODUCTION Antimalarials (AMs) are old drugs with a wide range of beneficial effects in systemic lupus erythematosus (SLE) beyond the control of activity. The most recent debate is focused on defining the optimal doses to assure the best benefit/risk ratio. AREAS COVERED We have reviewed the pharmacological basis underlying the various therapeutic effects of AMs and the beneficial and toxic effects of HCQ, also discussing the role of mepacrine not only as a substitute in cases of maculopathy, but also as a very effective therapy combined with HCQ. We searched PubMed and Embase for articles published in English at any time. We used the terms "hydroxychloroquine" or "mepacrine" or "chloroquine" or "antimalarials", "pharmacokinetics", "efficacy", "remission", "toxicity", "adherence". We reviewed original research articles, large observational studies, systematic reviews, and expert consensus statements. Additionally, studies were identified through the assessment of the reference lists of the evaluated manuscripts. EXPERT OPINION We advocate for the widespread use of HCQ at stable doses of 200 mg/d (≤4 mg/kg/d for most patients) and also for the early combination therapy with mepacrine to assure a good control of SLE activity, and also a durable and safe use of these essential drugs for the management of SLE.
Collapse
Affiliation(s)
- Diana Paredes-Ruiz
- Biobizkaia Health Research Institute, Hospital Universitario Cruces, Bizkaia, The Basque Country, Spain
| | - Daniel Martin-Iglesias
- Biobizkaia Health Research Institute, Hospital Universitario Cruces, Bizkaia, The Basque Country, Spain
- Internal Medicine Department, Hospital Universitario de Leon, Leon, Spain
| | - Laura Amo
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Guillermo Ruiz-Irastorza
- Biobizkaia Health Research Institute, Hospital Universitario Cruces, Bizkaia, The Basque Country, Spain
- Department of Medicine, University of The Basque Country, UPV/EHU, Bizkaia, The Basque Country, Spain
| |
Collapse
|
2
|
Liu Y, Wang Y, Zhang J, Peng Q, Wang X, Xiao X, Shi K. Nanotherapeutics targeting autophagy regulation for improved cancer therapy. Acta Pharm Sin B 2024; 14:2447-2474. [PMID: 38828133 PMCID: PMC11143539 DOI: 10.1016/j.apsb.2024.03.019] [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: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/29/2024] [Indexed: 06/05/2024] Open
Abstract
The clinical efficacy of current cancer therapies falls short, and there is a pressing demand to integrate new targets with conventional therapies. Autophagy, a highly conserved self-degradation process, has received considerable attention as an emerging therapeutic target for cancer. With the rapid development of nanomedicine, nanomaterials have been widely utilized in cancer therapy due to their unrivaled delivery performance. Hence, considering the potential benefits of integrating autophagy and nanotechnology in cancer therapy, we outline the latest advances in autophagy-based nanotherapeutics. Based on a brief background related to autophagy and nanotherapeutics and their impact on tumor progression, the feasibility of autophagy-based nanotherapeutics for cancer treatment is demonstrated. Further, emerging nanotherapeutics developed to modulate autophagy are reviewed from the perspective of cell signaling pathways, including modulation of the mammalian target of rapamycin (mTOR) pathway, autophagy-related (ATG) and its complex expression, reactive oxygen species (ROS) and mitophagy, interference with autophagosome-lysosome fusion, and inhibition of hypoxia-mediated autophagy. In addition, combination therapies in which nano-autophagy modulation is combined with chemotherapy, phototherapy, and immunotherapy are also described. Finally, the prospects and challenges of autophagy-based nanotherapeutics for efficient cancer treatment are envisioned.
Collapse
Affiliation(s)
- Yunmeng Liu
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Yaxin Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Jincheng Zhang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Qikai Peng
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xingdong Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xiyue Xiao
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Kai Shi
- College of Pharmacy, Nankai University, Tianjin 300350, China
| |
Collapse
|
3
|
Peng-Cheng L, Meng-Na L, Jian-Bin L, Shu-Jiao Y, Wu R. Advancements on the impact of hydroxychloroquine in systemic lupus erythematosus. Heliyon 2024; 10:e30393. [PMID: 38711668 PMCID: PMC11070867 DOI: 10.1016/j.heliyon.2024.e30393] [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: 01/08/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Hydroxychloroquine (HCQ) has gained significant attention as a therapeutic option for systemic lupus erythematosus (SLE) because of its multifaceted mechanism of action. It is a lipophilic, lysosomotropic drug, that easily traverses cell membranes and accumulates in lysosomes. Once accumulated, HCQ alkalizes lysosomes within the cytoplasm, thereby disrupting their function and interfering with processes like antigen presentation. Additionally, HCQ has shown potential in modulating T-cell responses, inhibiting cytokine production, and influencing Toll-like receptor signaling. Its immunomodulatory effects have generated interest in its application for autoimmune disorders. Despite its established efficacy, uncertainties persist regarding the optimal therapeutic concentrations and their correlation with adverse effects such as retinal toxicity. Therefore, standardized dosing and monitoring guidelines are crucial. In this study, we provide a comprehensive review of the mechanisms, efficacy, dosing variations, and retinal toxicity profiles of HCQ, which are essential to optimize SLE treatment protocols and ensure patient safety.
Collapse
Affiliation(s)
- Liu Peng-Cheng
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lv Meng-Na
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Li Jian-Bin
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yu Shu-Jiao
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Rui Wu
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Giannopoulos S, Bozkus CC, Zografos E, Athanasiou A, Bongiovanni AM, Doulaveris G, Bakoyiannis CN, Theodoropoulos GE, Zografos GC, Witkin SS, Orfanelli T. Targeting Both Autophagy and Immunotherapy in Breast Cancer Treatment. Metabolites 2022; 12:metabo12100966. [PMID: 36295867 PMCID: PMC9607060 DOI: 10.3390/metabo12100966] [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: 09/05/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
As clinical efforts towards breast-conserving therapy and prolonging survival of those with metastatic breast cancer increase, innovative approaches with the use of biologics are on the rise. Two areas of current focus are cancer immunotherapy and autophagy, both of which have been well-studied independently but have recently been shown to have intertwining roles in cancer. An increased understanding of their interactions could provide new insights that result in novel diagnostic, prognostic, and therapeutic strategies. In this breast cancer-focused review, we explore the interactions between autophagy and two clinically relevant immune checkpoint pathways; the programmed cell death-1 receptor with its ligand (PD-L1)/PD-1 and the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4)/CD80 and CD86 (B7-1 and B7-2). Furthermore, we discuss emerging preclinical and clinical data supporting targeting both immunotherapy and autophagy pathway manipulation as a promising approach in the treatment of breast cancer.
Collapse
Affiliation(s)
- Spyridon Giannopoulos
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cansu Cimen Bozkus
- Department of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY 10029, USA
| | - Eleni Zografos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Aikaterini Athanasiou
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ann Marie Bongiovanni
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Georgios Doulaveris
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Chris N Bakoyiannis
- First Department of Surgery, Division of Vascular Surgery, Laikon General Hospital, National Kapodistrian University of Athens, 15772 Athens, Greece
| | - Georgios E Theodoropoulos
- First Department of Propaedeutic Surgery, Hippocration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Georgios C Zografos
- First Department of Propaedeutic Surgery, Hippocration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Steven S Witkin
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Theofano Orfanelli
- First Department of Propaedeutic Surgery, Hippocration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
5
|
Yazal T, Bailleul J, Ruan Y, Sung D, Chu FI, Palomera D, Dao A, Sehgal A, Gurunathan V, Aryan L, Eghbali M, Vlashi E. Radiosensitizing Pancreatic Cancer via Effective Autophagy Inhibition. Mol Cancer Ther 2022; 21:79-88. [PMID: 34725193 DOI: 10.1158/1535-7163.mct-20-1103] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/02/2021] [Accepted: 10/29/2021] [Indexed: 12/09/2022]
Abstract
Despite aggressive treatments, pancreatic ductal adenocarcinoma (PDAC) remains an intractable disease, largely because it is refractory to therapeutic interventions. To overcome its nutrient-poor microenvironment, PDAC heavily relies on autophagy for metabolic needs to promote tumor growth and survival. Here, we explore autophagy inhibition as a method to enhance the effects of radiotherapy on PDAC tumors. Hydroxychloroquine is an autophagy inhibitor at the focus of many PDAC clinical trials, including in combination with radiotherapy. However, its acid-labile properties likely reduce its intratumoral efficacy. Here, we demonstrate that EAD1, a synthesized analogue of HCQ, is a more effective therapeutic for sensitizing PDAC tumors of various KRAS mutations to radiotherapy. Specifically, in vitro models show that EAD1 is an effective inhibitor of autophagic flux in PDAC cells, accompanied by a potent inhibition of proliferation. When combined with radiotherapy, EAD1 is consistently superior to HCQ not only as a single agent, but also in radiosensitizing PDAC cells, and perhaps most importantly, in decreasing the self-renewal capacity of PDAC cancer stem cells (PCSC). The more pronounced sensitizing effects of autophagy inhibitors on pancreatic stem over differentiated cells points to a new understanding that PCSCs may be more dependent on autophagy to counter the effects of radiation toxicity, a potential mechanism explaining the resistance of PCSCs to radiotherapy. Finally, in vivo subcutaneous tumor models demonstrate that combination of radiotherapy and EAD1 is the most successful at controlling tumor growth. The models also confirmed a similar toxicity profile between EAD1 and Hydroxychloroquine.
Collapse
Affiliation(s)
- Taha Yazal
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Yangjingyi Ruan
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - David Sung
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Fang-I Chu
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Daisy Palomera
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Amy Dao
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Anahita Sehgal
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Vibha Gurunathan
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Laila Aryan
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Mansoureh Eghbali
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
6
|
Aminoquinolines as Translational Models for Drug Repurposing: Anticancer Adjuvant Properties and Toxicokinetic-Related Features. JOURNAL OF ONCOLOGY 2021; 2021:3569349. [PMID: 34527050 PMCID: PMC8437624 DOI: 10.1155/2021/3569349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/21/2021] [Indexed: 01/04/2023]
Abstract
The indiscriminate consumption of antimalarials against coronavirus disease-2019 emphasizes the longstanding clinical weapons of medicines. In this work, we conducted a review on the antitumor mechanisms of aminoquinolines, focusing on the responses and differences of tumor histological tissues and toxicity related to pharmacokinetics. This well-defined analysis shows similar mechanistic forms triggered by aminoquinolines in different histological tumor tissues and under coexposure conditions, although different pharmacological potencies also occur. These molecules are lysosomotropic amines that increase the antiproliferative action of chemotherapeutic agents, mainly by cell cycle arrest, histone acetylation, physiological changes in tyrosine kinase metabolism, inhibition of PI3K/Akt/mTOR pathways, cyclin D1, E2F1, angiogenesis, ribosome biogenesis, triggering of ATM-ATR/p53/p21 signaling, apoptosis, and presentation of tumor peptides. Their chemo/radiotherapy sensitization effects may be an adjuvant option against solid tumors, since 4-aminoquinolines induce lysosomal-mediated programmed cytotoxicity of cancer cells and accumulation of key markers, predominantly, LAMP1, p62/SQSTM1, LC3 members, GAPDH, beclin-1/Atg6, α-synuclein, and granules of lipofuscin. Adverse effects are dose-dependent, though most common with chloroquine, hydroxychloroquine, amodiaquine, and other aminoquinolines are gastrointestinal changes, blurred vision ventricular arrhythmias, cardiac arrest, QTc prolongation, severe hypoglycemia with loss of consciousness, and retinopathy, and they are more common with chloroquine than with hydroxychloroquine and amodiaquine due to pharmacokinetic features. Additionally, psychological/neurological effects were also detected during acute or chronic use, but aminoquinolines do not cross the placenta easily and low quantity is found in breast milk despite their long mean residence times, which depends on the coexistence of hepatic diseases (cancer-related or not), first pass metabolism, and comedications. The low cost and availability on the world market have converted aminoquinolines into “star drugs” for pharmaceutical repurposing, but a continuous pharmacovigilance is necessary because these antimalarials have multiple modes of action/unwanted targets, relatively narrow therapeutic windows, recurrent adverse effects, and related poisoning self-treatment. Therefore, their use must obey strict rules, ethical and medical prescriptions, and clinical and laboratory monitoring.
Collapse
|
7
|
High-throughput screening for natural compound-based autophagy modulators reveals novel chemotherapeutic mode of action for arzanol. Cell Death Dis 2021; 12:560. [PMID: 34059630 PMCID: PMC8167120 DOI: 10.1038/s41419-021-03830-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022]
Abstract
Autophagy is an intracellular recycling pathway with implications for intracellular homeostasis and cell survival. Its pharmacological modulation can aid chemotherapy by sensitizing cancer cells toward approved drugs and overcoming chemoresistance. Recent translational data on autophagy modulators show promising results in reducing tumor growth and metastasis, but also reveal a need for more specific compounds and novel lead structures. Here, we searched for such autophagy-modulating compounds in a flow cytometry-based high-throughput screening of an in-house natural compound library. We successfully identified novel inducers and inhibitors of the autophagic pathway. Among these, we identified arzanol as an autophagy-modulating drug that causes the accumulation of ATG16L1-positive structures, while it also induces the accumulation of lipidated LC3. Surprisingly, we observed a reduction of the size of autophagosomes compared to the bafilomycin control and a pronounced accumulation of p62/SQSTM1 in response to arzanol treatment in HeLa cells. We, therefore, speculate that arzanol acts both as an inducer of early autophagosome biogenesis and as an inhibitor of later autophagy events. We further show that arzanol is able to sensitize RT-112 bladder cancer cells towards cisplatin (CDDP). Its anticancer activity was confirmed in monotherapy against both CDDP-sensitive and -resistant bladder cancer cells. We classified arzanol as a novel mitotoxin that induces the fragmentation of mitochondria, and we identified a series of targets for arzanol that involve proteins of the class of mitochondria-associated quinone-binding oxidoreductases. Collectively, our results suggest arzanol as a valuable tool for autophagy research and as a lead compound for drug development in cancer therapy.
Collapse
|
8
|
Ferreira PMP, Sousa RWRD, Ferreira JRDO, Militão GCG, Bezerra DP. Chloroquine and hydroxychloroquine in antitumor therapies based on autophagy-related mechanisms. Pharmacol Res 2021; 168:105582. [PMID: 33775862 DOI: 10.1016/j.phrs.2021.105582] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 02/09/2023]
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are the most common drugs used to relieve acute and chronic inflammatory diseases. In this article, we present a review about the use of CQ and HCQ in antitumor therapies based on autophagy mechanisms. These molecules break/discontinue autophagosome-lysosome fusions in initial phases and enhance antiproliferative action of chemotherapeutics. Their sensitizing effects of chemotherapy when used as an adjuvant option in clinical trials against cancer. However, human related-MDR genes are also under risk to develop chemo or radioresistance because cancer cells have ability to throw 4-aminoquinolines out from digestive vacuoles well. Additionally, they also have antitumor mechanism unrelated to autophagy, including cell death from apoptosis and necroptosis and immunomodulatory/anti-inflammatory properties. However, the link between some anticancer mechanisms, clinical efficacy and pharmacological safety has not yet been fully defined.
Collapse
Affiliation(s)
- Paulo Michel Pinheiro Ferreira
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, 64049-550 Teresina, Brazil.
| | - Rayran Walter Ramos de Sousa
- Department of Biophysics and Physiology, Laboratory of Experimental Cancerology, Federal University of Piauí, 64049-550 Teresina, Brazil
| | | | | | - Daniel Pereira Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ-BA), 40296-710 Salvador, Brazil
| |
Collapse
|
9
|
Xiao M, Benoit A, Hasmim M, Duhem C, Vogin G, Berchem G, Noman MZ, Janji B. Targeting Cytoprotective Autophagy to Enhance Anticancer Therapies. Front Oncol 2021; 11:626309. [PMID: 33718194 PMCID: PMC7951055 DOI: 10.3389/fonc.2021.626309] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/15/2021] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a highly regulated multi-step process that occurs at the basal level in almost all cells. Although the deregulation of the autophagy process has been described in several pathologies, the role of autophagy in cancer as a cytoprotective mechanism is currently well established and supported by experimental and clinical evidence. Our understanding of the molecular mechanism of the autophagy process has largely contributed to defining how we can harness this process to improve the benefit of cancer therapies. While the role of autophagy in tumor resistance to chemotherapy is extensively documented, emerging data point toward autophagy as a mechanism of cancer resistance to radiotherapy, targeted therapy, and immunotherapy. Therefore, manipulating autophagy has emerged as a promising strategy to overcome tumor resistance to various anti-cancer therapies, and autophagy modulators are currently evaluated in combination therapies in several clinical trials. In this review, we will summarize our current knowledge of the impact of genetically and pharmacologically modulating autophagy genes and proteins, involved in the different steps of the autophagy process, on the therapeutic benefit of various cancer therapies. We will also briefly discuss the challenges and limitations to developing potent and selective autophagy inhibitors that could be used in ongoing clinical trials.
Collapse
Affiliation(s)
- Malina Xiao
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Alice Benoit
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Meriem Hasmim
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Caroline Duhem
- Department of Hemato-oncology, Centre Hospitalier du Luxembourg, Luxembourg City, Luxembourg
| | - Guillaume Vogin
- Université de Lorraine - UMR 7365, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Vandoeuvre-lès-Nancy, France.,Centre François Baclesse, Esch-sur-Alzette, Luxembourg
| | - Guy Berchem
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg.,Department of Hemato-oncology, Centre Hospitalier du Luxembourg, Luxembourg City, Luxembourg
| | - Muhammad Zaeem Noman
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| | - Bassam Janji
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg City, Luxembourg
| |
Collapse
|
10
|
Tripathy S, Dassarma B, Roy S, Chabalala H, Matsabisa MG. A review on possible modes of action of chloroquine/hydroxychloroquine: repurposing against SAR-CoV-2 (COVID-19) pandemic. Int J Antimicrob Agents 2020; 56:106028. [PMID: 32450198 PMCID: PMC7243790 DOI: 10.1016/j.ijantimicag.2020.106028] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
Chloroquine (CQ) and its analogue hydroxychloroquine (HCQ) have long been used worldwide as frontline drugs for the treatment and prophylaxis of human malaria. Since the first reported cases in Wuhan, China, in late December 2019, humans have been under threat from coronavirus disease 2019 (COVID-19) caused by the novel coronavirus SARS-CoV-2 (previously known as 2019-nCoV), subsequently declared a pandemic. While the world is searching for expedited approval for a vaccine, which may be only preventative and not a cure, physicians and country leaders are considering several concerted clinical trials suggesting that the age-old antimalarial drugs CQ/HCQ could be a potent therapeutic against COVID-19. Based on accumulating scientific reports, here we highlight the possible modes of action of CQ/HCQ that could justify its use against viral infections. Considering the global health crisis of the COVID-19 pandemic, the option of repurposing old drugs, e.g. CQ/HCQ, particularly HCQ, for the treatment of SARS-CoV-2 infection could be a good choice. CQ/HCQ has diverse modes of action, including alteration of the acidic environment inside lysosomes and late endosomes, preventing endocytosis, exosome release and phagolysosomal fusion, and inhibition of the host cytokine storm. One or more diverse mechanisms might work against viral infections and reduce mortality. As there is no cure for COVID-19, clinical testing of HCQ is urgently required to determine its potency against SARS-CoV-2, as this is the currently available treatment option. There remains a need to find other innovative drug candidates as possible candidates to enter clinical evaluation and testing.
Collapse
Affiliation(s)
- Satyajit Tripathy
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Barsha Dassarma
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Somenath Roy
- Ex-Professor, Department of Human Physiology with Community Health, Vidyasagar University, Paschim Medinipur 721102, India
| | - Hlupheka Chabalala
- IK-based Technology Innovations, Department of Science and Innovations, Brummeria, Pretoria 0001, South Africa
| | - Motlalepula Gilbert Matsabisa
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa.
| |
Collapse
|
11
|
Nirk EL, Reggiori F, Mauthe M. Hydroxychloroquine in rheumatic autoimmune disorders and beyond. EMBO Mol Med 2020; 12:e12476. [PMID: 32715647 PMCID: PMC7411564 DOI: 10.15252/emmm.202012476] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022] Open
Abstract
Initially used as antimalarial drugs, hydroxychloroquine (HCQ) and, to a lesser extent, chloroquine (CQ) are currently being used to treat several diseases. Due to its cost‐effectiveness, safety and efficacy, HCQ is especially used in rheumatic autoimmune disorders (RADs), such as systemic lupus erythematosus, primary Sjögren's syndrome and rheumatoid arthritis. Despite this widespread use in the clinic, HCQ molecular modes of action are still not completely understood. By influencing several cellular pathways through different mechanisms, CQ and HCQ inhibit multiple endolysosomal functions, including autophagy, as well as endosomal Toll‐like receptor activation and calcium signalling. These effects alter several aspects of the immune system with the synergistic consequence of reducing pro‐inflammatory cytokine production and release, one of the most marked symptoms of RADs. Here, we review the current knowledge on the molecular modes of action of these drugs and the circumstances under which they trigger side effects. This is of particular importance as the therapeutic use of HCQ is expanding beyond the treatment of malaria and RADs.
Collapse
Affiliation(s)
- Eliise Laura Nirk
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mario Mauthe
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Afolayan FI, Adegbolagun O, Mwikwabe NN, Orwa J, Anumudu C. Cytokine modulation during malaria infections by some medicinal plants. SCIENTIFIC AFRICAN 2020. [DOI: 10.1016/j.sciaf.2020.e00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
13
|
Stagni V, Kaminari A, Sideratou Z, Sakellis E, Vlahopoulos SA, Tsiourvas D. Targeting breast cancer stem-like cells using chloroquine encapsulated by a triphenylphosphonium-functionalized hyperbranched polymer. Int J Pharm 2020; 585:119465. [PMID: 32497731 DOI: 10.1016/j.ijpharm.2020.119465] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 01/07/2023]
Abstract
Cancer stem cells (CSCs) have garnered increasing attention over the past decade, as they are believed to play a crucial role in tumor progression and drug resistance. Accumulating evidence provides insight into the function of autophagy in maintenance and survival of CSCs. Here, we studied the impact of a mitochondriotropic triphenylphosphonium-functionalized dendrimeric nanocarrier on cultured breast cancer cell lines, grown either as adherent cells or as mammospheres that mimic a stem-like phenotype. The nanocarrier manifested a substantial cytotoxicity both alone as well as after encapsulation of chloroquine, a well-known autophagy inhibitor. The cytotoxic effects of the nanocarrier could be ascribed to interference with mitochondrial function. Importantly, mammospheres were selectively sensitive to encapsulated chloroquine and this depends on the expression of the gene encoding ATM kinase. Ataxia-telangiectasia mutated (ATM) kinase is an enzyme that functions as an essential signaling mediator that enables growth of cancer stem cells through the regulation of autophagy. We noted that this ATM-dependent sensitivity of mammospheres to encapsulated chloroquine was independent of the status of the tumor suppressor gene p53. Our study suggests that breast cancer stem cells, as they are modeled by mammospheres, are sensitive to encapsulated chloroquine, depending on the expression of the ATM kinase, which is thereby characterized as a potential biomarker for sensitivity to this type of treatment.
Collapse
Affiliation(s)
- Venturina Stagni
- Laboratory of Cell Signaling, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy; Institute of Molecular Biology and Pathology, National Research Council (CNR), Via DegliApuli 4, 00185 Rome, Italy.
| | - Archontia Kaminari
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Zili Sideratou
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Elias Sakellis
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Spiros A Vlahopoulos
- Ηoremeio Research Laboratory, First Department of Paediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitris Tsiourvas
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece.
| |
Collapse
|
14
|
Varisli L, Cen O, Vlahopoulos S. Dissecting pharmacological effects of chloroquine in cancer treatment: interference with inflammatory signaling pathways. Immunology 2020; 159:257-278. [PMID: 31782148 PMCID: PMC7011648 DOI: 10.1111/imm.13160] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Chloroquines are 4-aminoquinoline-based drugs mainly used to treat malaria. At pharmacological concentrations, they have significant effects on tissue homeostasis, targeting diverse signaling pathways in mammalian cells. A key target pathway is autophagy, which regulates macromolecule turnover in the cell. In addition to affecting cellular metabolism and bioenergetic flow equilibrium, autophagy plays a pivotal role at the interface between inflammation and cancer progression. Chloroquines consequently have critical effects in tissue metabolic activity and importantly, in key functions of the immune system. In this article, we will review the work addressing the role of chloroquines in the homeostasis of mammalian tissue, and the potential strengths and weaknesses concerning their use in cancer therapy.
Collapse
Affiliation(s)
- Lokman Varisli
- Union of Education and Science Workers (EGITIM SEN), Diyarbakir Branch, Diyarbakir, Turkey
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, Turkey
| | - Osman Cen
- Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Natural Sciences, Joliet Jr College, Joliet, IL, USA
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Gao L, Zheng H, Cai Q, Wei L. Autophagy and Tumour Radiotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:375-387. [PMID: 32671760 DOI: 10.1007/978-981-15-4272-5_25] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Radiotherapy is an important component of cancer treatment modalities. With the rapid development of three-dimensional conformal, intensity-modulated, image-guided radiotherapy and the efficacy of radiotherapy continues to improve. Autophagy, as a catabolic process, is characterized by the formation of a double-membrane vesicle. Radiotherapy is known to induce autophagy in both cancer and normal cells. Here, we reviewed the interaction of radiotherapy and autophagy in the process of cancer treatment. The potential role of autophagy modification in enhancing radiotherapy treatment will also be reviewed.
Collapse
Affiliation(s)
- Lu Gao
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Huifei Zheng
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Quanyu Cai
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China.
| |
Collapse
|
16
|
Hounjet J, Habets R, Schaaf MB, Hendrickx TC, Barbeau LMO, Yahyanejad S, Rouschop KM, Groot AJ, Vooijs M. The anti-malarial drug chloroquine sensitizes oncogenic NOTCH1 driven human T-ALL to γ-secretase inhibition. Oncogene 2019; 38:5457-5468. [PMID: 30967635 DOI: 10.1038/s41388-019-0802-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 01/30/2019] [Accepted: 03/19/2019] [Indexed: 12/26/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer arising from T-cell progenitors. Although current treatments, including chemotherapy and glucocorticoids, have significantly improved survival, T-ALL remains a fatal disease and new treatment options are needed. Since more than 60% of T-ALL cases bear oncogenic NOTCH1 mutations, small molecule inhibitors of NOTCH1 signalling; γ-secretase inhibitors (GSI), are being actively investigated for the treatment of T-ALL. Unfortunately, GSI have shown limited clinical efficacy and dose-limiting toxicities. We hypothesized that by combining known drugs, blocking NOTCH activity through another mechanism, may synergize with GSI enabling equal efficacy at a lower concentration. Here, we show that the clinically used anti-malarial drug chloroquine (CQ), an inhibitor of lysosomal function and autophagy, decreases T-ALL cell viability and proliferation. This effect of CQ was not observed in GSI-resistant T-ALL cell lines. Mechanistically, CQ impairs the redox balance, induces ds DNA breaks and activates the DNA damage response. CQ also interferes with intracellular trafficking and processing of oncogenic NOTCH1. Interestingly, we show for the first time that the addition of CQ to γ-secretase inhibition has a synergistic therapeutic effect on T-ALL and reduces the concentration of GSI required to obtain a reduction in cell viability and a block of proliferation. Overall, our results suggest that CQ may be a promising repurposed drug in the treatment of T-ALL, as a single treatment or in combination with GSI, increasing the therapeutic ratio.
Collapse
Affiliation(s)
- Judith Hounjet
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands.,MAASTRO Clinic, Maastricht, The Netherlands
| | - Roger Habets
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Marco B Schaaf
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Tessa C Hendrickx
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Lydie M O Barbeau
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Sanaz Yahyanejad
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Kasper M Rouschop
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Arjan J Groot
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Marc Vooijs
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
17
|
Folkerts H, Hilgendorf S, Vellenga E, Bremer E, Wiersma VR. The multifaceted role of autophagy in cancer and the microenvironment. Med Res Rev 2018; 39:517-560. [PMID: 30302772 PMCID: PMC6585651 DOI: 10.1002/med.21531] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 12/12/2022]
Abstract
Autophagy is a crucial recycling process that is increasingly being recognized as an important factor in cancer initiation, cancer (stem) cell maintenance as well as the development of resistance to cancer therapy in both solid and hematological malignancies. Furthermore, it is being recognized that autophagy also plays a crucial and sometimes opposing role in the complex cancer microenvironment. For instance, autophagy in stromal cells such as fibroblasts contributes to tumorigenesis by generating and supplying nutrients to cancerous cells. Reversely, autophagy in immune cells appears to contribute to tumor‐localized immune responses and among others regulates antigen presentation to and by immune cells. Autophagy also directly regulates T and natural killer cell activity and is required for mounting T‐cell memory responses. Thus, within the tumor microenvironment autophagy has a multifaceted role that, depending on the context, may help drive tumorigenesis or may help to support anticancer immune responses. This multifaceted role should be taken into account when designing autophagy‐based cancer therapeutics. In this review, we provide an overview of the diverse facets of autophagy in cancer cells and nonmalignant cells in the cancer microenvironment. Second, we will attempt to integrate and provide a unified view of how these various aspects can be therapeutically exploited for cancer therapy.
Collapse
Affiliation(s)
- Hendrik Folkerts
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Susan Hilgendorf
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edo Vellenga
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Edwin Bremer
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Valerie R Wiersma
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
18
|
Autophagy therapeutics: preclinical basis and initial clinical studies. Cancer Chemother Pharmacol 2018; 82:923-934. [PMID: 30225602 DOI: 10.1007/s00280-018-3688-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Autophagy captures and degrades intracellular components such as proteins and organelles to sustain metabolism and homeostasis. Rapidly accumulating attention is being paid to the role of autophagy in the development of cancer, which makes autophagy attractive tools and targets for novel therapeutic approaches. Functional studies have confirmed that autophagy dysregulation is causal in many cases of cancer, with autophagy acting as tumor suppressors or tumor promoters, and autophagy inhibitor or promoter has shown promise in preclinical studies. The autophagy-targeted therapeutics using chloroquine/hydroxychloroquine have reached clinical development for treating cancer, but these drugs are actually not efficient probably because of a reduced penetration within the tumor. In this review, we first discuss the discoveries related to dual function of autophagy in cancer. Then, we provide an overview of preclinical studies and clinical trials involved in the development of autophagy therapeutics and finally discuss the future of such therapies.
Collapse
|
19
|
Current and Future Use of Chloroquine and Hydroxychloroquine in Infectious, Immune, Neoplastic, and Neurological Diseases: A Mini-Review. Clin Drug Investig 2018; 38:653-671. [DOI: 10.1007/s40261-018-0656-y] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
Kucharewicz K, Dudkowska M, Zawadzka A, Ogrodnik M, Szczepankiewicz AA, Czarnocki Z, Sikora E. Simultaneous induction and blockade of autophagy by a single agent. Cell Death Dis 2018; 9:353. [PMID: 29500364 PMCID: PMC5834631 DOI: 10.1038/s41419-018-0383-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 11/10/2022]
Abstract
Besides cell death, autophagy and cell senescence are the main outcomes of anticancer treatment. We demonstrate that tacrine-melatonin heterodimer C10, a potent anti-Alzheimer’s disease drug, has an antiproliferative effect on MCF-7 breast cancer cells. The main cell response to a 24 h-treatment with C10 was autophagy enhancement accompanied by inhibition of mTOR and AKT pathways. Significantly increased autophagy markers, such as LC3B- and ATG16L-positive vesicles, confirmed autophagy induction by C10. However, analysis of autophagic flux using mCherry-GFP-LC3B construct revealed inhibition of autophagy by C10 at the late-stage. Moreover, electron microscopy and analysis of colocalization of LC3B and LAMP-1 proteins provided evidence of autophagosome-lysosome fusion with concomitant inhibition of autolysosomal degradation function. After transient treatment with IC50 dose of C10 followed by cell culture without the drug, 20% of MCF-7 cells displayed markers of senescence. On the other hand, permanent cell treatment with C10 resulted in massive cell death on the 5th or 6th day. Recently, an approach whereby autophagy is induced by one compound and simultaneously blocked by the use of another one has been proposed as a novel anticancer strategy. We demonstrate that the same effect may be achieved using a single agent, C10. Our findings offer a new, promising strategy for anticancer treatment.
Collapse
Affiliation(s)
- Karolina Kucharewicz
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Magdalena Dudkowska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Anna Zawadzka
- Laboratory of Natural Products Chemistry, Faculty of Chemistry, University of Warsaw, 1 Pasteur Street, 02-093, Warsaw, Poland
| | - Mikolaj Ogrodnik
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.,Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Andrzej A Szczepankiewicz
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Zbigniew Czarnocki
- Laboratory of Natural Products Chemistry, Faculty of Chemistry, University of Warsaw, 1 Pasteur Street, 02-093, Warsaw, Poland
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| |
Collapse
|
21
|
Autophagy-associated immune responses and cancer immunotherapy. Oncotarget 2018; 7:21235-46. [PMID: 26788909 PMCID: PMC5008281 DOI: 10.18632/oncotarget.6908] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/06/2016] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an evolutionarily conserved catabolic process by which cellular components are sequestered into a double-membrane vesicle and delivered to the lysosome for terminal degradation and recycling. Accumulating evidence suggests that autophagy plays a critical role in cell survival, senescence and homeostasis, and its dysregulation is associated with a variety of diseases including cancer, cardiovascular disease, neurodegeneration. Recent studies show that autophagy is also an important regulator of cell immune response. However, the mechanism by which autophagy regulates tumor immune responses remains elusive. In this review, we will describe the role of autophagy in immune regulation and summarize the possible molecular mechanisms that are currently well documented in the ability of autophagy to control cell immune response. In addition, the scientific and clinical hurdles regarding the potential role of autophagy in cancer immunotherapy will be discussed.
Collapse
|
22
|
Liu S, Cai X, Xia L, Jiang C, Chen P, Wang X, Zhang B, Zhao HY. Chloroquine exerts antitumor effects on NB4 acute promyelocytic leukemia cells and functions synergistically with arsenic trioxide. Oncol Lett 2017; 15:2024-2030. [PMID: 29434902 DOI: 10.3892/ol.2017.7488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 10/26/2017] [Indexed: 01/17/2023] Open
Abstract
Chloroquine (CQ) has been confirmed to exhibit antitumor effects on different types of cancer cell, but whether it exerts the same effect on acute promyelocytic leukemia (APL) cells remains to be confirmed. In the present study, the effects of various concentrations of CQ on the growth, apoptosis and cell cycle distribution of NB4 cells, as well as the potential mechanisms underlying these effects, were examined. The combined effect of CQ and arsenic trioxide (ATO) on the growth of NB4 cells was also determined. The results of the present study demonstrated that CQ treatment inhibited cell proliferation, and induced mitochondrial pathway apoptosis and S phase arrest in a dose-dependent manner by regulating apoptosis- and cell cycle-related proteins. CQ and ATO had a synergistic effect on the growth inhibition of NB4 cells, which may have been induced through the inhibition of autophagy. In conclusion, the results of the present study indicated that CQ exhibits a cytotoxic effect on NB4 cells and has a synergistic effect when combined with ATO, which thereby improves the curative effect of ATO on APL.
Collapse
Affiliation(s)
- Shousheng Liu
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Xiuyu Cai
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Liangping Xia
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Chang Jiang
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Ping Chen
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaopai Wang
- Department of Pathology, Guangzhou First People's Hospital, Guangzhou, Guangdong 510080, P.R. China
| | - Bei Zhang
- Department of General Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| | - Hong Yun Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China
| |
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: 178] [Impact Index Per Article: 22.3] [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
|
McBride WH, Ganapathy E, Lee MH, Nesseler JP, Nguyen C, Schaue D. A perspective on the impact of radiation therapy on the immune rheostat. Br J Radiol 2017; 90:20170272. [PMID: 28707537 PMCID: PMC5853348 DOI: 10.1259/bjr.20170272] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The advent and success of immune checkpoint inhibitors (ICIs) in cancer treatment has broadened the spectrum of tumours that might be considered "immunogenic" and susceptible to immunotherapeutic (IT) intervention. Not all cancer types are sensitive, and not all patients with any given type respond. Combination treatment of ICIs with an established cytotoxic modality such as radiation therapy (RT) is a logical step towards improvement. For one, RT alone has been shown to be genuinely immunomodulatory and secondly pre-clinical data generally support combined ICI-RT approaches. This new integrated therapy for cancer treatment holds much promise, although there is still a lot to be learned about how best to schedule the treatments, manage the toxicities and determine what biomarkers might predict response, as well as many other issues. This review examines how RT alters the immune rheostat and how it might best be positioned to fully exploit IT.
Collapse
Affiliation(s)
- William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ekambaram Ganapathy
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Mi-Heon Lee
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jean P Nesseler
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Christine Nguyen
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
25
|
An J, Minie M, Sasaki T, Woodward JJ, Elkon KB. Antimalarial Drugs as Immune Modulators: New Mechanisms for Old Drugs. Annu Rev Med 2016; 68:317-330. [PMID: 27813878 DOI: 10.1146/annurev-med-043015-123453] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The best known of the naturally occurring antimalarial compounds are quinine, extracted from cinchona bark, and artemisinin (qinghao), extracted from Artemisia annua in China. These and other derivatives are now chemically synthesized and remain the mainstay of therapy to treat malaria. The beneficial effects of several of the antimalarial drugs (AMDs) on clinical features of autoimmune disorders were discovered by chance during World War II. In this review, we discuss the chemistry of AMDs and their mechanisms of action, emphasizing how they may impact multiple pathways of innate immunity. These pathways include Toll-like receptors and the recently described cGAS-STING pathway. Finally, we discuss the current and future impact of AMDs on systemic lupus erythematosus, rheumatoid arthritis, and devastating monogenic disorders (interferonopathies) characterized by expression of type I interferon in the brain.
Collapse
Affiliation(s)
- Jie An
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington 98195; email
| | | | | | | | - Keith B Elkon
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington 98195; email .,Department of Immunology, University of Washington, Seattle, Washington 98195; , , ,
| |
Collapse
|
26
|
Hu L, Wang H, Huang L, Zhao Y, Wang J. Crosstalk between autophagy and intracellular radiation response (Review). Int J Oncol 2016; 49:2217-2226. [PMID: 27748893 DOI: 10.3892/ijo.2016.3719] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/27/2016] [Indexed: 11/06/2022] Open
Abstract
Autophagy induced by radiation is critical to cell fate decision. Evidence now sheds light on the importance of autophagy induced by cancer radiotherapy. Traditional view considers radiation can directly or indirectly damage DNA which can activate DNA damage the repair signaling pathway, a large number of proteins participating in DNA damage repair signaling pathway such as p53, ATM, PARP1, FOXO3a, mTOR and SIRT1 involved in autophagy regulation. However, emerging recent evidence suggests radiation can also cause injury to extranuclear targets such as plasma membrane, mitochondria and endoplasmic reticulum (ER) and induce accumulation of ceramide, ROS, and Ca2+ concentration which activate many signaling pathways to modulate autophagy. Herein we review the role of autophagy in radiation therapy and the potent intracellular autophagic triggers induced by radiation. We aim to provide a more theoretical basis of radiation-induced autophagy, and provide novel targets for developing cytotoxic drugs to increase radiosensitivity.
Collapse
Affiliation(s)
- Lelin Hu
- Department of Radiation Oncology, Peking University Third Hospital, Haidian, Beijing 100191, P.R. China
| | - Hao Wang
- Department of Radiation Oncology, Peking University Third Hospital, Haidian, Beijing 100191, P.R. China
| | - Li Huang
- Department of Radiation Oncology, Peking University Third Hospital, Haidian, Beijing 100191, P.R. China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Haidian, Beijing 100191, P.R. China
| |
Collapse
|
27
|
HU TAO, LI PEI, LUO ZHONGGUANG, CHEN XIAOYU, ZHANG JINGYANG, WANG CHUNYAO, CHEN PING, DONG ZIMING. Chloroquine inhibits hepatocellular carcinoma cell growth in vitro and in vivo. Oncol Rep 2016; 35:43-9. [PMID: 26530158 PMCID: PMC4699623 DOI: 10.3892/or.2015.4380] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/29/2015] [Indexed: 01/07/2023] Open
Abstract
Recently, chloroquine (CQ) has been widely used to improve the efficacy of different chemotherapy drugs to treat tumors. However, the effects of single treatment of CQ on liver cancer have not been investigated. In the present study, we examined the effects of CQ on the growth and viability of liver cancer cells in vitro and in vivo, and revealed that CQ treatment triggered G0/G1 cell cycle arrest, induced DNA damage and apoptosis in a dose- and time-dependent manner in liver cancer cells. Moreover, administration of CQ to tumor-bearing mice suppressed the tumor growth in an orthotopic xenograft model of liver cancer. These findings extend our understanding and suggest that CQ could be repositioned as a treatment option for liver cancer as a single treatment or in combination.
Collapse
Affiliation(s)
- TAO HU
- College of Basic Medical Sciences, Zhengzhou University; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China
- Laboratory Animal Center, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - PEI LI
- College of Basic Medical Sciences, Zhengzhou University; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China
| | - ZHONGGUANG LUO
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - XIAOYU CHEN
- College of Basic Medical Sciences, Zhengzhou University; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China
| | - JINGYANG ZHANG
- College of Basic Medical Sciences, Zhengzhou University; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China
| | - CHUNYAO WANG
- Laboratory Animal Center, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - PING CHEN
- College of Basic Medical Sciences, Zhengzhou University; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China
- Correspondence to: Dr Ziming Dong or Dr Ping Chen, College of Basic Medical Sciences, Zhengzhou university; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China, E-mail: , E-mail:
| | - ZIMING DONG
- College of Basic Medical Sciences, Zhengzhou University; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China
- Correspondence to: Dr Ziming Dong or Dr Ping Chen, College of Basic Medical Sciences, Zhengzhou university; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, P.R. China, E-mail: , E-mail:
| |
Collapse
|
28
|
Time to use a dose of Chloroquine as an adjuvant to anti-cancer chemotherapies. Eur J Pharmacol 2015; 771:139-44. [PMID: 26687632 DOI: 10.1016/j.ejphar.2015.12.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/27/2015] [Accepted: 12/09/2015] [Indexed: 01/05/2023]
Abstract
Chloroquine, a drug used for over 80 years to treat and prevent malaria and, more recently, to treat autoimmune diseases, is very safe but has a plethora of dose-dependent effects. By increasing pH in acidic compartments it inhibits for example lysosomal enzymes. In the context of cancer, Chloroquine was found to have direct effects on different types of malignancies that could potentiate chemotherapies. For example, the anti-malaria drug may inhibit both the multidrug-resistance pump and autophagy (mechanisms that tumor cells may use to resist chemotherapies), intercalate in DNA and enhance the penetration of chemotherapeutic drugs in cells or solid cancer tissues. However, these activities were mostly demonstrated at high doses of Chloroquine (higher than 10mg/kg or 10mg/l i.e. ca. 31μM). Nevertheless, it was reported that daily uptake of clinically acceptable doses (less than 10mg/kg) of Chloroquine in addition to chemo-radio-therapy increases the survival of glioblastoma patients (Sotelo et al., 2006; Briceno et al., 2007). However, the optimal dose and schedule of this multi-active drug with respect to chemotherapy has never been experimentally determined. The present article reviews the several known direct and indirect effects of different doses of Chloroquine on cancer and how those effects may indicate that a fine tuning of the dose/schedule of Chloroquine administration versus chemotherapy may be critical to obtain an adjuvant effect of Chloroquine in anti-cancer treatments. We anticipate that the appropriate (time and dose) addition of Chloroquine to the standard of care may greatly and safely potentiate current anti-cancer treatments.
Collapse
|
29
|
Maes H, Kuchnio A, Carmeliet P, Agostinis P. Chloroquine anticancer activity is mediated by autophagy-independent effects on the tumor vasculature. Mol Cell Oncol 2015; 3:e970097. [PMID: 27308577 PMCID: PMC4845249 DOI: 10.4161/23723548.2014.970097] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 09/10/2014] [Accepted: 09/10/2014] [Indexed: 11/19/2022]
Abstract
Chloroquine is used clinically as an autophagy blocker to potentiate anticancer treatments. However, whether chloroquine acts solely through autophagy-dependent and cancer cell autonomous mechanisms has remained elusive. In a recent study we found that chloroquine reduced intratumoral hypoxia and metastasis, while improving chemotherapy response, largely through an autophagy-independent, NOTCH1-reliant mechanism of tumor vessel normalization.
Collapse
Affiliation(s)
- Hannelore Maes
- Department Cellular and Molecular Medicine; Laboratory of Cell Death and Therapy ; KU Leuven, Leuven, Belgium
| | - Anna Kuchnio
- Department of Oncology; Laboratory of Angiogenesis and Neurovascular Link; KU Leuven, Leuven, Belgium; Vesalius Research Center; Laboratory of Angiogenesis and Neurovascular Link; VIB, Leuven, Belgium
| | - Peter Carmeliet
- Department of Oncology; Laboratory of Angiogenesis and Neurovascular Link; KU Leuven, Leuven, Belgium; Vesalius Research Center; Laboratory of Angiogenesis and Neurovascular Link; VIB, Leuven, Belgium
| | - Patrizia Agostinis
- Department Cellular and Molecular Medicine; Laboratory of Cell Death and Therapy ; KU Leuven, Leuven, Belgium
| |
Collapse
|
30
|
Schaue D, McBride WH. Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol 2015; 12:527-40. [PMID: 26122185 DOI: 10.1038/nrclinonc.2015.120] [Citation(s) in RCA: 496] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The past 20 years have seen dramatic changes in the delivery of radiation therapy, but the impact of radiobiology on the clinic has been far less substantial. A major consideration in the use of radiotherapy has been on how best to exploit differences between the tumour and host tissue characteristics, which in the past has been achieved empirically by radiation-dose fractionation. New advances are uncovering some of the mechanistic processes that underlie this success story. In this Review, we focus on how these processes might be targeted to improve the outcome of radiotherapy at the individual patient level. This approach would seem a more productive avenue of treatment than simply trying to increase the radiation dose delivered to the tumour.
Collapse
Affiliation(s)
- Dörthe Schaue
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| | - William H McBride
- Department of Radiation Oncology, Room B3-109, Center for Health Sciences, Westwood, University of California, Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA
| |
Collapse
|
31
|
Murray D, McBride WH, Schwartz JL. Radiation biology in the context of changing patterns of radiotherapy. Radiat Res 2014; 182:259-72. [PMID: 25029108 DOI: 10.1667/rr13740.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The last decade has witnessed a revolution in the clinical application of high-dose "ablative" radiation therapy. Initially this approach was limited to the treatment of brain tumors, but more recently we have seen its successful extension to tumors outside the brain, e.g., for small lung nodules. These advances have been driven largely by improvements in image-guided inverse treatment planning that allow the dose per fraction to the tumor to be increased over the conventional 2 Gy dose while keeping the late normal tissue complications at an acceptable level by dose limitation. Despite initial concerns about excessive late complications, as might be expected based on dose extrapolations using the linear-quadratic equation, these approaches have shown considerable clinical promise. Our knowledge of the biological consequences of high-doses of ionizing radiation in normal and cancerous tissues has lagged behind these clinical advances. Our intent here is to survey recent experimental findings from the perspective of better understanding the biological effects of high-dose therapy and whether they are truly different from conventional doses. We will also consider the implications of this knowledge for further refining and improving these approaches on the basis of underlying mechanisms.
Collapse
Affiliation(s)
- David Murray
- a Department of Oncology, Division of Experimental Oncology, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
32
|
Nagelkerke A, Bussink J, Geurts-Moespot A, Sweep FCGJ, Span PN. Therapeutic targeting of autophagy in cancer. Part II: pharmacological modulation of treatment-induced autophagy. Semin Cancer Biol 2014; 31:99-105. [PMID: 24933034 DOI: 10.1016/j.semcancer.2014.06.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Autophagy, the catabolic pathway in which cells recycle organelles and other parts of their own cytoplasm, is increasingly recognised as an important cytoprotective mechanism in cancer cells. Several cancer treatments stimulate the autophagic process and when autophagy is inhibited, cancer cells show an enhanced response to multiple treatments. These findings have nourished the theory that autophagy provides cancer cells with a survival advantage during stressful conditions, including exposure to therapeutics. Therefore, interference with the autophagic response can potentially enhance the efficacy of cancer therapy. In this review we examine two approaches to modulate autophagy as complementary cancer treatment: inhibition and induction. Inhibition of autophagy during cancer treatment eliminates its cytoprotective effects. Conversely, induction of autophagy combined with conventional cancer therapy exerts severe cytoplasmic degradation that can ultimately lead to cell death. We will discuss how autophagy can be therapeutically manipulated in cancer cells and how interactions between the conventional cancer therapies and autophagy modulation influence treatment outcome.
Collapse
Affiliation(s)
- Anika Nagelkerke
- Department of Laboratory Medicine, Radboud university medical center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Department of Radiation Oncology, Radboud university medical center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Johan Bussink
- Department of Radiation Oncology, Radboud university medical center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Anneke Geurts-Moespot
- Department of Laboratory Medicine, Radboud university medical center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboud university medical center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Paul N Span
- Department of Radiation Oncology, Radboud university medical center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|