1
|
Lavudi K, Nuguri SM, Pandey P, Kokkanti RR, Wang QE. ALDH and cancer stem cells: Pathways, challenges, and future directions in targeted therapy. Life Sci 2024; 356:123033. [PMID: 39222837 DOI: 10.1016/j.lfs.2024.123033] [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: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Human ALDH comprise 19 subfamilies in which ALDH1A1, ALDH1A3, ALDH3A1, ALDH5A1, ALDH7A1, and ALDH18A1 are implicated in CSC. Studies have shown that ALDH can also be involved in drug resistance and standard chemotherapy regimens are ineffective in treating patients at the stage of disease recurrence. Existing chemotherapeutic drugs eliminate the bulk of tumors but are usually not effective against CSC which express ALDH+ population. Henceforth, targeting ALDH is convincing to treat the patient's post-relapse. Combination therapies that interlink signaling mechanisms seem promising to increase the overall disease-free survival rate. Therefore, targeting ALDH through ALDH inhibitors along with immunotherapies may create a novel platform for translational research. This review aims to fill in the gap between ALDH1 family members in relation to its cell signaling mechanisms, highlighting their potential as molecular targets to sensitize recurrent tumors and bring forward the future development concerning the current progress and draw backs. This review summarizes the role of cancer stem cells and their upregulation by maintaining the tumor microenvironment in which ALDH is specifically highlighted. It discusses the regulation of ALDH family proteins and the crosstalk between ALDH and CSC in relation to cancer metabolism. Furthermore, it establishes the correlation between ALDH involved signaling mechanisms and their specific targeted inhibitors, as well as their functional modularity, bioavailability, and mechanistic role in various cancers.
Collapse
Affiliation(s)
- Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States
| | - Shreya Madhav Nuguri
- Department of Food science and Technology, The Ohio State University, Columbus, OH, United States
| | - Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, U.P., India; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | | | - Qi-En Wang
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
2
|
Esposito M, Amory JK, Kang Y. The pathogenic role of retinoid nuclear receptor signaling in cancer and metabolic syndromes. J Exp Med 2024; 221:e20240519. [PMID: 39133222 PMCID: PMC11318670 DOI: 10.1084/jem.20240519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024] Open
Abstract
The retinoid nuclear receptor pathway, activated by the vitamin A metabolite retinoic acid, has been extensively investigated for over a century. This study has resulted in conflicting hypotheses about how the pathway regulates health and how it should be pharmaceutically manipulated. These disagreements arise from a fundamental contradiction: retinoid agonists offer clear benefits to select patients with rare bone growth disorders, acute promyelocytic leukemia, and some dermatologic diseases, yet therapeutic retinoid pathway activation frequently causes more harm than good, both through acute metabolic dysregulation and a delayed cancer-promoting effect. In this review, we discuss controlled clinical, mechanistic, and genetic data to suggest several disease settings where inhibition of the retinoid pathway may be a compelling therapeutic strategy, such as solid cancers or metabolic syndromes, and also caution against continued testing of retinoid agonists in cancer patients. Considerable evidence suggests a central role for retinoid regulation of immunity and metabolism, with therapeutic opportunities to antagonize retinoid signaling proposed in cancer, diabetes, and obesity.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Kayothera, Inc , Seattle, WA, USA
| | | | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Institute for Cancer Research Princeton Branch , Princeton, NJ, USA
| |
Collapse
|
3
|
Takahashi C, Chtcherbinine M, Huddle BC, Wilson MW, Emmel T, Hohlman RM, McGonigal S, Buckanovich RJ, Larsen SD, Hurley TD. Development of substituted benzimidazoles as inhibitors of human aldehyde dehydrogenase 1A isoenzymes. Chem Biol Interact 2024; 391:110910. [PMID: 38364885 PMCID: PMC11062403 DOI: 10.1016/j.cbi.2024.110910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
Aldehyde dehydrogenase 1A (ALDH1A) isoforms may be a useful target for overcoming chemotherapy resistance in high-grade serous ovarian cancer (HGSOC) and other solid tumor cancers. However, as different cancers express different ALDH1A isoforms, isoform selective inhibitors may have a limited therapeutic scope. Furthermore, resistance to an ALDH1A isoform selective inhibitor could arise via induction of expression of other ALDH1A isoforms. As such, we have focused on the development of pan-ALDH1A inhibitors, rather than on ALDH1A isoform selective compounds. Herein, we report the development of a new group of pan-ALDH1A inhibitors to assess whether broad spectrum ALDH1A inhibition is an effective adjunct to chemotherapy in HGSOC. Optimization of the CM10 scaffold, aided by ALDH1A1 crystal structures, led to improved biochemical potencies, improved cellular efficacy as demonstrated by reduction in ALDEFLUOR signal in HGSOC cells, and substantial improvements in liver microsomal stability. Based on this work we identified two compounds 17 and 25 suitable for future in vivo proof of concept experiments.
Collapse
Affiliation(s)
- Cyrus Takahashi
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brandt C Huddle
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael W Wilson
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Timothy Emmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert M Hohlman
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stacy McGonigal
- Department of Obstetrics, Gynecology, and Reproductive Sciences, the Magee-Women's Research Institute, Pittsburgh, PA 15213, USA
| | - Ronald J Buckanovich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, the Magee-Women's Research Institute, Pittsburgh, PA 15213, USA; Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Women's Research Institute, Pittsburgh, PA, 15213, USA
| | - Scott D Larsen
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
4
|
Xanthis V, Mantso T, Dimtsi A, Pappa A, Fadouloglou VE. Human Aldehyde Dehydrogenases: A Superfamily of Similar Yet Different Proteins Highly Related to Cancer. Cancers (Basel) 2023; 15:4419. [PMID: 37686694 PMCID: PMC10650815 DOI: 10.3390/cancers15174419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The superfamily of human aldehyde dehydrogenases (hALDHs) consists of 19 isoenzymes which are critical for several physiological and biosynthetic processes and play a major role in the organism's detoxification via the NAD(P) dependent oxidation of numerous endogenous and exogenous aldehyde substrates to their corresponding carboxylic acids. Over the last decades, ALDHs have been the subject of several studies as it was revealed that their differential expression patterns in various cancer types are associated either with carcinogenesis or promotion of cell survival. Here, we attempt to provide a thorough review of hALDHs' diverse functions and 3D structures with particular emphasis on their role in cancer pathology and resistance to chemotherapy. We are especially interested in findings regarding the association of structural features and their changes with effects on enzymes' functionalities. Moreover, we provide an updated outline of the hALDHs inhibitors utilized in experimental or clinical settings for cancer therapy. Overall, this review aims to provide a better understanding of the impact of ALDHs in cancer pathology and therapy from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | - Vasiliki E. Fadouloglou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
5
|
Chen TM, Huang CM, Setiawan SA, Hsieh MS, Sheen CC, Yeh CT. KDM5D Histone Demethylase Identifies Platinum-Tolerant Head and Neck Cancer Cells Vulnerable to Mitotic Catastrophe. Int J Mol Sci 2023; 24:ijms24065310. [PMID: 36982384 PMCID: PMC10049674 DOI: 10.3390/ijms24065310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a major contributor to cancer incidence globally and is currently managed by surgical resection followed by adjuvant chemoradiotherapy. However, local recurrence is the major cause of mortality, indicating the emergence of drug-tolerant persister cells. A specific histone demethylase, namely lysine-specific demethylase 5D (KDM5D), is overexpressed in diverse types of cancers and involved in cancer cell cycle regulation. However, the role of KDM5D in the development of cisplatin-tolerant persister cells remains unexplored. Here, we demonstrated that KDM5D contributes to the development of persister cells. Aurora Kinase B (AURKB) disruption affected the vulnerability of persister cells in a mitotic catastrophe–dependent manner. Comprehensive in silico, in vitro, and in vivo experiments were performed. KDM5D expression was upregulated in HNSCC tumor cells, cancer stem cells, and cisplatin-resistant cells with biologically distinct signaling alterations. In an HNSCC cohort, high KDM5D expression was associated with a poor response to platinum treatment and early disease recurrence. KDM5D knockdown reduced the tolerance of persister cells to platinum agents and caused marked cell cycle deregulation, including the loss of DNA damage prevention, and abnormal mitosis-enhanced cell cycle arrest. By modulating mRNA levels of AURKB, KDM5D promoted the generation of platinum-tolerant persister cells in vitro, leading to the identification of the KDM5D/AURKB axis, which regulates cancer stemness and drug tolerance of HNSCC. Treatment with an AURKB inhibitor, namely barasertib, resulted in a lethal consequence of mitotic catastrophe in HNSCC persister cells. The cotreatment of cisplatin and barasertib suppressed tumor growth in the tumor mouse model. Thus, KDM5D might be involved in the development of persister cells, and AURKB disruption can overcome tolerance to platinum treatment in HNSCC.
Collapse
Affiliation(s)
- Tsung-Ming Chen
- Department of Otolaryngology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan;
- Department of Otolaryngology-Head and Neck Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Chih-Ming Huang
- Department of Otolaryngology, Taitung Mackay Memorial Hospital, Taitung City 950408, Taiwan;
- Department of Nursing, Tajen University, Pingtung 90741, Taiwan
| | - Syahru Agung Setiawan
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan;
- Department of Medical Research & Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Ming-Shou Hsieh
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan;
- Department of Dentistry, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 235, Taiwan
- Department of Periodontics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Chih-Chi Sheen
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan;
- Department of Dentistry, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 235, Taiwan
- Department of Periodontics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Correspondence: (C.-C.S.); (C.-T.Y.); Tel.: +886-2-249-0088 (ext. 8885) (C.-C.S.); +886-2-249-0088 (ext. 8881) (C.-T.Y.); Fax: +886-2-2248-0900 (C.-C.S.); +886-2-2248-0900 (C.-T.Y.)
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung 95092, Taiwan
- Correspondence: (C.-C.S.); (C.-T.Y.); Tel.: +886-2-249-0088 (ext. 8885) (C.-C.S.); +886-2-249-0088 (ext. 8881) (C.-T.Y.); Fax: +886-2-2248-0900 (C.-C.S.); +886-2-2248-0900 (C.-T.Y.)
| |
Collapse
|
6
|
Li J, Li X, Guo Q. Drug Resistance in Cancers: A Free Pass for Bullying. Cells 2022; 11:3383. [PMID: 36359776 PMCID: PMC9654341 DOI: 10.3390/cells11213383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
The cancer burden continues to grow globally, and drug resistance remains a substantial challenge in cancer therapy. It is well established that cancerous cells with clonal dysplasia generate the same carcinogenic lesions. Tumor cells pass on genetic templates to subsequent generations in evolutionary terms and exhibit drug resistance simply by accumulating genetic alterations. However, recent evidence has implied that tumor cells accumulate genetic alterations by progressively adapting. As a result, intratumor heterogeneity (ITH) is generated due to genetically distinct subclonal populations of cells coexisting. The genetic adaptive mechanisms of action of ITH include activating "cellular plasticity", through which tumor cells create a tumor-supportive microenvironment in which they can proliferate and cause increased damage. These highly plastic cells are located in the tumor microenvironment (TME) and undergo extreme changes to resist therapeutic drugs. Accordingly, the underlying mechanisms involved in drug resistance have been re-evaluated. Herein, we will reveal new themes emerging from initial studies of drug resistance and outline the findings regarding drug resistance from the perspective of the TME; the themes include exosomes, metabolic reprogramming, protein glycosylation and autophagy, and the relates studies aim to provide new targets and strategies for reversing drug resistance in cancers.
Collapse
Affiliation(s)
| | | | - Qie Guo
- The Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
7
|
Substance GP-2250 as a New Therapeutic Agent for Malignant Peritoneal Mesothelioma—A 3-D In Vitro Study. Int J Mol Sci 2022; 23:ijms23137293. [PMID: 35806313 PMCID: PMC9267014 DOI: 10.3390/ijms23137293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Malignant peritoneal mesothelioma is a rare tumor entity. Although cytoreductive surgery and hyperthermic intraperitoneal chemotherapy have increased overall survival, its prognosis remains poor. Established chemotherapeutics include mitomycin C (MMC) and cisplatin (CP), both characterized by severe side effects. GP-2250 is a novel antineoplastic agent, currently under clinical investigation. This in vitro study aims to investigate effects of GP-2250 including combinations with CP and MMC on malignant mesothelioma. JL-1 and MSTO-211H mesothelioma cell lines were treated with increasing doses of GP-2250, CP, MMC and combination therapies of GP-2250 + CP/MMC. Microscopic effects were documented, and a flow-cytometric apoptosis/necrosis assay was performed. Synergistic and antagonistic effects were analyzed by computing the combination index by Chou-Talalay. GP-2250 showed an antiadhesive effect on JL-1 and MSTO-211H spheroids. It had a dose-dependent cytotoxic effect on both monolayer and spheroid cultured cells, inducing apoptosis and necrosis. Combination treatments of GP-2250 with MMC and CP led to significant reductions of the effective doses of CP/MMC. Synergistic and additive effects were observed. GP-2250 showed promising antineoplastic effects on malignant mesothelioma cells in vitro especially in combination with CP/MMC. This forms the basis for further in vivo and clinical investigations in order to broaden treatment options.
Collapse
|
8
|
Anobile DP, Montenovo G, Pecoraro C, Franczak M, Ait Iddouch W, Peters GJ, Riganti C, Giovannetti E. Splicing deregulation, microRNA and Notch aberrations: fighting the three-headed dog to overcome drug resistance in malignant mesothelioma. Expert Rev Clin Pharmacol 2022; 15:305-322. [PMID: 35533249 DOI: 10.1080/17512433.2022.2074835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Malignant mesothelioma (MMe) is an aggressive rare cancer of the mesothelium, associated with asbestos exposure. MMe is currently an incurable disease at all stages mainly due to resistance to treatments. It is therefore necessary to elucidate key mechanisms underlying chemoresistance, in an effort to exploit them as novel therapeutic targets. AREAS COVERED Chemoresistance is frequently elicited by microRNA (miRNA) alterations and splicing deregulations. Indeed, several miRNAs, such as miR-29c, have been shown to exert oncogenic or oncosuppressive activity. Alterations in the splicing machinery might also be involved in chemoresistance. Moreover, the Notch signaling pathway, often deregulated in MMe, plays a key role in cancer stem cells formation and self-renewal, leading to drug resistance and relapses. EXPERT OPINION The prognosis of MMe in patients varies among different tumors and patient characteristics, and novel biomarkers and therapies are warranted. This work aims at giving an overview of MMe, with a special focus on state-of-the-art treatments and new therapeutic strategies against vulnerabilities emerging from studies on epigenetics factors. Besides, this review is also the first to discuss the interplay between miRNAs and alternative splicing as well as the role of Notch as new promising frontiers to overcome drug resistance in MMe.
Collapse
Affiliation(s)
- Dario P Anobile
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, Netherlands.,Department of Oncology, University of Torino, 10043 Orbassano, Italy
| | - Giulia Montenovo
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, Netherlands.,Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Camilla Pecoraro
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, Netherlands.,Dipartimento Di Scienze E Tecnologie Biologiche Chimiche E Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Marika Franczak
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, Netherlands.,Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Widad Ait Iddouch
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, Netherlands
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, Netherlands.,Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10043 Orbassano, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081HV Amsterdam, Netherlands.,Fondazione Pisana per la Scienza Pisa, 56100 Pisa, Italy
| |
Collapse
|
9
|
Bronte G, Procopio AD, Graciotti L. The application of cancer stem cell model in malignant mesothelioma. Crit Rev Oncol Hematol 2022; 174:103698. [PMID: 35525390 DOI: 10.1016/j.critrevonc.2022.103698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 12/22/2022] Open
Abstract
The high mortality rate of malignant pleural mesothelioma led to study the mechanisms for chemoresistance. The cancer stem cell (CSC) model has been proposed to explain chemoresistance. CSCs are characterized by self-renewal capacity, that is detected through tumor-initiating cell assays. As in other malignancies, many studies sought to identify surface markers to isolate CSCs from malignant mesothelioma. Other studies characterized malignant mesothelioma CSCs for the expression of specific genes involved in stemness and the expression of proteins involved in chemoresistance. However, the main methods to characterize isolated CSCs include sphere formation, invasiveness, tumor-initiating capacity and expression of specific surface markers. The better knowledge of malignant mesothelioma CSCs allowed exploring new potential targets to develop specific treatments.
Collapse
Affiliation(s)
- Giuseppe Bronte
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy.
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, National Institute of Health and Sciences on Ageing (INRCA-IRCCS), Ancona, Italy
| | - Laura Graciotti
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
10
|
Forchlorfenuron and Novel Analogs Cause Cytotoxic Effects in Untreated and Cisplatin-Resistant Malignant Mesothelioma-Derived Cells. Int J Mol Sci 2022; 23:ijms23073963. [PMID: 35409322 PMCID: PMC8999537 DOI: 10.3390/ijms23073963] [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: 02/22/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Malignant mesothelioma (MM) is a currently incurable, aggressive cancer derived from mesothelial cells, most often resulting from asbestos exposure. The current first-line treatment in unresectable MM is cisplatin/pemetrexed, which shows very little long-term effectiveness, necessitating research for novel therapeutic interventions. The existing chemotherapies often act on the cytoskeleton, including actin filaments and microtubules, but recent advances indicate the ‘fourth’ form consisting of the family of septins, representing a novel target. The septin inhibitor forchlorfenuron (FCF) and FCF analogs inhibit MM cell growth in vitro, but at concentrations which are too high for clinical applications. Based on the reported requirement of the chloride group in the 2-position of the pyridine ring of FCF for MM cell growth inhibition and cytotoxicity, we systematically investigated the importance (cell growth-inhibiting capacity) of the halogen atoms fluorine, chlorine, bromine and iodine in the 2- or 3-position of the pyridine ring. The MM cell lines ZL55, MSTO-211H, and SPC212, and—as a control—immortalized Met-5A mesothelial cells were used. The potency of the various halogen substitutions in FCF was mostly correlated with the atom size (covalent radius); the small fluoride analogs showed the least effect, while the largest one (iodide) most strongly decreased the MTT signals, in particular in MM cells derived from epithelioid MM. In the latter, the strongest effects in vitro were exerted by the 2-iodo and, unexpectedly, the 2-trifluoromethyl (2-CF3) FCF analogs, which were further tested in vivo in mice. However, FCF-2-I and, more strongly, FCF-2-CF3 caused rapidly occurring strong symptoms of systemic toxicity at doses lower than those previously obtained with FCF. Thus, we investigated the effectiveness of FCF (and selected analogs) in vitro in MM cells which were first exposed to cisplatin. The slowly appearing population of cisplatin-resistant cells was still susceptible to the growth-inhibiting/cytotoxic effect of FCF and its analogs, indicating that cisplatin and FCF target non-converging pathways in MM cells. Thus, a combination therapy of cisplatin and FCF (analogs) might represent a new avenue for the treatment of repopulating chemo-resistant MM cells in this currently untreatable cancer.
Collapse
|
11
|
Zanoni M, Bravaccini S, Fabbri F, Arienti C. Emerging Roles of Aldehyde Dehydrogenase Isoforms in Anti-cancer Therapy Resistance. Front Med (Lausanne) 2022; 9:795762. [PMID: 35299840 PMCID: PMC8920988 DOI: 10.3389/fmed.2022.795762] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/10/2022] [Indexed: 12/19/2022] Open
Abstract
Aldehyde dehydrogenases (ALDHs) are a family of detoxifying enzymes often upregulated in cancer cells and associated with therapeutic resistance. In humans, the ALDH family comprises 19 isoenzymes active in the majority of mammalian tissues. Each ALDH isoform has a specific differential expression pattern and most of them have individual functional roles in cancer. ALDHs are overexpressed in subpopulations of cancer cells with stem-like features, where they are involved in several processes including cellular proliferation, differentiation, detoxification and survival, participating in lipids and amino acid metabolism and retinoic acid synthesis. In particular, ALDH enzymes protect cancer cells by metabolizing toxic aldehydes in less reactive and more soluble carboxylic acids. High metabolic activity as well as conventional anticancer therapies contribute to aldehyde accumulation, leading to DNA double strand breaks (DSB) through the generation of reactive oxygen species (ROS) and lipid peroxidation. ALDH overexpression is crucial not only for the survival of cancer stem cells but can also affect immune cells of the tumour microenvironment (TME). The reduction of ROS amount and the increase in retinoic acid signaling impairs immunogenic cell death (ICD) inducing the activation and stability of immunosuppressive regulatory T cells (Tregs). Dissecting the role of ALDH specific isoforms in the TME can open new scenarios in the cancer treatment. In this review, we summarize the current knowledge about the role of ALDH isoforms in solid tumors, in particular in association with therapy-resistance.
Collapse
Affiliation(s)
- Michele Zanoni
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | | | | | - Chiara Arienti
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
12
|
Optimization of tumor spheroid model in mesothelioma and lung cancers and anti-cancer drug testing in H2052/484 spheroids. Oncotarget 2021; 12:2375-2387. [PMID: 34853659 PMCID: PMC8629400 DOI: 10.18632/oncotarget.28134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/10/2021] [Indexed: 01/08/2023] Open
Abstract
Advanced lung cancers and mesothelioma remain incurable diseases. Despite some promising new therapy strategies, predicting whether an individual patient will be sensitive to a given therapy is challenging. The purpose of this study is to establish and evaluate the efficiency of a three-dimensional spheroid model of human thoracic cancer in predicting the efficacy of drugs. Human mesothelioma and lung tumor spheroids were established from cell lines and primary cells derived from the patient. The growth kinetics and cell viability of microtumors were assessed using spheroid size and intracellular ATP level. The sensitivity of the mesothelioma spheroids to the cisplatin or cisplatin/pemetrexed combination was determined. We determined that studying the kinetics of the spheroid growth for 15 days after seeding 1000 cells/well in a 96-well plate was optimal. Monitoring the growth kinetic and intracellular ATP of spheroids allowed the identification of early changes in spheroid viability. Finally, we validated this model by measuring a dose-dependent reduction in the cell viability of mesothelioma H2052/484 spheroids treated with both first-line treatments, cisplatin and the cisplatin/pemetrexed combination. In conclusion, we have developed a three-dimensional spheroid model of thoracic tumor cells useful for tailoring the medical treatment to the specific characteristics of each patient.
Collapse
|
13
|
Cersosimo F, Barbarino M, Lonardi S, Vermi W, Giordano A, Bellan C, Giurisato E. Mesothelioma Malignancy and the Microenvironment: Molecular Mechanisms. Cancers (Basel) 2021; 13:cancers13225664. [PMID: 34830817 PMCID: PMC8616064 DOI: 10.3390/cancers13225664] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
Several studies have reported that cellular and soluble components of the tumor microenvironment (TME) play a key role in cancer-initiation and progression. Considering the relevance and the complexity of TME in cancer biology, recent research has focused on the investigation of the TME content, in terms of players and informational exchange. Understanding the crosstalk between tumor and non-tumor cells is crucial to design more beneficial anti-cancer therapeutic strategies. Malignant pleural mesothelioma (MPM) is a complex and heterogenous tumor mainly caused by asbestos exposure with few treatment options and low life expectancy after standard therapy. MPM leukocyte infiltration is rich in macrophages. Given the failure of macrophages to eliminate asbestos fibers, these immune cells accumulate in pleural cavity leading to the establishment of a unique inflammatory environment and to the malignant transformation of mesothelial cells. In this inflammatory landscape, stromal and immune cells play a driven role to support tumor development and progression via a bidirectional communication with tumor cells. Characterization of the MPM microenvironment (MPM-ME) may be useful to understand the complexity of mesothelioma biology, such as to identify new molecular druggable targets, with the aim to improve the outcome of the disease. In this review, we summarize the known evidence about the MPM-ME network, including its prognostic and therapeutic relevance.
Collapse
Affiliation(s)
- Francesca Cersosimo
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| | - Marcella Barbarino
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.); (C.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy; (S.L.); (W.V.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy; (S.L.); (W.V.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.); (C.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Cristiana Bellan
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.); (C.B.)
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
- Correspondence: ; Tel.: +39-057-723-2125
| |
Collapse
|
14
|
Insights into Intra-Tumoral Heterogeneity: Transcriptional Profiling of Chemoresistant MPM Cell Subpopulations Reveals Involvement of NFkB and DNA Repair Pathways and Contributes a Prognostic Signature. Int J Mol Sci 2021; 22:ijms222112071. [PMID: 34769499 PMCID: PMC8585077 DOI: 10.3390/ijms222112071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Chemoresistance is a hallmark of malignant pleural mesothelioma (MPM) management and the expression of ALDH1A3 is responsible for the survival and activity of MPM chemoresistant cell subpopulations (ALDHbright cells). We enriched mesothelioma ALDHbright cells to near homogeneity by FACS sorting and an Aldefluor assay and performed unbiased Affymetrix gene expression profiling. Viability and ELISA assays were used to rule out significant apoptosis in the sorted cell subpopulations and to assess target engagement by butein. Statistical analysis of the results, pathway enrichment and promoter enrichment were employed for the generation of the data. Q-RTPCR was used to validate a subset of the identified, modulated mRNAs In this work, we started from the observation that the mRNA levels of the ALDH1A3 isoform could prognostically stratify MPM patients. Thus, we purified MPM ALDHbright cells from NCI-H2595 cells and interrogated their gene expression (GES) profile. We analyzed the GES of the purified cells at both a steady state and upon treatment with butein (a multifunctional tetrahydroxy-chalcone), which abates the ALDHbright cell number, thereby exerting chemo-sensitizing effects in vitro and in vivo. We identified 924 genes modulated in a statistically significant manner as a function of ALDH status and of the response to the inhibitor. Pathway and promoter enrichment identified the molecular determinant of high ALDH status and how butein treatment altered the molecular portrait of those chemoresistant cell subpopulations. Further, we unraveled an eighteen-gene signature with high prognostic significance for MPM patients, and showed that most of the identified prognostic contributors escaped the analysis of unfractionated samples. This work proves that digging into the unexplored field of intra-tumor heterogeneity (ITH) by working at the cell subpopulation level may provide findings of prognostic relevance, in addition to mechanistic insights into tumor resistance to therapy.
Collapse
|
15
|
Cioce M, Canino C, Pass H, Blandino G, Strano S, Fazio VM. Arachidonic acid drives adaptive responses to chemotherapy-induced stress in malignant mesothelioma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:344. [PMID: 34727953 PMCID: PMC8561918 DOI: 10.1186/s13046-021-02118-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Background High resistance to therapy and poor prognosis characterizes malignant pleural mesothelioma (MPM). In fact, the current lines of treatment, based on platinum and pemetrexed, have limited impact on the survival of MPM patients. Adaptive response to therapy-induced stress involves complex rearrangements of the MPM secretome, mediated by the acquisition of a senescence-associated-secretory-phenotype (SASP). This fuels the emergence of chemoresistant cell subpopulations, with specific gene expression traits and protumorigenic features. The SASP-driven rearrangement of MPM secretome takes days to weeks to occur. Thus, we have searched for early mediators of such adaptive process and focused on metabolites differentially released in mesothelioma vs mesothelial cell culture media, after treatment with pemetrexed. METHODS Mass spectrometry-based (LC/MS and GC/MS) identification of extracellular metabolites and unbiased statistical analysis were performed on the spent media of mesothelial and mesothelioma cell lines, at steady state and after a pulse with pharmacologically relevant doses of the drug. ELISA based evaluation of arachidonic acid (AA) levels and enzyme inhibition assays were used to explore the role of cPLA2 in AA release and that of LOX/COX-mediated processing of AA. QRT-PCR, flow cytometry analysis of ALDH expressing cells and 3D spheroid growth assays were employed to assess the role of AA at mediating chemoresistance features of MPM. ELISA based detection of p65 and IkBalpha were used to interrogate the NFkB pathway activation in AA-treated cells. RESULTS We first validated what is known or expected from the mechanism of action of the antifolate. Further, we found increased levels of PUFAs and, more specifically, arachidonic acid (AA), in the transformed cell lines treated with pemetrexed. We showed that pharmacologically relevant doses of AA tightly recapitulated the rearrangement of cell subpopulations and the gene expression changes happening in pemetrexed -treated cultures and related to chemoresistance. Further, we showed that release of AA following pemetrexed treatment was due to cPLA2 and that AA signaling impinged on NFkB activation and largely affected anchorage-independent, 3D growth and the resistance of the MPM 3D cultures to the drug. CONCLUSIONS AA is an early mediator of the adaptive response to pem in chemoresistant MPM and, possibly, other malignancies.
Collapse
Affiliation(s)
- Mario Cioce
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy.
| | - Claudia Canino
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA.,Radiation Oncology Unit, UPMC Hillmann Cancer Center, San Pietro Hospital FBF, Rome, Italy
| | - Harvey Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Sabrina Strano
- SAFU Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Vito Michele Fazio
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy. .,Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133, Rome, Italy. .,Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy.
| |
Collapse
|
16
|
MacDonagh L, Santiago RM, Gray SG, Breen E, Cuffe S, Finn SP, O'Byrne KJ, Barr MP. Exploitation of the vitamin A/retinoic acid axis depletes ALDH1-positive cancer stem cells and re-sensitises resistant non-small cell lung cancer cells to cisplatin. Transl Oncol 2021; 14:101025. [PMID: 33550205 PMCID: PMC7868629 DOI: 10.1016/j.tranon.2021.101025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 12/23/2022] Open
Abstract
Despite advances in personalised medicine and the emerging role of immune checkpoints in directing treatment decisions in subsets of lung cancer patients, non-small cell lung cancer (NSCLC) remains the most common cause of cancer-related deaths worldwide. The development of drug resistance plays a key role in the relapse of lung cancer patients in the clinical setting, mainly due to the unlimited renewal capacity of residual cancer stem cells (CSCs) within the tumour cell population during chemotherapy. In this study, we investigated the function of the CSC marker, aldehyde dehydrogenase (ALDH1) in retinoic acid cell signalling using an in vitro model of cisplatin resistant NSCLC. The addition of key components in retinoic acid cell signalling, all-trans retinoic acid (ATRA) and retinol to cisplatin chemotherapy, significantly reduced ALDH1-positive cell subsets in cisplatin resistant NSCLC cells relative to their sensitive counterparts resulting in the re-sensitisation of chemo-resistant cells to the cytotoxic effects of cisplatin. Furthermore, combination of ATRA or retinol with cisplatin significantly inhibited cell proliferation, colony formation and increased cisplatin-induced apoptosis. This increase in apoptosis may, at least in part, be due to differential gene expression of the retinoic acid (RARα/β) and retinoid X (RXRα) nuclear receptors in cisplatin-resistant lung cancer cells. These data support the concept of exploiting the retinoic acid signalling cascade as a novel strategy in targeting subsets of CSCs in cisplatin resistant lung tumours.
Collapse
Affiliation(s)
- Lauren MacDonagh
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Ireland.
| | - Rhyla Mae Santiago
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Ireland.
| | - Steven G Gray
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Ireland.
| | - Eamon Breen
- Flow Cytometry Facility, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Ireland.
| | - Sinead Cuffe
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Ireland; Medical Oncology Department, St James's Hospital, Dublin, Ireland.
| | - Stephen P Finn
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Ireland; Histopathology Department, St James's Hospital & Trinity College Dublin, Ireland.
| | - Kenneth J O'Byrne
- Cancer & Ageing Research Program, Queensland University of Technology, Brisbane, Australia.
| | - Martin P Barr
- Thoracic Oncology Research Group, School of Clinical Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital & Trinity College Dublin, Ireland.
| |
Collapse
|
17
|
Huddle BC, Grimley E, Chtcherbinine M, Buchman CD, Takahashi C, Debnath B, McGonigal SC, Mao S, Li S, Felton J, Pan S, Wen B, Sun D, Neamati N, Buckanovich RJ, Hurley TD, Larsen SD. Development of 2,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one inhibitors of aldehyde dehydrogenase 1A (ALDH1A) as potential adjuncts to ovarian cancer chemotherapy. Eur J Med Chem 2020; 211:113060. [PMID: 33341649 DOI: 10.1016/j.ejmech.2020.113060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
There is strong evidence that inhibition of one or more Aldehyde Dehydrogenase 1A (ALDH1A) isoforms may be beneficial in chemotherapy-resistant ovarian cancer and other tumor types. While many previous efforts have focused on development of ALDH1A1 selective inhibitors, the most deadly ovarian cancer subtype, high-grade serous (HGSOC), exhibits elevated expression of ALDH1A3. Herein, we report continued development of pan-ALDH1A inhibitors to assess whether broad spectrum ALDH1A inhibition is an effective adjunct to chemotherapy in this critical tumor subtype. Optimization of the CM39 scaffold, aided by metabolite ID and several new ALDH1A1 crystal structures, led to improved biochemical potencies, improved cellular ALDH inhibition in HGSOC cell lines, and substantial improvements in microsomal stability culminating in orally bioavailable compounds. We demonstrate that two compounds 68 and 69 are able to synergize with chemotherapy in a resistant cell line and patient-derived HGSOC tumor spheroids, indicating their suitability for future in vivo proof of concept experiments.
Collapse
Affiliation(s)
- Brandt C Huddle
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Edward Grimley
- Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cameron D Buchman
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cyrus Takahashi
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bikash Debnath
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stacy C McGonigal
- Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Shuai Mao
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Siwei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jeremy Felton
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shu Pan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ronald J Buckanovich
- Division of Hematology-Oncology, Departments of Internal Medicine and Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh Medical Center and the Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Scott D Larsen
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
18
|
Zhang H, Xu H, Ashby CR, Assaraf YG, Chen ZS, Liu HM. Chemical molecular-based approach to overcome multidrug resistance in cancer by targeting P-glycoprotein (P-gp). Med Res Rev 2020; 41:525-555. [PMID: 33047304 DOI: 10.1002/med.21739] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/01/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) remains one of the major impediments for efficacious cancer chemotherapy. Increased efflux of multiple chemotherapeutic drugs by transmembrane ATP-binding cassette (ABC) transporter superfamily is considered one of the primary causes for cancer MDR, in which the role of P-glycoprotein (P-gp/ABCB1) has been most well-established. The clinical co-administration of P-gp drug efflux inhibitors, in combination with anticancer drugs which are P-gp transport substrates, was considered to be a treatment modality to surmount MDR in anticancer therapy by blocking P-gp-mediated multidrug efflux. Extensive attempts have been carried out to screen for sets of nontoxic, selective, and efficacious P-gp efflux inhibitors. In this review, we highlight the recent achievements in drug design, characterization, structure-activity relationship (SAR) studies, and mechanisms of action of the newly synthetic, potent small molecules P-gp inhibitors in the past 5 years. The development of P-gp inhibitors will increase our knowledge of the mechanisms and functions of P-gp-mediated drug efflux which will benefit drug discovery and clinical cancer therapeutics where P-gp transporter overexpression has been implicated in MDR.
Collapse
Affiliation(s)
- Hang Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Co-innovation Center of Henan Province for New Drug R & D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiwei Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Co-innovation Center of Henan Province for New Drug R & D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, New York, USA
| | - Yehuda G Assaraf
- Department of Biology, The Fred Wyszkowski Cancer Research Laboratory, Technion-Israel Institute of Technology, Haifa, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, Queens, New York, USA
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Co-innovation Center of Henan Province for New Drug R & D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Xu D, Yang H, Schmid RA, Peng RW. Therapeutic Landscape of Malignant Pleural Mesothelioma: Collateral Vulnerabilities and Evolutionary Dependencies in the Spotlight. Front Oncol 2020; 10:579464. [PMID: 33072611 PMCID: PMC7538645 DOI: 10.3389/fonc.2020.579464] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is the epitome of a recalcitrant cancer driven by pharmacologically intractable tumor suppressor proteins. A significant but largely unmet challenge in the field is the translation of genetic information on alterations in tumor suppressor genes (TSGs) into effective cancer-specific therapies. The notion that abnormal tumor genome subverts physiological cellular processes, which creates collateral vulnerabilities contextually related to specific genetic alterations, offers a promising strategy to target TSG-driven MPM. Moreover, emerging evidence has increasingly appreciated the therapeutic potential of genetic and pharmacological dependencies acquired en route to cancer development and drug resistance. Here, we review the most recent progress on vulnerabilities co-selected by functional loss of major TSGs and dependencies evolving out of cancer development and resistance to cisplatin based chemotherapy, the only first-line regimen approved by the US Food and Drug Administration (FDA). Finally, we highlight CRISPR-based functional genomics that has emerged as a powerful platform for cancer drug discovery in MPM. The repertoire of MPM-specific “Achilles heel” rises on the horizon, which holds the promise to elucidate therapeutic landscape and may promote precision oncology for MPM.
Collapse
Affiliation(s)
- Duo Xu
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Haitang Yang
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralph A Schmid
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ren-Wang Peng
- Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Schmidtova S, Dorssers LCJ, Kalavska K, Gillis AJM, Oosterhuis JW, Stoop H, Miklikova S, Kozovska Z, Burikova M, Gercakova K, Durinikova E, Chovanec M, Mego M, Kucerova L, Looijenga LHJ. Napabucasin overcomes cisplatin resistance in ovarian germ cell tumor-derived cell line by inhibiting cancer stemness. Cancer Cell Int 2020; 20:364. [PMID: 32774158 PMCID: PMC7397611 DOI: 10.1186/s12935-020-01458-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/25/2020] [Indexed: 02/08/2023] Open
Abstract
Background Cisplatin resistance of ovarian yolk sac tumors (oYST) is a clinical challenge due to dismal patient prognosis, even though the disease is extremely rare. We investigated potential association between cisplatin resistance and cancer stem cell (CSC) markers in chemoresistant oYST cells and targeting strategies to overcome resistance in oYST. Methods Chemoresistant cells were derived from chemosensitive human oYST cells by cultivation in cisplatin in vitro. Derivative cells were characterized by chemoresistance, functional assays, flow cytometry, gene expression and protein arrays focused on CSC markers. RNAseq, methylation and microRNA profiling were performed. Quail chorioallantoic membranes (CAM) with implanted oYST cells were used to analyze the micro-tumor extent and interconnection with the CAM. Tumorigenicity in vivo was determined on immunodeficient mouse model. Chemoresistant cells were treated by inhibitors intefering with the CSC properties to examine the chemosensitization to cisplatin. Results Long-term cisplatin exposure resulted in seven-fold higher IC50 value in resistant cells, cross-resistance to oxaliplatin and carboplatin, and increased migratory capacity, invasiveness and tumorigenicity, associated with hypomethylation of differentially methylated genes/promotors. Resistant cells exhibited increased expression of prominin-1 (CD133), ATP binding cassette subfamily G member 2 (ABCG2), aldehyde dehydrogenase 3 isoform A1 (ALDH3A1), correlating with reduced gene and promoter methylation, as well as increased expression of ALDH1A3 and higher overall ALDH enzymatic activity, rendering them cross-resistant to DEAB, disulfiram and napabucasin. Salinomycin and tunicamycin were significantly more toxic to resistant cells. Pretreatment with napabucasin resensitized the cells to cisplatin and reduced their tumorigenicity in vivo. Conclusions The novel chemoresistant cells represent unique model of refractory oYST. CSC markers are associated with cisplatin resistance being possible targets in chemorefractory oYST.
Collapse
Affiliation(s)
- Silvia Schmidtova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.,Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
| | - Lambert C J Dorssers
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Katarina Kalavska
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.,Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Ad J M Gillis
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - J Wolter Oosterhuis
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Hans Stoop
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Svetlana Miklikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Kozovska
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Monika Burikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Katarina Gercakova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Erika Durinikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Michal Chovanec
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Michal Mego
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
| | - Lucia Kucerova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Leendert H J Looijenga
- Department of Pathology, Laboratory for Experimental Patho-Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
21
|
Ling M, Yang X. Correlation between cancer stem cells (CSCs) and tumor-infiltrating lymphocytes (TILs): do TILs interact with CSCs in non-small cell lung cancer? ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:914. [PMID: 32953714 PMCID: PMC7475387 DOI: 10.21037/atm-20-1556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Min Ling
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
22
|
Horio D, Minami T, Kitai H, Ishigaki H, Higashiguchi Y, Kondo N, Hirota S, Kitajima K, Nakajima Y, Koda Y, Fujimoto E, Negi Y, Niki M, Kanemura S, Shibata E, Mikami K, Takahashi R, Yokoi T, Kuribayashi K, Kijima T. Tumor-associated macrophage-derived inflammatory cytokine enhances malignant potential of malignant pleural mesothelioma. Cancer Sci 2020; 111:2895-2906. [PMID: 32530527 PMCID: PMC7419052 DOI: 10.1111/cas.14523] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/24/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an asbestos-related aggressive malignant neoplasm. Due to the difficulty of achieving curative surgical resection in most patients with MPM, a combination chemotherapy of cisplatin and pemetrexed has been the only approved regimen proven to improve the prognosis of MPM. However, the median overall survival time is at most 12 mo even with this regimen. There has been therefore a pressing need to develop a novel chemotherapeutic strategy to bring about a better outcome for MPM. We found that expression of interleukin-1 receptor (IL-1R) was upregulated in MPM cells compared with normal mesothelial cells. We also investigated the biological significance of the interaction between pro-inflammatory cytokine IL-1β and the IL-1R in MPM cells. Stimulation by IL-1β promoted MPM cells to form spheroids along with upregulating a cancer stem cell marker CD26. We also identified tumor-associated macrophages (TAMs) as the major source of IL-1β in the MPM microenvironment. Both high mobility group box 1 derived from MPM cells and the asbestos-activated inflammasome in TAMs induced the production of IL-1β, which resulted in enhancement of the malignant potential of MPM. We further performed immunohistochemical analysis using clinical MPM samples obtained from patients who were treated with the combination of platinum plus pemetrexed, and found that the overexpression of IL-1R tended to correlate with poor overall survival. In conclusion, the interaction between MPM cells and TAMs through a IL-1β/IL-1R signal could be a promising candidate as the target for novel treatment of MPM (Hyogo College of Medicine clinical trial registration number: 2973).
Collapse
Affiliation(s)
- Daisuke Horio
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
| | - Toshiyuki Minami
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Hidemi Kitai
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Hirotoshi Ishigaki
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
| | - Yoko Higashiguchi
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
| | - Nobuyuki Kondo
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic SurgeryHyogo College of MedicineNishinomiyaJapan
| | - Seiichi Hirota
- Department of Surgical PathologyHyogo College of MedicineNishinomiyaJapan
| | - Kazuhiro Kitajima
- Division of Nuclear Medicine and PET CenterDepartment of RadiologyHyogo College of MedicineNishinomiyaJapan
| | - Yasuhiro Nakajima
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
| | - Yuichi Koda
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Eriko Fujimoto
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Yoshiki Negi
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Maiko Niki
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Shingo Kanemura
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Eisuke Shibata
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Koji Mikami
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Ryo Takahashi
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Takashi Yokoi
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Kozo Kuribayashi
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| | - Takashi Kijima
- Department of Respiratory Medicine and HematologyHyogo College of MedicineNishinomiyaJapan
- Department of Thoracic OncologyHyogo College of MedicineNishinomiyaJapan
| |
Collapse
|
23
|
Wong JJW, Selbo PK. High aldehyde dehydrogenase activity does not protect colon cancer cells against TPCS 2a-sensitized photokilling. Photochem Photobiol Sci 2020; 19:308-312. [PMID: 32108197 DOI: 10.1039/c9pp00453j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aldehyde dehydrogenases (ALDH) are detoxifying enzymes that are upregulated in cancer stem cells (CSCs) and may cause chemo- and ionizing radiation (IR) therapy resistance. By using the ALDEFLUOR assay, CD133 + human colon cancer cells HT-29, were FACSorted into three populations: ALDHbright, ALDHdim and unsorted (bulk) and treated with chemo-, radio- or photodynamic therapy (PDT) using the clinical relevant photosensitizer disulfonated tetraphenyl chlorin (TPCS2a/fimaporfin). Here we show that there is no difference in cytotoxic responses to TPCS2a-PDT in ALHDbright, ALDHdim or bulk cancer cells. Likewise, both 5-FU and oxaliplatin chemotherapy efficacy was not reduced in ALDHbright as compared to ALDHdim cancer cells. However, we found that ALHDbright HT-29 cells are significantly less sensitive to ionizing radiation compared to ALDHdim cells. This study demonstrates that the cytotoxic response to PDT (using TPCS2a as photosensitizer) is independent of ALDH activity in HT-29 cancer cells. Our results further strengthen the use of TPCS2a to target CSCs.
Collapse
Affiliation(s)
- Judith Jing Wen Wong
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital Oslo University Hospital Montebello, 0379, Oslo, Norway
| | - Pål Kristian Selbo
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital Oslo University Hospital Montebello, 0379, Oslo, Norway
| |
Collapse
|
24
|
Williams M, Cheng YY, Phimmachanh M, Winata P, van Zandwijk N, Reid G. Tumour suppressor microRNAs contribute to drug resistance in malignant pleural mesothelioma by targeting anti-apoptotic pathways. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:1193-1206. [PMID: 35582270 PMCID: PMC9019216 DOI: 10.20517/cdr.2019.41] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/17/2019] [Accepted: 09/28/2019] [Indexed: 11/28/2022]
Abstract
Aim: Aberrant microRNA expression is a common event in cancer drug resistance, however its involvement in malignant pleural mesothelioma (MPM) drug resistance is largely unexplored. We aimed to investigate the contribution of microRNAs to the resistance to drugs commonly used in the treatment of MPM. Methods: Drug resistant MPM cell lines were generated by treatment with cisplatin, gemcitabine or vinorelbine. Expression of microRNAs was quantified using RT-qPCR. Apoptosis and drug sensitivity assays were carried out following transfection with microRNA mimics or BCL2 siRNAs combined with drugs. Results: Expression of miR-15a, miR-16 and miR-34a was downregulated in MPM cells with acquired drug resistance. Transfection with miR-15a or miR-16 mimics reversed the resistance to cisplatin, gemcitabine or vinorelbine, whereas miR-34a reversed cisplatin and vinorelbine resistance only. Similarly, in parental cell lines, miR-15a or miR-16 mimics sensitised cells to all drugs, whereas miR-34a increased response to cisplatin and vinorelbine. Increased microRNA expression increased drug-induced apoptosis and caused BCL2 mRNA and protein reduction. RNAi-mediated knockdown of BCL2 partly recapitulated the increase in drug sensitivity in cisplatin and vinorelbine treated cells. Conclusion: Drug-resistant MPM cell lines exhibited reduced expression of tumour suppressor microRNAs. Increasing tumour suppressor of microRNA expression sensitised both drug resistant and parental cell lines to chemotherapeutic agents, in part through targeting of BCL2. Taken together, these data suggest that miR-15a, miR-16 and miR-34a are involved in the acquired and intrinsic drug resistance phenotype of MPM cells.
Collapse
Affiliation(s)
- Marissa Williams
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
| | - Yuen Yee Cheng
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
| | - Monica Phimmachanh
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
- Current address: Garvan Institute of Medical Research, Darlinghurst, Sydney NSW2010, Australia
| | - Patrick Winata
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
| | - Nico van Zandwijk
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
- Current address: Sydney Local Health District, Concord, Sydney NSW2194, Australia
| | - Glen Reid
- Asbestos Diseases Research Institute, Sydney NSW2139, Australia
- Sydney Medical School, The University of Sydney, Sydney NSW2050, Australia
- Current address: Department of Pathology, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
25
|
Masciale V, Grisendi G, Banchelli F, D'Amico R, Maiorana A, Sighinolfi P, Stefani A, Morandi U, Dominici M, Aramini B. Isolation and Identification of Cancer Stem-Like Cells in Adenocarcinoma and Squamous Cell Carcinoma of the Lung: A Pilot Study. Front Oncol 2019; 9:1394. [PMID: 31921651 PMCID: PMC6930193 DOI: 10.3389/fonc.2019.01394] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Lung cancer stem cells (CSCs) share many characteristics with normal stem cells, such as self-renewal and multipotentiality. High expression of aldehyde dehydrogenase (ALDH) has been detected in many tumors, particularly in the CSC compartment, and it plays an important role in tumor proliferation, metastasis, and drug resistance. CD44 is commonly used as a cell surface marker of cancer stem-like cells in epithelial tumors. The aim of this study was to isolate and analyze cancer stem-like cells from surgically removed specimens to compare lung adenocarcinoma (ADENO) and squamous (SQUAMO) cell carcinoma. Methods: The ALDEFLUOR assay was used to identify and sort ALDHhigh and ALDHlow human lung cancer cells following tissue digestion. Fluorescence-activated cell sorting analysis for CD44 was performed with tumor cells. Quantitative real-time PCR was performed to assess the expression of SOX2 and NANOG as stemness markers. ALDH1A1 expression was additionally determined by immunohistochemistry. Anchorage-independent ALDHhigh cell growth was also evaluated. ALDHhigh ADENO and SQUAMO cells were cultured to analyze spheroid formation. Results: All specimens contained 0.5-12.5% ALDHhigh cells with 3.8-18.9% CD44-positive cells. SOX2 and NANOG relative expression in ALDHhigh compared to ALDHlow cells in ADENO and SQUAMO was analyzed and compared between the histotypes. Immunohistochemistry confirmed the presence of ALDH1A1 in the sections. SOX2 and NANOG were expressed at higher levels in the ALDHhigh subpopulation than in the ALDHlow subpopulation only in ADENO cells, and the opposite result was seen in SQUAMO cells. In vitro functional assays demonstrated that ALDHhigh cells exhibited migration capacity with distinct behaviors between ALDHhigh spheres in ADENO vs. SQUAMO samples. Conclusions: Our results highlight the importance of a better characterization of cancer stem-like cells in ADENO and SQUAMO histotypes. This may suggest new differential approaches for prognostic and therapeutic purposes in patients with non-small-cell lung cancer.
Collapse
Affiliation(s)
- Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy.,Rigenerand SRL, Modena, Italy
| | - Federico Banchelli
- Department of Medical and Surgical Sciences for Children & Adults, Center of Medical Statistic, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D'Amico
- Department of Medical and Surgical Sciences for Children & Adults, Center of Medical Statistic, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Department of Medical and Surgical Sciences for Children & Adults, Institute of Pathology, University of Modena and Reggio Emilia, Modena, Italy
| | - Pamela Sighinolfi
- Department of Medical and Surgical Sciences for Children & Adults, Institute of Pathology, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Stefani
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
26
|
Dinavahi SS, Gowda R, Bazewicz CG, Battu MB, Lin JM, Chitren RJ, Pandey MK, Amin S, Robertson GP, Gowda K. Design, synthesis characterization and biological evaluation of novel multi-isoform ALDH inhibitors as potential anticancer agents. Eur J Med Chem 2019; 187:111962. [PMID: 31887569 DOI: 10.1016/j.ejmech.2019.111962] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 12/31/2022]
Abstract
The aldehyde dehydrogenases (ALDHs) are a family of detoxifying enzymes that are overexpressed in various cancers. Increased expression of ALDH is associated with poor prognosis, stemness, and drug resistance. Because of the critical role of ALDH in cancer stem cells, several ALDH inhibitors have been developed. Nonetheless, all these inhibitors either lack efficacy or are too toxic or have not been tested extensively. Thus, the continued development of ALDH inhibitors is warranted. In this study, we designed and synthesized potent multi-ALDH isoform inhibitors based on the isatin backbone. The early molecular docking studies and enzymatic tests revealed that 3(a-l) and 4(a-l) are the potent ALDH1A1, ALDHA2, and ALDH3A1 inhibitors. ALDH inhibitory IC50s of 3(a-l) and 4(a-l) were 230 nM to >10,000 nM for ALDH1A1, 939 nM to >10,000 nM for ALDH2 and 193 nM to >10,000 nM for ALDH3A1. The most potent compounds 3(h-l) had IC50s for killing melanoma cells ranged from 2.1 to 5.7 μM, while for colon cancer cells, it ranged from 2.5 to 5.8 μM and for multiple myeloma cells ranging from 0.3 to 4.7 μM. Toxicity studies of 3(h-l) revealed that 3h to be the least toxic multi-ALDH isoform inhibitor. Mechanistically, 3(h-l) caused increased ROS activity, lipid peroxidation, and toxic aldehyde accumulation, secondary to potent multi-ALDH isoform inhibition leading to increased apoptosis and G2/M cell cycle arrest. Together, the study details the design, synthesis, and evaluation of potent, multi-isoform ALDH inhibitors to treat cancers.
Collapse
Affiliation(s)
- Saketh S Dinavahi
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Raghavendra Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Christopher G Bazewicz
- Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; College of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Madhu Babu Battu
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, 500039, India
| | - Jyh Ming Lin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Robert J Chitren
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, United States
| | - Manoj K Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, United States
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States
| | - Krishne Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States; Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, United States.
| |
Collapse
|
27
|
Masciale V, Grisendi G, Banchelli F, D'Amico R, Maiorana A, Sighinolfi P, Pinelli M, Lovati E, Stefani A, Morandi U, Dominici M, Aramini B. Correlating tumor-infiltrating lymphocytes and lung cancer stem cells: a cross-sectional study. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:619. [PMID: 31930020 DOI: 10.21037/atm.2019.11.27] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Lung cancer stem cells (LCSCs) are endowed with high aldehyde dehydrogenase (ALDH) expression and play roles in tumor proliferation, metastasis, and drug resistance. Their elusive nature may allow them to escape the immune response by tumor-infiltrating lymphocytes (TILs), which can positively affect the outcome in non-small cell lung cancer (NSCLC) patients. Despite independent investigations on both LCSCs and TILs, the relationship between the two has been very marginally considered. We analyzed whether these two cell types may be related as a prerequisite for novel diagnostic and therapeutic approaches. Methods In this cross-sectional study, NSCLC human surgical specimens from 12 patients were tested by ALDEFLUOR assay to identify ALDHhigh cells. Fluorescence-activated cell sorting (FACS) analyses for CD3+, CD4+, and CD8+ TILs were performed in combination with immunohistochemistry evaluation. Results Statistically positive correlations were found between ALDH+ and CD8+, and between ALDH+ and CD3+ cells populations; no correlation was found between ALDH+ and CD4+ cells. The expression of CD3+ and CD8+ by cells accounted for 40.1% and 58.7%, respectively, of the variability of ALDH+ cell expression by an R-squared index, which highlights the strong correlation between TILs and LCSCs. Immunohistochemistry revealed 6-25% positive cells. Conclusions We report a correlation between cytotoxic TILs and LCSCs, which may contribute to the future development of targeted therapies focusing on the different roles of lymphocytes against lung cancer.
Collapse
Affiliation(s)
- Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Center of Statistic, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D'Amico
- Center of Statistic, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Institute of Pathology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Pamela Sighinolfi
- Institute of Pathology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Pinelli
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Eleonora Lovati
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Stefani
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
28
|
Dinavahi SS, Bazewicz CG, Gowda R, Robertson GP. Aldehyde Dehydrogenase Inhibitors for Cancer Therapeutics. Trends Pharmacol Sci 2019; 40:774-789. [DOI: 10.1016/j.tips.2019.08.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/29/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
|
29
|
Papazoglou ED, Jagirdar RM, Kouliou OA, Pitaraki E, Hatzoglou C, Gourgoulianis KI, Zarogiannis SG. In Vitro Characterization of Cisplatin and Pemetrexed Effects in Malignant Pleural Mesothelioma 3D Culture Phenotypes. Cancers (Basel) 2019; 11:cancers11101446. [PMID: 31569615 PMCID: PMC6826727 DOI: 10.3390/cancers11101446] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with poor prognosis. The main treatment for MPM is doublet chemotherapy with Cisplatin and Pemetrexed, while ongoing trials test the efficacy of pemetrexed monotherapy. However, there is lack of evidence regarding the effects of Cisplatin and Pemetrexed on MPM cell phenotypes, especially in three-dimensional (3D) cell cultures. In this study, we evaluated the effects Cisplatin and Pemetrexed on cell viability using homologous cell derived extracellular matrix (hECM) as substratum and subsequently in the following 3D cell culture phenotypes: tumor spheroid formation, tumor spheroid invasion, and collagen gel contraction. We used benign mesothelial MeT-5A cells as controls and the MPM cell lines M14K (epithelioid), MSTO (biphasic), and ZL34 (sarcomatoid). Cell viability of all cell lines was significantly decreased with all treatments. Mean tumor spheroid perimeter was reduced after treatment with Pemetrexed or the doublet therapy in all cell lines, while Cisplatin reduced the mean spheroid perimeter of MeT-5A and MSTO cells. Doublet treatment reduced the invasive capacity of spheroids of cell lines into collagenous matrices, while Cisplatin lowered the invasion of the MSTO and ZL34 cell lines, and Pemetrexed lowered the invasion of MeT-5A and ZL34 cell lines. Treatment with Pemetrexed or the combination significantly reduced the collagen gel contraction of all cell lines, while Cisplatin treatment affected only the MeT-5A and M14K cells. The results of the current study can be used as an in vitro 3D platform for testing novel drugs against MPM for ameliorating the effects of first line chemotherapeutics.
Collapse
Affiliation(s)
- Eleftherios D Papazoglou
- Department of Physiology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Rajesh M Jagirdar
- Department of Physiology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Olympia A Kouliou
- Department of Physiology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Eleanna Pitaraki
- Department of Physiology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Chrissi Hatzoglou
- Department of Physiology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Konstantinos I Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| | - Sotirios G Zarogiannis
- Department of Physiology, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece.
| |
Collapse
|
30
|
Mutti L, Peikert T, Robinson BWS, Scherpereel A, Tsao AS, de Perrot M, Woodard GA, Jablons DM, Wiens J, Hirsch FR, Yang H, Carbone M, Thomas A, Hassan R. Scientific Advances and New Frontiers in Mesothelioma Therapeutics. J Thorac Oncol 2019; 13:1269-1283. [PMID: 29966799 DOI: 10.1016/j.jtho.2018.06.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/07/2018] [Accepted: 06/17/2018] [Indexed: 12/20/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare and aggressive cancer that arises from the mesothelial surface of the pleural and peritoneal cavities, the pericardium, and rarely, the tunica vaginalis. The incidence of MPM is expected to increase worldwide in the next two decades. However, even with the use of multimodality treatment, MPM remains challenging to treat, with a 5-year survival rate of less than 5%. The International Association for the Study of Lung Cancer has gathered experts in different areas of mesothelioma research and management to summarize the most significant scientific advances and new frontiers related to mesothelioma therapeutics.
Collapse
Affiliation(s)
- Luciano Mutti
- School of Environment and Life Sciences, College of Science and Technology, Cockcroft Building, University of Salford, Salford, United Kingdom
| | - Tobias Peikert
- Department of Pulmonary Medicine, Mayo Clinic, Rochester, Minnesota
| | - Bruce W S Robinson
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Arnaud Scherpereel
- Pulmonary and Thoracic Oncology, CHU de Lille, Univ Lille, Lille, France; French National Network of Clinical Expert Centres for Malignant Pleural Mesothelioma Management
| | - Anne S Tsao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Marc de Perrot
- Division of Thoracic Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Gavitt A Woodard
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - David M Jablons
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Jacinta Wiens
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Fred R Hirsch
- International Association for the Study of Lung Cancer, Aurora, Colorado; Division of Medical Oncology, University of Colorado Cancer Center, Aurora, Colorado
| | - Haining Yang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Anish Thomas
- Development Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Raffit Hassan
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
31
|
Marcq E, Audenaerde JRV, Waele JD, Jacobs J, Loenhout JV, Cavents G, Pauwels P, Meerbeeck JPV, Smits EL. Building a Bridge between Chemotherapy and Immunotherapy in Malignant Pleural Mesothelioma: Investigating the Effect of Chemotherapy on Immune Checkpoint Expression. Int J Mol Sci 2019; 20:E4182. [PMID: 31455014 PMCID: PMC6747385 DOI: 10.3390/ijms20174182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/30/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
In light of the promising results of immune checkpoint blockade (ICPB) in malignant pleural mesothelioma (MPM), we investigated the effect of different chemotherapeutic agents on the expression of immune checkpoints (ICPs) in order to rationally design a good treatment schedule for their combination with ICP blocking antibodies. Cisplatin, oxaliplatin and pemetrexed are interesting chemotherapeutic agents to combine with immunotherapy given their immunomodulatory capacities. We looked into cisplatin and pemetrexed because their combination is used as first-line treatment of MPM. Additionally, the effect of the immunogenic chemotherapeutic agent, oxaliplatin, was also studied. Three different MPM cell lines were used for representation of both epithelioid and sarcomatoid subtypes. The desired inhibitory concentrations of the chemotherapeutic agents were determined with the SRB-assay. Allogeneic co-cultures of MPM cells with healthy donor peripheral blood mononuclear cells (PBMC) were set up to assess the effect of these chemotherapeutic agents on the expression of ICPs (PD-1, LAG-3, TIM-3) and their ligands (PD-L1, PD-L2, galectin-9). Cisplatin might be a promising treatment to combine with ICP blocking antibodies since our MPM cell lines were most susceptible to this stand-alone treatment. We found that the expression of ICPs and their ligands on both MPM cells and PBMC was mostly downregulated or unaltered when treated with chemotherapeutic agents, though no clear trend could be determined.
Collapse
Affiliation(s)
- Elly Marcq
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium.
| | | | - Jorrit De Waele
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Julie Jacobs
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Jinthe Van Loenhout
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Glenn Cavents
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
- Department of Pathology, Antwerp University Hospital, Antwerp 2650, Belgium
| | - Jan P van Meerbeeck
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
- Department of Pulmonology & Thoracic Oncology, Antwerp University Hospital, Antwerp 2650, Belgium
| | - Evelien Lj Smits
- Center for Oncological Research, University of Antwerp, Antwerp 2000, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp 2650, Belgium
| |
Collapse
|
32
|
Schmidtova S, Kalavska K, Gercakova K, Cierna Z, Miklikova S, Smolkova B, Buocikova V, Miskovska V, Durinikova E, Burikova M, Chovanec M, Matuskova M, Mego M, Kucerova L. Disulfiram Overcomes Cisplatin Resistance in Human Embryonal Carcinoma Cells. Cancers (Basel) 2019; 11:E1224. [PMID: 31443351 PMCID: PMC6769487 DOI: 10.3390/cancers11091224] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 12/17/2022] Open
Abstract
Cisplatin resistance in testicular germ cell tumors (TGCTs) is a clinical challenge. We investigated the underlying mechanisms associated with cancer stem cell (CSC) markers and modalities circumventing the chemoresistance. Chemoresistant models (designated as CisR) of human embryonal carcinoma cell lines NTERA-2 and NCCIT were derived and characterized using flow cytometry, gene expression, functional and protein arrays. Tumorigenicity was determined on immunodeficient mouse model. Disulfiram was used to examine chemosensitization of resistant cells. ALDH1A3 isoform expression was evaluated by immunohistochemistry in 216 patients' tissue samples. Chemoresistant cells were significantly more resistant to cisplatin, carboplatin and oxaliplatin compared to parental cells. NTERA-2 CisR cells exhibited altered morphology and increased tumorigenicity. High ALDH1A3 expression and increased ALDH activity were detected in both refractory cell lines. Disulfiram in combination with cisplatin showed synergy for NTERA-2 CisR and NCCIT CisR cells and inhibited growth of NTERA-2 CisR xenografts. Significantly higher ALDH1A3 expression was detected in TGCTs patients' tissue samples compared to normal testicular tissue. We characterized novel clinically relevant model of chemoresistant TGCTs, for the first time identified the ALDH1A3 as a therapeutic target in TGCTs and more importantly, showed that disulfiram represents a viable treatment option for refractory TGCTs.
Collapse
Affiliation(s)
- Silvia Schmidtova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.
| | - Katarina Kalavska
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
- Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
| | - Katarina Gercakova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Cierna
- Department of Pathology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Svetlana Miklikova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Bozena Smolkova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Verona Buocikova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Viera Miskovska
- Department of Oncology, Faculty of Medicine, Comenius University and St. Elisabeth Cancer Institute, Kolarska 12, 812 50 Bratislava, Slovakia
| | - Erika Durinikova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Monika Burikova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Michal Chovanec
- Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
| | - Miroslava Matuskova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Michal Mego
- Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
- Translational Research Unit, Faculty of Medicine, Comenius University, Klenova 1, 833 10 Bratislava, Slovakia
| | - Lucia Kucerova
- Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
33
|
Milosevic V, Kopecka J, Salaroglio IC, Libener R, Napoli F, Izzo S, Orecchia S, Ananthanarayanan P, Bironzo P, Grosso F, Tabbò F, Comunanza V, Alexa-Stratulat T, Bussolino F, Righi L, Novello S, Scagliotti GV, Riganti C. Wnt/IL-1β/IL-8 autocrine circuitries control chemoresistance in mesothelioma initiating cells by inducing ABCB5. Int J Cancer 2019; 146:192-207. [PMID: 31107974 DOI: 10.1002/ijc.32419] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a tumor with high chemoresistance and poor prognosis. MPM-initiating cells (ICs) are known to be drug resistant, but it is unknown if and how stemness-related pathways determine chemoresistance. Moreover, there are no predictive markers of IC-associated chemoresistance. Aim of this work is to clarify if and by which mechanisms the chemoresistant phenotype of MPM IC was due to specific stemness-related pathways. We generated MPM IC from primary MPM samples and compared the gene expression and chemo-sensitivity profile of IC and differentiated/adherent cells (AC) of the same patient. Compared to AC, IC had upregulated the drug efflux transporter ABCB5 that determined resistance to cisplatin and pemetrexed. ABCB5-knocked-out (KO) IC clones were resensitized to the drugs in vitro and in patient-derived xenografts. ABCB5 was transcriptionally activated by the Wnt/GSK3β/β-catenin/c-myc axis that also increased IL-8 and IL-1β production. IL-8 and IL-1β-KO IC clones reduced the c-myc-driven transcription of ABCB5 and reacquired chemosensitivity. ABCB5-KO clones had lower IL-8 and IL-1β secretion, and c-myc transcriptional activity, suggesting that either Wnt/GSK3β/β-catenin and IL-8/IL-1β signaling drive c-myc-mediated transcription of ABCB5. ABCB5 correlated with lower time-to-progression and overall survival in MPM patients treated with cisplatin and pemetrexed. Our work identified multiple autocrine loops linking stemness pathways and resistance to cisplatin and pemetrexed in MPM IC. ABCB5 may represent a new target to chemosensitize MPM IC and a potential biomarker to predict the response to the first-line chemotherapy in MPM patients.
Collapse
Affiliation(s)
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Torino, Italy
| | | | - Roberta Libener
- Pathology Division, S. Antonio and Biagio Hospital, Alessandria, Italy
| | - Francesca Napoli
- Department of Oncology, University of Torino, Torino, Italy.,Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Stefania Izzo
- Department of Oncology, University of Torino, Torino, Italy.,Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Sara Orecchia
- Pathology Division, S. Antonio and Biagio Hospital, Alessandria, Italy
| | | | - Paolo Bironzo
- Department of Oncology, University of Torino, Torino, Italy.,Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Federica Grosso
- Oncology Division, S. Antonio and Biagio Hospital, Alessandria, Italy
| | - Fabrizio Tabbò
- Department of Oncology, University of Torino, Torino, Italy.,Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Valentina Comunanza
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute - FPO IRCCS, Candiolo, Italy
| | | | - Federico Bussolino
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute - FPO IRCCS, Candiolo, Italy
| | - Luisella Righi
- Department of Oncology, University of Torino, Torino, Italy.,Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Silvia Novello
- Department of Oncology, University of Torino, Torino, Italy.,Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Giorgio V Scagliotti
- Department of Oncology, University of Torino, Torino, Italy.,Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy.,Interdepartmental Center "G. Scansetti" for the Study of Asbestos and Other Toxic Particulates, University of Torino, Torino, Italy
| |
Collapse
|
34
|
Chefetz I, Grimley E, Yang K, Hong L, Vinogradova EV, Suciu R, Kovalenko I, Karnak D, Morgan CA, Chtcherbinine M, Buchman C, Huddle B, Barraza S, Morgan M, Bernstein KA, Yoon E, Lombard DB, Bild A, Mehta G, Romero I, Chiang CY, Landen C, Cravatt B, Hurley TD, Larsen SD, Buckanovich RJ. A Pan-ALDH1A Inhibitor Induces Necroptosis in Ovarian Cancer Stem-like Cells. Cell Rep 2019; 26:3061-3075.e6. [PMID: 30865894 PMCID: PMC7061440 DOI: 10.1016/j.celrep.2019.02.032] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 01/19/2019] [Accepted: 02/07/2019] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer is typified by the development of chemotherapy resistance. Chemotherapy resistance is associated with high aldehyde dehydrogenase (ALDH) enzymatic activity, increased cancer "stemness," and expression of the stem cell marker CD133. As such, ALDH activity has been proposed as a therapeutic target. Although it remains controversial which of the 19 ALDH family members drive chemotherapy resistance, ALDH1A family members have been primarily linked with chemotherapy resistant and stemness. We identified two ALDH1A family selective inhibitors (ALDH1Ai). ALDH1Ai preferentially kills CD133+ ovarian cancer stem-like cells (CSCs). ALDH1Ai induce necroptotic CSC death, mediated, in part, by the induction of mitochondrial uncoupling proteins and reduction in oxidative phosphorylation. ALDH1Ai is highly synergistic with chemotherapy, reducing tumor initiation capacity and increasing tumor eradication in vivo. These studies link ALDH1A with necroptosis and confirm the family as a critical therapeutic target to overcome chemotherapy resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Ilana Chefetz
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Edward Grimley
- Division of Hematology-Oncology, Department of Internal Medicine, Division of Gynecology-Oncology, Department of Obstetrics and Gynecology, and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kun Yang
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Linda Hong
- Division of Gynecology-Oncology, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | | | - Radu Suciu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Ilya Kovalenko
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - David Karnak
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Cynthia A Morgan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cameron Buchman
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brandt Huddle
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Scott Barraza
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Meredith Morgan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kara A Bernstein
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA
| | - David B Lombard
- Department of Pathology and Institute of Gerontology, University of Michigan, Ann Arbor, MI, USA
| | - Andrea Bild
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Iris Romero
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL, USA
| | - Chun-Yi Chiang
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL, USA
| | - Charles Landen
- Department of Obstetrics and Gynecology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Benjamin Cravatt
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Scott D Larsen
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Ronald J Buckanovich
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Division of Gynecology-Oncology, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Division of Hematology-Oncology, Department of Internal Medicine, Division of Gynecology-Oncology, Department of Obstetrics and Gynecology, and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA; Magee-Womens Research Institute, Pittsburgh, PA, USA.
| |
Collapse
|
35
|
Ghasemi F, Sarabi PZ, Athari SS, Esmaeilzadeh A. Therapeutics strategies against cancer stem cell in breast cancer. Int J Biochem Cell Biol 2019; 109:76-81. [PMID: 30772480 DOI: 10.1016/j.biocel.2019.01.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/21/2019] [Accepted: 01/28/2019] [Indexed: 01/06/2023]
Abstract
Breast cancer is known as a most prevalent cancer and second deadly cancer, among women worldwide. Due to the high incidence rate of breast cancer and limitations of conventional therapy it seemed essential to look for new targets in cancer cells and directly target them such as target therapy on breast cancer stem cells. In this review we indicate some of therapeutic uses of cancer stem cells in breast cancer. Some strategies are targeting surface specific markers and activated signaling pathways in their microenvironment such as Notch, Hedgehog, Wnt/b-catenin, PI3K/Akt, NF-kB, BMP and TGF-β and their maintenance and drug resistance, using various miRNAs, enhancement of CSCs apoptosis, differentiation therapy, blocking epithelial to mesenchymal transition and using different natural compounds. Recent studies have shown that cancer stem cells play major roles in target therapy on breast cancer. The new manipulation approaches of cancer stem cells can be used as target therapy of breast cancer that were highlighted for immunotherapy of cancer.
Collapse
Affiliation(s)
- Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Parisa Zia Sarabi
- Molecular and Medicine Research Center, Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
36
|
Riganti C, Contino M, Guglielmo S, Perrone MG, Salaroglio IC, Milosevic V, Giampietro R, Leonetti F, Rolando B, Lazzarato L, Colabufo NA, Fruttero R. Design, Biological Evaluation, and Molecular Modeling of Tetrahydroisoquinoline Derivatives: Discovery of A Potent P-Glycoprotein Ligand Overcoming Multidrug Resistance in Cancer Stem Cells. J Med Chem 2018; 62:974-986. [DOI: 10.1021/acs.jmedchem.8b01655] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Chiara Riganti
- Dipartimento di Oncologia, Università degli Studi di Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Marialessandra Contino
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Stefano Guglielmo
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - Maria G. Perrone
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Iris C. Salaroglio
- Dipartimento di Oncologia, Università degli Studi di Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Vladan Milosevic
- Dipartimento di Oncologia, Università degli Studi di Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Roberta Giampietro
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Barbara Rolando
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - Loretta Lazzarato
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - Nicola A. Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
- Biofordrug s.r.l., Spin-off dell’Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Roberta Fruttero
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| |
Collapse
|
37
|
Bazewicz CG, Dinavahi SS, Schell TD, Robertson GP. Aldehyde dehydrogenase in regulatory T-cell development, immunity and cancer. Immunology 2018; 156:47-55. [PMID: 30387499 DOI: 10.1111/imm.13016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/10/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
The role of aldehyde dehydrogenase (ALDH) in carcinogenesis and resistance to cancer therapies is well known. Mounting evidence also suggests a potentially important role for ALDH in the induction and function of regulatory T (Treg) cells. Treg cells are important cells of the immune system involved in promoting immune tolerance and preventing aberrant immune responses to beneficial or non-harmful antigens. However, Treg cells also impair tumor immunity, leading to the progression of various carcinomas. ALDH expression and the subsequent production of retinoic acid by numerous cells, including dendritic cells, macrophages, eosinophils and epithelial cells, seems important in Treg induction and function in multiple organ systems. This is particularly evident in the gastrointestinal tract, pulmonary tract and skin, which are exposed to a myriad of environmental antigens and represent interfaces between the human body and the outside world. Expression of ALDH in Treg cells themselves may also be involved in the proliferation of these cells and resistance to certain cytotoxic therapies. Hence, inhibition of ALDH expression may be useful to treat cancer. Besides the direct effect of ALDH inhibition on carcinogenesis and resistance to cancer therapies, inhibition of ALDH could potentially augment the immune response to tumor antigens by inhibiting Treg induction, function and ability to promote immune tolerance to tumor cells in multiple cancer types.
Collapse
Affiliation(s)
- Christopher G Bazewicz
- College of Medicine, The Pennsylvania State University Medical Center, Hershey, PA, USA.,The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University Medical Center, Hershey, PA, USA
| | - Saketh S Dinavahi
- The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Pharmacology, The Pennsylvania State University Medical Center, Hershey, PA, USA
| | - Todd D Schell
- Department of Microbiology and Immunology, The Pennsylvania State University Medical Center, Hershey, PA, USA
| | - Gavin P Robertson
- The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Pharmacology, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Pathology, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Dermatology, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Department of Surgery, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Penn State Melanoma Therapeutics Program, The Pennsylvania State University Medical Center, Hershey, PA, USA.,Foreman Foundation for Melanoma Research, The Pennsylvania State University Medical Center, Hershey, PA, USA
| |
Collapse
|
38
|
Mesothelial to mesenchyme transition as a major developmental and pathological player in trunk organs and their cavities. Commun Biol 2018; 1:170. [PMID: 30345394 PMCID: PMC6191446 DOI: 10.1038/s42003-018-0180-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/28/2018] [Indexed: 12/18/2022] Open
Abstract
The internal organs embedded in the cavities are lined by an epithelial monolayer termed the mesothelium. The mesothelium is increasingly implicated in driving various internal organ pathologies, as many of the normal embryonic developmental pathways acting in mesothelial cells, such as those regulating epithelial-to-mesenchymal transition, also drive disease progression in adult life. Here, we summarize observations from different animal models and organ systems that collectively point toward a central role of epithelial-to-mesenchymal transition in driving tissue fibrosis, acute scarring, and cancer metastasis. Thus, drugs targeting pathways of mesothelium’s transition may have broad therapeutic benefits in patients suffering from these diseases. Tim Koopmans and Yuval Rinkevich review recent findings linking the mesothelium’s embryonic programs that drive epithelial-to-mesenchyme transition with adult pathologies, such as fibrosis, acute scarring, and cancer metastasis. They highlight new avenues for drug development that would target pathways of the mesothelium’s mesenchymal transition.
Collapse
|
39
|
Yang T, Gu J, Liu T, Ma H, Ma X, Tao J, Jin Y, Liang X. [Expression of Acetaldehyde Dehydrogenase in Gefitinib-resistant Human Lung Adenocarcinoma HCC-827/GR Cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2018; 21:431-436. [PMID: 29945700 PMCID: PMC6022030 DOI: 10.3779/j.issn.1009-3419.2018.06.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Tumor recurrence and drug resistance are the main causes of death in tumor patients. The family of acetaldehyde dehydrogenase (ALDH) is closely related to the proliferation, migration, invasion and resistance of tumor cells, and different ALDH subtypes are expressed in different tumor cells. The aim of this study is to elucidate the ALDH subtype in human lung adenocarcinoma HCC-827/GR cells, which resistant to the gefitinib. METHODS The human lung adenocarcinoma HCC-827 cells were used to generate the gefitinib-resistant HCC-827/GR cells; the expression of ALDH subtype in either HCC-827 or HCC-827/GR was detected by flow cytometry; The proliferative capacity and sensitivity to gefitinib of hcc-827/GR cells were analyzed by MTT assay before and after treatment with 100 μmol/L diethyllaminaldehyde (DEAB); Real-time quantitative PCR was used to detect the expression of ALDH subtypes at mRNA levels in hcc-827 cells and hcc-827/GR cells. RESULTS Compared with HCC-827 cells, the positive rate of ALDH in HCC-827/GR cells increased. The proliferation ability of HCC-827/GR cells decreased after treatment with 100 μmol/L DEAB. Compared with HCC-827 cells, the expression of ALDH1A1 and ALDH1L1 mRNA was increased in hcc-827/GR cells, but the ALDH3B2 expression was decreased. CONCLUSIONS ALDH might be used as a molecular biomarker to test the gefitinib-resistant to lung adenocarcinoma cancer cells, and the ALDH1A1 may play a role in gefitinib resistance in lung cancer.
Collapse
Affiliation(s)
- Tingting Yang
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| | - Jingjing Gu
- College of Clinical Medicine, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| | - Ting Liu
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| | - Haibin Ma
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| | - Xiaona Ma
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| | - Jin Tao
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| | - Yiran Jin
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| | - Xueyun Liang
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University,
Yinchuan 750004, China
| |
Collapse
|
40
|
Huddle BC, Grimley E, Buchman CD, Chtcherbinine M, Debnath B, Mehta P, Yang K, Morgan CA, Li S, Felton J, Sun D, Mehta G, Neamati N, Buckanovich RJ, Hurley TD, Larsen SD. Structure-Based Optimization of a Novel Class of Aldehyde Dehydrogenase 1A (ALDH1A) Subfamily-Selective Inhibitors as Potential Adjuncts to Ovarian Cancer Chemotherapy. J Med Chem 2018; 61:8754-8773. [PMID: 30221940 DOI: 10.1021/acs.jmedchem.8b00930] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aldehyde dehydrogenase (ALDH) activity is commonly used as a marker to identify cancer stem-like cells. The three ALDH1A isoforms have all been individually implicated in cancer stem-like cells and in chemoresistance; however, which isoform is preferentially expressed varies between cell lines. We sought to explore the structural determinants of ALDH1A isoform selectivity in a series of small-molecule inhibitors in support of research into the role of ALDH1A in cancer stem cells. An SAR campaign guided by a cocrystal structure of the HTS hit CM39 (7) with ALDH1A1 afforded first-in-class inhibitors of the ALDH1A subfamily with excellent selectivity over the homologous ALDH2 isoform. We also discovered the first reported modestly selective single isoform 1A2 and 1A3 inhibitors. Two compounds, 13g and 13h, depleted the CD133+ putative cancer stem cell pool, synergized with cisplatin, and achieved efficacious concentrations in vivo following IP administration. Compound 13h additionally synergized with cisplatin in a patient-derived ovarian cancer spheroid model.
Collapse
Affiliation(s)
| | | | - Cameron D Buchman
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | - Mikhail Chtcherbinine
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | | | - Pooja Mehta
- Department of Materials Science Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Kun Yang
- Division of Hematology Oncology, Department of Internal Medicine , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Cynthia A Morgan
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | - Siwei Li
- Department of Pharmaceutical Sciences, College of Pharmacy ; University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Jeremy Felton
- Department of Pharmaceutical Sciences, College of Pharmacy ; University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy ; University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Geeta Mehta
- Department of Materials Science Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Department of Biomedical Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States.,Macromolecular Science and Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | | | - Ronald J Buckanovich
- Division of Hematology Oncology, Department of Internal Medicine , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , Indiana 46202 , United States
| | | |
Collapse
|
41
|
Kim MC, Hwang SH, Kim NY, Lee HS, Ji S, Yang Y, Kim Y. Hypoxia promotes acquisition of aggressive phenotypes in human malignant mesothelioma. BMC Cancer 2018; 18:819. [PMID: 30111297 PMCID: PMC6094475 DOI: 10.1186/s12885-018-4720-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 08/02/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Hypoxia is a hallmark of the solid tumor microenvironment and is associated with poor outcomes in cancer patients. The present study was performed to investigate mechanisms underlying the hypoxia-induced phenotypic changes using human malignant mesothelioma (HMM) cells. METHODS Hypoxic conditions were achieved by incubating HMM cells in the air chamber. The effect of hypoxia on phenotype changes in HMM cells was investigated by performing in vitro clonogenicity, drug resistance, migration, and invasion assays. Signaling pathways and molecules involved in the more aggressive behaviors of HMM cells under hypoxia were investigated. A two-tailed unpaired Student's t-test or one-way ANOVA with Bonferroni post-test correction was used in this study. RESULTS Hypoxic conditions upregulated hypoxia-inducible factor 1 alpha (HIF-1α) and HIF-2α in parallel with the upregulation of its target, Glut-1, in HMM cells. In vitro clonogenicity of HMM cells was significantly increased in hypoxic conditions, but the proliferation of cells at a high density in hypoxia was lower than that in normoxic conditions. The expression levels of HIF-2α and Oct4 were increased in hypoxic HMM cells. The percentage of cells with high CD44 expression was significantly higher in HMM cells cultured in hypoxia than those cultured in normoxia. Hypoxia significantly enhanced the resistance of HMM cells to cisplatin, which occurred through cytoprotection against cisplatin-induced apoptosis. While cisplatin treatment decreased the ratio of Bcl-2 to Bax in normoxic condition, hypoxia conversely increased the ratio in HMM cells treated with cisplatin. Hypoxia increased the mobility and invasiveness of HMM cells. Epithelial to mesenchymal transition was promoted, which was indicated by the repression of E-cadherin and the concomitant increase of vimentin in HMM cells. CONCLUSIONS The data illustrated that hypoxic conditions augmented the aggressive phenotypes of HMM cells at the biological and molecular levels. The present study provides valuable background information beginning to understand aggressiveness of HMM in tumor microenvironments, suggesting that a control measure for tumor hypoxia may be an effective therapeutic strategy to reduce the aggressiveness of cancer cells in HMM patients.
Collapse
Affiliation(s)
- Myung-Chul Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Sung-Hyun Hwang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Na-Yon Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Hong-Seok Lee
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Sumin Ji
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Yeseul Yang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.,BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Yongbaek Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea. .,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
42
|
Differential regulation of the sphere formation and maintenance of cancer-initiating cells of malignant mesothelioma via CD44 and ALK4 signaling pathways. Oncogene 2018; 37:6357-6367. [PMID: 30061637 PMCID: PMC6283855 DOI: 10.1038/s41388-018-0405-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 06/03/2018] [Accepted: 06/19/2018] [Indexed: 01/06/2023]
Abstract
Malignant mesothelioma (MM) has a poor prognosis and is largely resistant to standard treatments, so it is important to seek novel therapeutic strategies for this disease. Cancer-initiating cells (CICs) were previously identified in MM using stem cell-associated markers in combination with spheroid cultures. However, the mechanisms underlying the induction and maintenance of CICs in MM remain to be fully explored. Here we showed that the CICs, which had high aldehyde dehydrogenase levels (ALDHbright) and stem cell-associated genes, were expanded in MM cells cultured under sphere-forming conditions. The MM spheroids also initiated tumors in immunodeficient mice more efficiently than did conventional adherent MM cells. In the MM spheroids, the expression of hyaluronan (HA) synthases was upregulated. Inhibiting the HA synthesis or CD44 functions by gene knockdown or neutralizing antibody abolished the formation of large-sized spheroids and the expansion of ALDHbright CICs. The expression of activin-A was also increased in the spheroids, and type I activin-A receptor subunit (ALK4) was upregulated in the ALDHbright CICs. The neutralization of activin-A or functional inactivation of ALK4 diminished the ALDHbright CICs without affecting spheroid formation. The knockdown of CD44 or ALK4 strongly suppressed the tumor growth in immunodeficient mice. These results together suggest that the HA–CD44 and activin-A–ALK4 pathways differentially regulate the spheroid formation and maintenance of ALDHbright CICs in MM cells, and that both pathways play critical roles in tumor growth in immunodeficient hosts. Our findings provide a novel therapeutic option for MM that targets signaling pathways that promote the CIC compartment through CD44 and ALK4.
Collapse
|
43
|
Wei L, Chintala S, Ciamporcero E, Ramakrishnan S, Elbanna M, Wang J, Hu Q, Glenn ST, Murakami M, Liu L, Gomez EC, Sun Y, Conroy J, Miles KM, Malathi K, Ramaiah S, Anbarasu A, Woloszynska-Read A, Johnson CS, Conroy J, Liu S, Morrison CD, Pili R. Genomic profiling is predictive of response to cisplatin treatment but not to PI3K inhibition in bladder cancer patient-derived xenografts. Oncotarget 2018; 7:76374-76389. [PMID: 27823983 PMCID: PMC5363516 DOI: 10.18632/oncotarget.13062] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 10/22/2016] [Indexed: 11/25/2022] Open
Abstract
Purpose Effective systemic therapeutic options are limited for bladder cancer. In this preclinical study we tested whether bladder cancer gene alterations may be predictive of treatment response. Experimental design We performed genomic profiling of two bladder cancer patient derived tumor xenografts (PDX). We optimized the exome sequence analysis method to overcome the mouse genome interference. Results We identified a number of somatic mutations, mostly shared by the primary tumors and PDX. In particular, BLCAb001, which is less responsive to cisplatin than BLCAb002, carried non-sense mutations in several genes associated with cisplatin resistance, including MLH1, BRCA2, and CASP8. Furthermore, RNA-Seq analysis revealed the overexpression of cisplatin resistance associated genes such as SLC7A11, TLE4, and IL1A in BLCAb001. Two different PIK3CA mutations, E542K and E545K, were identified in BLCAb001 and BLCAb002, respectively. Thus, we tested whether the genomic profiling was predictive of response to a dual PI3K/mTOR targeting agent, LY3023414. Despite harboring similar PIK3CA mutations, BLCAb001 and BLCAb002 exhibited differential response, both in vitro and in vivo. Sustained target modulation was observed in the sensitive model BLCAb002 but not in BLCAb001, as well as decreased autophagy. Interestingly, computational modelling of mutant structures and affinity binding to PI3K revealed that E542K mutation was associated with weaker drug binding than E545K. Conclusions Our results suggest that the presence of activating PIK3CA mutations may not necessarily predict in vivo treatment response to PI3K targeted therapies, while specific gene alterations may be predictive for cisplatin response in bladder cancer models and, potentially, in patients as well.
Collapse
Affiliation(s)
- Lei Wei
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sreenivasulu Chintala
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Eric Ciamporcero
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Swathi Ramakrishnan
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - May Elbanna
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Jianmin Wang
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Qiang Hu
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sean T Glenn
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Mitsuko Murakami
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lu Liu
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Eduardo Cortes Gomez
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Yuchen Sun
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jacob Conroy
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kiersten Marie Miles
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kullappan Malathi
- Medical & Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical & Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Anand Anbarasu
- Medical & Biological Computing Laboratory, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Anna Woloszynska-Read
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Candace S Johnson
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jeffrey Conroy
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Carl D Morrison
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Roberto Pili
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, NY, USA.,Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
44
|
di Martino S, Amoreo CA, Nuvoli B, Galati R, Strano S, Facciolo F, Alessandrini G, Pass HI, Ciliberto G, Blandino G, De Maria R, Cioce M. HSP90 inhibition alters the chemotherapy-driven rearrangement of the oncogenic secretome. Oncogene 2018; 37:1369-1385. [PMID: 29311642 DOI: 10.1038/s41388-017-0044-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/24/2017] [Accepted: 09/10/2017] [Indexed: 12/29/2022]
Abstract
Adaptive resistance to therapy is a hallmark of cancer progression. To date, it is not entirely clear how microenvironmental stimuli would mediate emergence of therapy-resistant cell subpopulations, although a rearrangement of the cancer cell secretome following therapy-induced stress can be pivotal for such a process. Here, by using the highly chemoresistant malignant pleural mesothelioma (MPM) as an experimental model, we unveiled a key contribution of the chaperone HSP90 at assisting a chemotherapy-instigated Senescence-Associated-Secretory-Phenotype (SASP). Thus, administration of a clinical trial grade, HSP90, inhibitor blunted the release of several cytokines by the chemotherapy-treated MPM cells, including interleukin (IL)-8. Reduction of IL-8 levels hampered the FAK-AKT signaling and inhibited 3D growth and migration. This correlated with downregulation of key EMT and chemoresistance genes and affected the survival of chemoresistant ALDHbright cell subpopulations. Altogether, inhibition of HSP90 provoked a switch from a pro-tumorigenic SASP to a pro-apoptotic senescence status, thus resulting in chemosensitizing effects. In mouse xenografts treated with first-line agents, inhibiting HSP90 blunted FAK activation and reduced the expression of ALDH1A3 and the levels of circulating human IL-8, these latter strongly correlating with the effect on tumor growth. We validated the above findings in primary mesothelioma cultures, a more clinically relevant model. We unveiled here a key contribution of the chaperone HSP90 at assisting the secretory stress in chemotherapy-treated cells, which may warrant further investigation in combinatorial therapeutic settings.
Collapse
Affiliation(s)
- Simona di Martino
- Oncogenomic and Epigenetic Unit Regina Elena National Cancer Institute, Rome, Italy
| | | | - Barbara Nuvoli
- Preclinical Models and New Therapeutic Agents Unit, Regina Elena National Cancer Institute, Rome, Italy
| | - Rossella Galati
- Preclinical Models and New Therapeutic Agents Unit, Regina Elena National Cancer Institute, Rome, Italy
| | - Sabrina Strano
- Molecular Chemoprevention Unit, Regina Elena National Cancer Institute, Rome, Italy.,Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Francesco Facciolo
- Department of Thoracic Surgery, Regina Elena National Cancer Institute, Rome, Italy
| | | | - Harvey I Pass
- New York University School of Medicine, Department of Cardiothoracic Surgery, New York, NY, USA
| | - Gennaro Ciliberto
- Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit Regina Elena National Cancer Institute, Rome, Italy.,Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Ruggero De Maria
- Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy. .,Current address: Institute of General Pathology, Catholic University and Gemelli Polyclinic, Rome, Italy.
| | - Mario Cioce
- Oncogenomic and Epigenetic Unit Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
45
|
Sai S, Suzuki M, Kim EH, Hayashi M, Vares G, Yamamoto N, Miyamoto T. Effects of carbon ion beam alone or in combination with cisplatin on malignant mesothelioma cells in vitro. Oncotarget 2017; 9:14849-14861. [PMID: 29599911 PMCID: PMC5871082 DOI: 10.18632/oncotarget.23756] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 12/15/2017] [Indexed: 12/22/2022] Open
Abstract
Malignant mesothelioma (MM) is extremely aggressive and a typical refractory cancer. In this study we investigated how effective on killing MM cells by carbon ion beam alone or in combination with cisplatin (CDDP) in vitro. Carbon ion beam (at the center of SOBP with 50 keV/µm of average LET) dose-independently suppressed MM cells MESO-1 and H226 cell viability and in combination with CDDP (25 μM) significantly enhanced its action. Relative biological effectiveness (RBE) values at 73 keV/μm and 13 keV/μm portion of carbon ion beam was estimated as 2.82-2.93 and 1.19-1.22 at D10 level relative to X-ray, respectively by using colony formation assay. Quantitative real time PCR analysis showed that expression of apoptosis-related BAX and autophagy-related Beclin1 and ATG7 was significantly enhanced by carbon ion beam alone or in combination with CDDP. Apoptosis analysis showed that caspase 3/7 activity and the percentage of apoptotic cells was dose-dependently increased after carbon ion beam and it was further increased when combined with CDDP. Spheroid formation ability of cancer stem like CD44+/CD26+ cells was significantly inhibited by carbon ion beam combined with CDDP. Besides, carbon ion beam combined with cisplatin significantly inhibited cell cycle progression (sub-G1 arrest) and induced more large number of γH2AX foci. In conclusion, carbon ion beam combined with CDDP has superior potential to kill MM cells including CSCs with enhanced apoptosis.
Collapse
Affiliation(s)
- Sei Sai
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masao Suzuki
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Eun Ho Kim
- Division of Applied Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Gongneung-dong, Nowon-Gu, Seoul, South Korea
| | - Mitsuhiro Hayashi
- Breast Center, Dokkyo Medical University Hospital, Mibu-machi, Shimotsuga-gun, Tochigi, Japan
| | - Guillaume Vares
- Okinawa Institute of Science and Technology (OIST), Advanced Medical Instrumentation Unit, Onna-son, Okinawa, Japan
| | - Naoyoshi Yamamoto
- Hospital of the National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Sciences and Technology, Chiba, Japan
| | - Tadaaki Miyamoto
- Chiba Foundation for Health Promotion and Disease Prevention, Chiba, Japan
| |
Collapse
|
46
|
Chen X, Liao R, Li D, Sun J. Induced cancer stem cells generated by radiochemotherapy and their therapeutic implications. Oncotarget 2017; 8:17301-17312. [PMID: 28038467 PMCID: PMC5370042 DOI: 10.18632/oncotarget.14230] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/13/2016] [Indexed: 12/26/2022] Open
Abstract
Local and distant recurrence of malignant tumors following radio- and/or chemotherapy correlates with poor prognosis of patients. Among the reasons for cancer recurrence, preexisting cancer stem cells (CSCs) are considered the most likely cause due to their properties of self-renewal, pluripotency, plasticity and tumorigenicity. It has been demonstrated that preexisting cancer stem cells derive from normal stem cells and differentiated somatic cells that undergo transformation and dedifferentiation respectively under certain conditions. However, recent studies have revealed that cancer stem cells can also be induced from non-stem cancer cells by radiochemotherapy, constituting the subpopulation of induced cancer stem cells (iCSCs). These findings suggest that radiochemotherapy has the side effect of directly transforming non-stem cancer cells into induced cancer stem cells, possibly contributing to tumor recurrence and metastasis. Therefore, drugs targeting cancer stem cells or preventing dedifferentiation of non-stem cancer cells can be combined with radiochemotherapy to improve its antitumor efficacy. The current review is to investigate the mechanisms by which induced cancer stem cells are generated by radiochemotherapy and hence provide new strategies for cancer treatment.
Collapse
Affiliation(s)
- Xiewan Chen
- Medical English Department, College of Basic Medicine, Third Military Medical University, Chongqing, China.,Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rongxia Liao
- Medical English Department, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Dezhi Li
- Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jianguo Sun
- Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
47
|
Mine N, Yamamoto S, Saito N, Sato T, Sakakibara K, Kufe DW, VonHoff DD, Kawabe T. CBP501 suppresses macrophage induced cancer stem cell like features and metastases. Oncotarget 2017; 8:64015-64031. [PMID: 28969049 PMCID: PMC5609981 DOI: 10.18632/oncotarget.19292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/02/2017] [Indexed: 02/07/2023] Open
Abstract
CBP501 is an anti-cancer drug candidate which has been shown to increase cis-diamminedichloro-platinum (II) (CDDP) uptake into cancer cell through calmodulin (CaM) inhibition. However, the effects of CBP501 on the cells in the tumor microenvironment have not been addressed. Here, we investigated new aspects of the potential anti-tumor mechanism of action of CBP501 by examining its effects on the macrophages. Macrophages contribute to cancer-related inflammation and sequential production of cytokines such as IL-6 and TNF-α which cause various biological processes that promote tumor initiation, growth and metastasis (1). These processes include the epithelial to mesenchymal transition (EMT) and cancer stem cell (CSC) formation, which are well-known, key events for metastasis. The present work demonstrates that CBP501 suppresses lipopolysaccharide (LPS)-induced production of IL-6, IL-10 and TNF-α by macrophages. CBP501 also suppressed formation of the tumor spheroids by culturing with conditioned medium from the LPS-stimulated macrophage cell line RAW264.7. Moreover, CBP501 suppressed expression of ABCG2, a marker for CSCs, by inhibiting the interaction between cancer cells expressing VCAM-1 and macrophages expressing VLA-4. Consistently with these results, CBP501 in vivo suppressed metastases of a tumor cell line, 4T1, one which is insensitive to combination treatment of CBP501 and CDDP in vitro. Taken together, these results offer potential new, unanticipated advantages of CBP501 treatment in anti-tumor therapy through a mechanism that entails the suppression of interactions between macrophages and cancer cells with suppression of sequential CSC-like cell formation in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel D VonHoff
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, USA
| | | |
Collapse
|
48
|
Nishino M, Ozaki M, Hegab AE, Hamamoto J, Kagawa S, Arai D, Yasuda H, Naoki K, Soejima K, Saya H, Betsuyaku T. Variant CD44 expression is enriching for a cell population with cancer stem cell-like characteristics in human lung adenocarcinoma. J Cancer 2017; 8:1774-1785. [PMID: 28819374 PMCID: PMC5556640 DOI: 10.7150/jca.19732] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022] Open
Abstract
Background: Preliminary studies have identified cancer stem cells (CSCs) in various cancers and there are several ongoing clinical studies targeting these cells. CD44 (standard or variant isoforms) and/or aldehyde dehydrogenase (ALDH) expression is the most commonly used markers for the identification of CSCs. The goal of the current study was to examine the ability of CD44v, either alone or in combination with ALDH, to identify CSCs within human lung cancer cells lines. Methods: We examined several lung adenocarcinoma cell lines for the ability of CD44v and/or ALDH expression to enrich for cells with CSC characteristics such as in vitro differential proliferation rate, chemotherapeutic-resistance, tumorsphere formation, and in vivo tumorigenicity. We also compared their in vivo secondary tumor formation, and histological characteristics of their xenograft tumors, and examined their expression of PD-L1, EGFR, xCT, and reactive oxygen species (ROS). Results: Both CD44vhigh/ALDHhigh and CD44vhigh/ALDHlow cells were enriched in cells with CSC characteristics, with the CD44vhigh/ALDHlow cells being more proliferative and more resistant to chemotherapeutics, whereas CD44vhigh/ALDHhigh cells were more efficient in forming tumorspheres in vitro, in making primary xenograft tumors, and in propagating secondary tumors in vivo. Applying stricter sorting gates to select for cells with the highest CD44v/ALDH expression caused the CD44vhigh/ALDHlow cells to lose their high proliferation rates and chemotherapeutic resistance ability, but enriched for the tumorsphere-forming cells among the CD44vhigh/ALDHhigh and CD44vhigh/ALDHlow cells. CD44vhigh expression was associated with PD-L1 and xCT expression in both H1650 and HCC827 cells. This association was not modified by ALDH expression in the H1650 cell line. However, in the HCC827 cell line, ALDH expression was negatively associated with PD-L1 and positively associated with xCT expression. Conclusion: Lung adenocarcinoma cells with high CD44v expression are enriched for CSCs. Addition of ALDH as an enrichment marker bestowed some CSCs characteristics to CD44vhigh/ALDHlow cells and others to CD44vhigh/ALDHhigh cells. We propose that lung adenocarcinoma contains different CSCs, each of them endowed with different CSC characteristics.
Collapse
Affiliation(s)
- Makoto Nishino
- Division of Pulmonary Medicine, Department of Medicine, and
| | - Mari Ozaki
- Division of Pulmonary Medicine, Department of Medicine, and
| | - Ahmed E Hegab
- Division of Pulmonary Medicine, Department of Medicine, and
| | - Junko Hamamoto
- Division of Pulmonary Medicine, Department of Medicine, and
| | - Shizuko Kagawa
- Division of Pulmonary Medicine, Department of Medicine, and
| | - Daisuke Arai
- Division of Pulmonary Medicine, Department of Medicine, and
| | | | | | - Kenzo Soejima
- Division of Pulmonary Medicine, Department of Medicine, and
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | |
Collapse
|
49
|
Singh A, Pruett N, Hoang CD. In vitro experimental models of mesothelioma revisited. Transl Lung Cancer Res 2017; 6:248-258. [PMID: 28713670 DOI: 10.21037/tlcr.2017.04.12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a biologically unusual, highly aggressive cancer that defies current multimodality treatments. Epidemiologic data suggest that this malignancy has not abated despite increasingly strict environmental regulations on asbestos, the putative causative agent for sporadic cases. An incomplete understanding of all the factors mechanistically driving mesothelioma is largely responsible for the current lack of curative treatments. Many approaches have been employed to ascertain the step-by-step molecular events involved in mesothelioma oncogenesis including in vitro, small animal in vivo, and human experimental models; though clearly defined, druggable mechanisms still are elusive. Importantly, the foundation of the latest accepted model of tumor initiation is derived from in vitro systems. A thorough review of in vitro mesothelioma oncogenesis models may suggest further opportunities for discovery.
Collapse
Affiliation(s)
- Anand Singh
- Section of Thoracic Surgery, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nathanael Pruett
- Section of Thoracic Surgery, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chuong D Hoang
- Section of Thoracic Surgery, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
Woodard GA, Yang YL, You L, Jablons DM. Drug development against the hippo pathway in mesothelioma. Transl Lung Cancer Res 2017; 6:335-342. [PMID: 28713678 DOI: 10.21037/tlcr.2017.06.02] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advances in the treatments for malignant pleural mesothelioma (MPM) have been disappointing until recently. Conventional cytotoxic drugs fail in MPM in part because they do not address the cancer stem cell population or stem cell pathways that drive tumor resistance and resurgence following treatment. The Hippo stem cell pathway regulates cell contact inhibition with tumor suppressor genes such as NF2 (Neurofibromatosis 2) upstream controlling YAP (Yes-associated protein 1) oncogenes. NF2 is mutated in 40-50% of all MPM and downstream YAP is constitutively active in greater than 70% of MPM, making the downstream YAP/TEAD (transcriptional enhancer associate domain) complex the ultimate target. Novel small molecule YAP inhibitors are showing promising results in preclinical studies and may prove to be effective chemotherapy drugs in MPM.
Collapse
Affiliation(s)
- Gavitt A Woodard
- Department of Surgery, University of California, San Francisco, USA
| | - Yi-Lin Yang
- Department of Surgery, University of California, San Francisco, USA
| | - Liang You
- Department of Surgery, University of California, San Francisco, USA
| | - David M Jablons
- Department of Surgery, University of California, San Francisco, USA
| |
Collapse
|