1
|
Thakur A, Rana M, Ritika, Mathew J, Nepali S, Pan CH, Liou JP, Nepali K. Small molecule tractable PARP inhibitors: Scaffold construction approaches, mechanistic insights and structure activity relationship. Bioorg Chem 2023; 141:106893. [PMID: 37783100 DOI: 10.1016/j.bioorg.2023.106893] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Diverse drug design strategies viz. molecular hybridization, substituent installation, scaffold hopping, isosteric replacement, high-throughput screening, induction and separation of chirality, structure modifications of phytoconstituents and use of structural templates have been exhaustively leveraged in the last decade to load the chemical toolbox of PARP inhibitors. Resultantly, numerous promising scaffolds have been pinpointed that in turn have led to the resuscitation of the credence to PARP inhibitors as cancer therapeutics. This review briefly presents the physiological functions of PARPs, the pharmacokinetics, and pharmacodynamics, and the interaction profiles of FDA-approved PARP inhibitors. Comprehensively covered is the section on the drug design strategies employed by drug discovery enthusiasts for furnishing PARP inhibitors. The impact of structural variations in the template of designed scaffolds on enzymatic and cellular activity (structure-activity relationship studies) has been discussed. The insights gained through the biological evaluation such as profiling of physicochemical properties andin vitroADME properties, PK assessments, and high-dose pharmacology are covered.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Ritika
- College of Medicine, Taipei Medical University, Taipei 110031, Taiwan
| | - Jacob Mathew
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Sanya Nepali
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Chun-Hsu Pan
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
2
|
Yi XF, Gao RL, Sun L, Wu ZX, Zhang SL, Huang LT, Han CB, Ma JT. Dual antitumor immunomodulatory effects of PARP inhibitor on the tumor microenvironment: A counterbalance between anti-tumor and pro-tumor. Biomed Pharmacother 2023; 163:114770. [PMID: 37105074 DOI: 10.1016/j.biopha.2023.114770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/10/2023] [Accepted: 04/22/2023] [Indexed: 04/29/2023] Open
Abstract
Poly (ADP-ribose)-polymerases (PARPs) play an essential role in the maintenance of genome integrity, DNA repair, and apoptosis. PARP inhibitors (PARPi) exert antitumor effects via synthetic lethality and PARP trapping. PARPi impact the antitumor immune response by modulating the tumor microenvironment, and their effect has dual properties of promoting and inhibiting the antitumor immune response. PARPi promote M1 macrophage polarization, antigen presentation by dendritic cells, infiltration of B and T cells and their killing capacity and inhibit tumor angiogenesis. PARPi can also inhibit the activation and function of immune cells by upregulating PD-L1. In this review, we summarize the dual immunomodulatory effects and possible underlying mechanisms of PARPi, providing a basis for the design of combination regimens for clinical treatment and the identification of populations who may benefit from these therapies.
Collapse
Affiliation(s)
- Xiao-Fang Yi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ruo-Lin Gao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhi-Xuan Wu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shu-Ling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng-Bo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Jie-Tao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
PARP Inhibitor Inhibits the Vasculogenic Mimicry through a NF-κB-PTX3 Axis Signaling in Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms232416171. [PMID: 36555812 PMCID: PMC9785325 DOI: 10.3390/ijms232416171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) are targeted therapies that inhibit PARP proteins which are involved in a variety of cell functions. PARPi may act as modulators of angiogenesis; however, the relationship between PARPi and the vasculogenic mimicry (VM) in breast cancer remains unclear. To determine whether PARPi regulate the vascular channel formation, we assessed whether the treatment with olaparib, talazoparib and veliparib inhibits the vascular channel formation by breast cancer cell lines. Here, we found that PARPi act as potent inhibitors of the VM formation in triple negative breast cancer cells, independently of the BRCA status. Mechanistically, we find that PARPi trigger and inhibit the NF-κB signaling, leading to the inhibition of the VM. We further show that PARPi decrease the expression of the angiogenic factor PTX3. Moreover, PTX3 rescued the PARPi-inhibited VM inhibition. In conclusion, our results indicate that PARPi, by targeting the VM, may provide a new therapeutic approach for triple negative breast cancer.
Collapse
|
4
|
Mou K, Zhou Y, Mu X, Zhang J, Wang L, Ge R. PARP1 Is a Prognostic Marker and Targets NFATc2 to Promote Carcinogenesis in Melanoma. Genet Test Mol Biomarkers 2022; 26:503-511. [DOI: 10.1089/gtmb.2021.0214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Kuanhou Mou
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yan Zhou
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xin Mu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jian Zhang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Lijuan Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Rui Ge
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
5
|
Demény MA, Virág L. The PARP Enzyme Family and the Hallmarks of Cancer Part 2: Hallmarks Related to Cancer Host Interactions. Cancers (Basel) 2021; 13:2057. [PMID: 33923319 PMCID: PMC8123211 DOI: 10.3390/cancers13092057] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/07/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Poly (ADP-ribose) polymerases (PARPs) modify target proteins with a single ADP-ribose unit or with a poly (ADP-ribose) (PAR) polymer. PARP inhibitors (PARPis) recently became clinically available for the treatment of BRCA1/2 deficient tumors via the synthetic lethality paradigm. This personalized treatment primarily targets DNA damage-responsive PARPs (PARP1-3). However, the biological roles of PARP family member enzymes are broad; therefore, the effects of PARPis should be viewed in a much wider context, which includes complex effects on all known hallmarks of cancer. In the companion paper (part 1) to this review, we presented the fundamental roles of PARPs in intrinsic cancer cell hallmarks, such as uncontrolled proliferation, evasion of growth suppressors, cell death resistance, genome instability, replicative immortality, and reprogrammed metabolism. In the second part of this review, we present evidence linking PARPs to cancer-associated inflammation, anti-cancer immune response, invasion, and metastasis. A comprehensive overview of the roles of PARPs can facilitate the identification of novel cancer treatment opportunities and barriers limiting the efficacy of PARPi compounds.
Collapse
Affiliation(s)
- Máté A. Demény
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, 4032 Debrecen, Hungary
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, 4032 Debrecen, Hungary
| |
Collapse
|
6
|
Lisi L, Chiavari M, Ciotti GMP, Lacal PM, Navarra P, Graziani G. DNA inhibitors for the treatment of brain tumors. Expert Opin Drug Metab Toxicol 2020; 16:195-207. [PMID: 32067518 DOI: 10.1080/17425255.2020.1729352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction: The worldwide incidence of central nervous system (CNS) primary tumors is increasing. Most of the chemotherapeutic agents used for treating these cancer types induce DNA damage, and their activity is affected by the functional status of repair systems involved in the detection or correction of DNA lesions. Unfortunately, treatment of malignant high-grade tumors is still an unmet medical need.Areas covered: We summarize the action mechanisms of the main DNA inhibitors used for the treatment of brain tumors. In addition, studies on new agents or drug combinations investigated for this indication are reviewed, focusing our attention on clinical trials that in the last 3 years have been completed, terminated or are still recruiting patients.Expert opinion: Much still needs to be done to render aggressive CNS tumors curable or at least to transform them from lethal to chronic diseases, as it is possible for other cancer types. Drugs with improved penetration in the CNS, toxicity profile, and activity against primary and recurrent tumors are eagerly needed. Targeted agents with innovative mechanisms of action and ability to harness the cells of the tumor microenvironment against cancer cells represent a promising approach for improving the clinical outcome of CNS tumors.
Collapse
Affiliation(s)
- Lucia Lisi
- Department of Safety and Bioethics, Catholic University Medical School, Rome, Italy
| | - Marta Chiavari
- Department of Safety and Bioethics, Catholic University Medical School, Rome, Italy
| | | | - Pedro M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - Pierluigi Navarra
- Department of Safety and Bioethics, Catholic University Medical School, Rome, Italy.,Department of Safety and Bioethics, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
7
|
Raineri A, Prodomini S, Fasoli S, Gotte G, Menegazzi M. Influence of onconase in the therapeutic potential of PARP inhibitors in A375 malignant melanoma cells. Biochem Pharmacol 2019; 167:173-181. [PMID: 31185226 DOI: 10.1016/j.bcp.2019.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022]
Abstract
Human malignant melanoma is one of the most aggressive cancers, accompanied with poor prognosis, metastatic evolution and high mortality. Many strategies have been developed using BRAF and MEK inhibitors in spite of the classic therapy with alkylating agents, but failure related to the ability of the tumor to activate alternative proliferation pathways occurred after promising initial successes. Poly(ADP-ribose) polymerase (PARP) enzymes are well known to be crucial for DNA damage response, and PARP inhibition results in the accumulation of DNA strand breaks that induce cell injury. For this reason, PARP-inhibitors (PARPi) have become promising tools to counteract many cancer types. One of the most used by clinicians is olaparib, that, however, showed again cancer resistance in patients. Thus, new generation molecules have been designed mainly to counteract this problem. Among them, we chose to test AZD2461 on the particularly aggressive human melanoma A375 cell line. This drug is a PARPi significantly less prone than olaparib to undergo the P-glycoprotein-mediated efflux mechanism, one of those responsible for resistance, that in turn is the main adversity in melanoma therapy. Then, we analysed AZD2461 also together with the enzyme onconase (ONC) on the same A375 cells, to investigate if the combination of drugs could possibly increase the in vitro antitumor activity. ONC is a small amphibian "pancreatic-type" ribonuclease that is able to exert a remarkable antitumor activity against many cancers, either in vitro or in vivo, principally because it can evade the ubiquitous ribonuclease cytosolic inhibitor thanks to its structural determinants. Hence, ONC became relevant in the use of protein-drug strategies against incurable cancers. The studies performed in this work showed that both drugs definitely affect A375 cells viability by inducing cytostatic and pro-apoptotic effects in a time- and dose-dependent manner, either if administered alone or in combination. Although we registered low synergistic effects with the combination of the two drugs, we found that AZD2461 did not induce resistance in A375 after two months treatment with high concentration of this molecule. Moreover, we underline that A375 cells treated for a prolonged time with AZD2461 were definitely more susceptible than parental A375 cells to the pro-apoptotic action of ONC. Considering also the different inhibitory effects of the two drugs on TNF-α gene expression and NF-κB DNA-binding, the tuning of their combined delivery to the A375 tumor cell line might open a promising scenario for future therapeutic applications devoted to defeat human melanoma.
Collapse
Affiliation(s)
- Alice Raineri
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Sara Prodomini
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Sabrina Fasoli
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Giovanni Gotte
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Marta Menegazzi
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy.
| |
Collapse
|
8
|
Fratangelo F, Camerlingo R, Carriero MV, Pirozzi G, Palmieri G, Gentilcore G, Ragone C, Minopoli M, Ascierto PA, Motti ML. Effect of ABT-888 on the apoptosis, motility and invasiveness of BRAFi-resistant melanoma cells. Int J Oncol 2018; 53:1149-1159. [PMID: 29956724 PMCID: PMC6065454 DOI: 10.3892/ijo.2018.4457] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/12/2018] [Indexed: 12/26/2022] Open
Abstract
Melanoma is a molecularly heterogeneous disease with many genetic mutations and altered signaling pathways. Activating mutations in the BRAF oncogene are observed in approximately 50% of cutaneous melanomas and the use of BRAF inhibitor (BRAFi) compounds has been reported to improve the outcome of patients with BRAF-mutated metastatic melanoma. However, the majority of these patients develop resistance within 6-8 months following the initiation of BRAFi treatment. In this study, we examined the possible use of the poly(ADP-ribose) polymerase 1 (PARP1) inhibitor, ABT-888 (veliparib), as a novel molecule that may be successfully employed in the treatment of BRAFi-resistant melanoma cells. Sensitive and resistant to BRAFi dabrafenib A375 cells were exposed to increasing concentrations of ABT-888. Cell viability and apoptosis were assessed by MTT assay and Annexin V-FITC analysis, respectively. The cell migratory and invasive ability was investigated using the xCELLigence technology and Boyden chamber assays, respectively. ABT-888 was found to reduce cell viability and exhibited pro-apoptotic activity in melanoma cell lines, independently from the BRAF/NRAS mutation status, in a dose-dependent manner, with the maximal effect being reached in the 25-50 µM concentration range. Moreover, ABT-888 promoted apoptosis in both the sensitive and resistant A375 cells, suggesting that ABT-888 may be useful in the treatment of BRAFi-resistant subsets of melanoma cells. Finally, in accordance with the involvement of PARP1 in actin cytoskeletal machinery, we found that the cytoskeletal organization, motility and invasive capability of both the A375 and A375R cells decreased upon exposure to 5 µM ABT-888 for 24 h. On the whole, the findings of this study highlight the pivotal role of PARP1 in the migration and invasion of melanoma cells, suggesting that ABT-888 may indeed be effective, not only as a pro-apoptotic drug for use in the treatment of BRAFi-resistant melanoma cells, but also in suppressing their migratory and invasive activities.
Collapse
Affiliation(s)
| | - Rosa Camerlingo
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
| | | | - Giuseppe Pirozzi
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, 07100 Sassari, Italy
| | - Giusy Gentilcore
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
- Division of Translational Medicine, Sidra Medical and Research Centre, 26999 Doha, Qatar
| | - Concetta Ragone
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
- Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', 81100 Caserta
| | - Michele Minopoli
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
| | | | - Maria Letizia Motti
- Istituto Nazionale Tumori -IRCCS- 'Fondazione G. Pascale', 80131 Naples
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, 07100 Sassari, Italy
- Division of Translational Medicine, Sidra Medical and Research Centre, 26999 Doha, Qatar
- Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', 81100 Caserta
- Department of Sport Science and Wellness, University 'Parthenope', 80133 Naples, Italy
| |
Collapse
|
9
|
Wouters J, Vizoso M, Martinez-Cardus A, Carmona FJ, Govaere O, Laguna T, Joseph J, Dynoodt P, Aura C, Foth M, Cloots R, van den Hurk K, Balint B, Murphy IG, McDermott EW, Sheahan K, Jirström K, Nodin B, Mallya-Udupi G, van den Oord JJ, Gallagher WM, Esteller M. Comprehensive DNA methylation study identifies novel progression-related and prognostic markers for cutaneous melanoma. BMC Med 2017; 15:101. [PMID: 28578692 PMCID: PMC5458482 DOI: 10.1186/s12916-017-0851-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/03/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Cutaneous melanoma is the deadliest skin cancer, with an increasing incidence and mortality rate. Currently, staging of patients with primary melanoma is performed using histological biomarkers such as tumor thickness and ulceration. As disruption of the epigenomic landscape is recognized as a widespread feature inherent in tumor development and progression, we aimed to identify novel biomarkers providing additional clinical information over current factors using unbiased genome-wide DNA methylation analyses. METHODS We performed a comprehensive DNA methylation analysis during all progression stages of melanoma using Infinium HumanMethylation450 BeadChips on a discovery cohort of benign nevi (n = 14) and malignant melanoma from both primary (n = 33) and metastatic (n = 28) sites, integrating the DNA methylome with gene expression data. We validated the discovered biomarkers in three independent validation cohorts by pyrosequencing and immunohistochemistry. RESULTS We identified and validated biomarkers for, and pathways involved in, melanoma development (e.g., HOXA9 DNA methylation) and tumor progression (e.g., TBC1D16 DNA methylation). In addition, we determined a prognostic signature with potential clinical applicability and validated PON3 DNA methylation and OVOL1 protein expression as biomarkers with prognostic information independent of tumor thickness and ulceration. CONCLUSIONS Our data underscores the importance of epigenomic regulation in triggering metastatic dissemination through the inactivation of central cancer-related pathways. Inactivation of cell-adhesion and differentiation unleashes dissemination, and subsequent activation of inflammatory and immune system programs impairs anti-tumoral defense pathways. Moreover, we identify several markers of tumor development and progression previously unrelated to melanoma, and determined a prognostic signature with potential clinical utility.
Collapse
Affiliation(s)
- Jasper Wouters
- Translational Cell and Tissue Research, KU Leuven (University of Leuven), Leuven, Belgium
- OncoMark Ltd, NovaUCD, Dublin 4, Ireland
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven (University of Leuven), Leuven, Belgium
| | - Miguel Vizoso
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Anna Martinez-Cardus
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - F Javier Carmona
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Olivier Govaere
- Translational Cell and Tissue Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Teresa Laguna
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
- Institute of Molecular Biology (IMB), Mainz, Germany
| | | | | | - Claudia Aura
- Translational Cell and Tissue Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Mona Foth
- OncoMark Ltd, NovaUCD, Dublin 4, Ireland
- Cancer Research UK, Beatson Institute, Glasgow, G61 1BD, UK
| | - Roy Cloots
- OncoMark Ltd, NovaUCD, Dublin 4, Ireland
- Department of Pathology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Karin van den Hurk
- OncoMark Ltd, NovaUCD, Dublin 4, Ireland
- Department of Pathology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Balazs Balint
- OncoMark Ltd, NovaUCD, Dublin 4, Ireland
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Ian G Murphy
- Department of Surgery, St. Vincent's University Hospital, Dublin 4, Ireland
| | - Enda W McDermott
- Department of Surgery, St. Vincent's University Hospital, Dublin 4, Ireland
| | - Kieran Sheahan
- Department of Pathology and Laboratory Medicine, St. Vincent's University Hospital, Dublin 4, Ireland
| | - Karin Jirström
- Department of Clinical Sciences, Division of Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | - Bjorn Nodin
- Department of Clinical Sciences, Division of Pathology, Lund University, Skåne University Hospital, 221 85, Lund, Sweden
| | | | - Joost J van den Oord
- Translational Cell and Tissue Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - William M Gallagher
- OncoMark Ltd, NovaUCD, Dublin 4, Ireland.
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.
- Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain.
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain.
| |
Collapse
|
10
|
PARP1 enhances lung adenocarcinoma metastasis by novel mechanisms independent of DNA repair. Oncogene 2016; 35:4569-79. [DOI: 10.1038/onc.2016.3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 12/28/2015] [Accepted: 12/29/2015] [Indexed: 12/31/2022]
|
11
|
Mao H, Lockyer P, Townley-Tilson WHD, Xie L, Pi X. LRP1 Regulates Retinal Angiogenesis by Inhibiting PARP-1 Activity and Endothelial Cell Proliferation. Arterioscler Thromb Vasc Biol 2015; 36:350-60. [PMID: 26634655 DOI: 10.1161/atvbaha.115.306713] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/15/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE We recently demonstrated that low-density lipoprotein receptor-related protein 1 (LRP1) is required for cardiovascular development in zebrafish. However, what role LRP1 plays in angiogenesis remains to be determined. To better understand the role of LRP1 in endothelial cell function, we investigated how LRP1 regulates mouse retinal angiogenesis. APPROACH AND RESULTS Depletion of LRP1 in endothelial cells results in increased retinal neovascularization in a mouse model of oxygen-induced retinopathy. Specifically, retinas in mice lacking endothelial LRP1 have more branching points and angiogenic sprouts at the leading edge of the newly formed vasculature. Increased endothelial proliferation as detected by Ki67 staining was observed in LRP1-deleted retinal endothelium in response to hypoxia. Using an array of biochemical and cell biology approaches, we demonstrate that poly(ADP-ribose) polymerase-1 (PARP-1) directly interacts with LRP1 in human retinal microvascular endothelial cells. This interaction between LRP1 and PARP-1 decreases under hypoxic condition. Moreover, LRP1 knockdown results in increased PARP-1 activity and subsequent phosphorylation of both retinoblastoma protein and cyclin-dependent kinase 2, which function to promote cell cycle progression and angiogenesis. CONCLUSIONS Together, these data reveal a pivotal role for LRP1 in endothelial cell proliferation and retinal neovascularization induced by hypoxia. In addition, we demonstrate for the first time the interaction between LRP1 and PARP-1 and the LRP1-dependent regulation of PARP-1-signaling pathways. These data bring forth the possibility of novel therapeutic approaches for pathological angiogenesis.
Collapse
Affiliation(s)
- Hua Mao
- From the Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (H.M., W.H.D.T.-T., L.X., X.P.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.)
| | - Pamela Lockyer
- From the Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (H.M., W.H.D.T.-T., L.X., X.P.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.)
| | - W H Davin Townley-Tilson
- From the Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (H.M., W.H.D.T.-T., L.X., X.P.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.)
| | - Liang Xie
- From the Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (H.M., W.H.D.T.-T., L.X., X.P.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.)
| | - Xinchun Pi
- From the Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (H.M., W.H.D.T.-T., L.X., X.P.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (P.L.).
| |
Collapse
|
12
|
Park SH, Noh SJ, Kim KM, Bae JS, Kwon KS, Jung SH, Kim JR, Lee H, Chung MJ, Moon WS, Kang MJ, Jang KY. Expression of DNA Damage Response Molecules PARP1, γH2AX, BRCA1, and BRCA2 Predicts Poor Survival of Breast Carcinoma Patients. Transl Oncol 2015; 8:239-49. [PMID: 26310369 PMCID: PMC4562981 DOI: 10.1016/j.tranon.2015.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/18/2015] [Accepted: 04/24/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: Poly(ADP-ribose) polymerase 1 (PARP1), γH2AX, BRCA1, and BRCA2 are conventional molecular indicators of DNA damage in cells and are often overexpressed in various cancers. In this study, we aimed, using immunohistochemical detection, whether the co-expression of PARP1, γH2AX, BRCA1, and BRCA2 in breast carcinoma (BCA) tissue can provide more reliable prediction of survival of BCA patients. MATERIALS AND METHODS: We investigated immunohistochemical expression and prognostic significance of the expression of PARP1, γH2AX, BRCA1, and BRCA2 in 192 cases of BCAs. RESULTS: The expression of these four molecules predicted earlier distant metastatic relapse, shorter overall survival (OS), and relapse-free survival (RFS) by univariate analysis. Multivariate analysis revealed the expression of PARP1, γH2AX, and BRCA2 as independent poor prognostic indicators of OS and RFS. In addition, the combined expressional pattern of BRCA1, BRCA2, PARP1, and γH2AX (CSbbph) was an additional independent prognostic predictor for OS (P < .001) and RFS (P < .001). The 10-year OS rate was 95% in the CSbbph-low (CSbbph scores 0 and 1) subgroup, but that was only 35% in the CSbbph-high (CSbbph score 4) subgroup. CONCLUSION: This study has demonstrated that the individual and combined expression patterns of PARP1, γH2AX, BRCA1, and BRCA2 could be helpful in determining an accurate prognosis for BCA patients and for the selection of BCA patients who could potentially benefit from anti-PARP1 therapy with a combination of genotoxic chemotherapeutic agents.
Collapse
Affiliation(s)
- See-Hyoung Park
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sang Jae Noh
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Kyoung Min Kim
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Jun Sang Bae
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Keun Sang Kwon
- Department of Preventive Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Sung Hoo Jung
- Department of Surgery, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Jung Ryul Kim
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Ho Lee
- Department of Forensic Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Myoung Ja Chung
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Woo Sung Moon
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Myoung Jae Kang
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, Jeonju, Republic of Korea.
| |
Collapse
|
13
|
Yélamos J, Galindo M, Navarro J, Albanell J, Rovira A, Rojo F, Oliver J. Enhancing tumor-targeting monoclonal antibodies therapy by PARP inhibitors. Oncoimmunology 2015; 5:e1065370. [PMID: 26942084 DOI: 10.1080/2162402x.2015.1065370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become a successful therapeutic approach in cancer. However, some patients do not achieve long-term clinical benefit and most mAbs only exert modest effects as monotherapies. Therefore, combinations with chemotherapy are currently being investigated. Emerging studies have shown a synergistic therapeutic effect of PARP inhibitors and mAbs in cancer. PARP enzymes catalytically cleave β-NAD+ and transfer the ADP-ribose moiety to acceptor proteins, modifying their function. In here, we update recent data about the therapeutic effect of the combination of PARP inhibitors with mAbs in cancer treatment and discuss the molecular mechanisms involved in this synergy.
Collapse
Affiliation(s)
- José Yélamos
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Immunology, Hospital del Mar, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Miguel Galindo
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM) , Barcelona, Spain
| | - Judith Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM) , Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Oncology, Hospital del Mar, Barcelona, Spain
| | - Ana Rovira
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Oncology, Hospital del Mar, Barcelona, Spain
| | - Federico Rojo
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Patology, IIS Fundación Jiménez Díaz, Madrid, Spain
| | - Javier Oliver
- Department of Cell Biology and Immunology; Instituto de Parasitología y Biomedicina López Neyra , Granada, Spain
| |
Collapse
|
14
|
Sistigu A, Manic G, Obrist F, Vitale I. Trial watch - inhibiting PARP enzymes for anticancer therapy. Mol Cell Oncol 2015; 3:e1053594. [PMID: 27308587 DOI: 10.1080/23723556.2015.1053594] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/16/2015] [Accepted: 05/18/2015] [Indexed: 12/25/2022]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are a members of family of enzymes that catalyze poly(ADP-ribosyl)ation (PARylation) and/or mono(ADP-ribosyl)ation (MARylation), two post-translational protein modifications involved in crucial cellular processes including (but not limited to) the DNA damage response (DDR). PARP1, the most abundant family member, is a nuclear protein that is activated upon sensing distinct types of DNA damage and contributes to their resolution by PARylating multiple DDR players. Recent evidence suggests that, along with DDR, activated PARP1 mediates a series of prosurvival and proapoptotic processes aimed at preserving genomic stability. Despite this potential oncosuppressive role, upregulation and/or overactivation of PARP1 or other PARP enzymes has been reported in a variety of human neoplasms. Over the last few decades, several pharmacologic inhibitors of PARP1 and PARP2 have been assessed in preclinical and clinical studies showing potent antineoplastic activity, particularly against homologous recombination (HR)-deficient ovarian and breast cancers. In this Trial Watch, we describe the impact of PARP enzymes and PARylation in cancer, discuss the mechanism of cancer cell killing by PARP1 inactivation, and summarize the results of recent clinical studies aimed at evaluating the safety and therapeutic profile of PARP inhibitors in cancer patients.
Collapse
Affiliation(s)
| | - Gwenola Manic
- Regina Elena National Cancer Institute , Rome, Italy
| | - Florine Obrist
- Université Paris-Sud/Paris XI, Le Kremlin-Bicêtre, France; INSERM, UMRS1138, Paris, France; Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, Rome, Italy; Department of Biology, University of Rome "TorVergata", Rome, Italy
| |
Collapse
|
15
|
Motta C, D'Angeli F, Scalia M, Satriano C, Barbagallo D, Naletova I, Anfuso CD, Lupo G, Spina-Purrello V. PJ-34 inhibits PARP-1 expression and ERK phosphorylation in glioma-conditioned brain microvascular endothelial cells. Eur J Pharmacol 2015; 761:55-64. [PMID: 25934569 DOI: 10.1016/j.ejphar.2015.04.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 04/17/2015] [Accepted: 04/20/2015] [Indexed: 12/19/2022]
Abstract
Inhibitors of PARP-1(Poly(ADP-ribose) polymerase-1) act by competing with NAD(+), the enzyme physiological substrate, which play a protective role in many pathological conditions characterized by PARP-1 overactivation. It has been shown that PARP-1 also promotes tumor growth and progression through its DNA repair activity. Since angiogenesis is an essential requirement for these activities, we sought to determine whether PARP inhibition might affect rat brain microvascular endothelial cells (GP8.3) migration, stimulated by C6-glioma conditioned medium (CM). Through wound-healing experiments and MTT analysis, we demonstrated that PARP-1 inhibitor PJ-34 [N-(6-Oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide] abolishes the migratory response of GP8.3 cells and reduces their viability. PARP-1 also acts in a DNA independent way within the Extracellular-Regulated-Kinase (ERK) signaling cascade, which regulates cell proliferation and differentiation. By western analysis and confocal laser scanning microscopy (LSM), we analyzed the effects of PJ-34 on PARP-1 expression, phospho-ERK and phospho-Elk-1 activation. The effect of MEK (mitogen-activated-protein-kinase-kinase) inhibitor PD98059 (2-(2-Amino-3-methoxyphenyl)-4 H-1-benzopyran-4-one) on PARP-1 expression in unstimulated and in CM-stimulated GP8.3 cells was analyzed by RT-PCR. PARP-1 expression and phospho-ERK activation were significantly reduced by treatment of GP8.3 cells with PJ-34 or PD98059. By LSM, we further demonstrated that PARP-1 and phospho-ERK are coexpressed and share the same subcellular localization in GP8.3 cells, in the cytoplasm as well as in nucleoplasm. Based on these data, we propose that PARP-1 and phospho-ERK interact in the cytosol and then translocate to the nucleus, where they trigger a proliferative response. We also propose that PARP-1 inhibition blocks CM-induced endothelial migration by interfering with ERK signal-transduction pathway.
Collapse
Affiliation(s)
- Carla Motta
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Floriana D'Angeli
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Marina Scalia
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Cristina Satriano
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Davide Barbagallo
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Irina Naletova
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Carmelina Daniela Anfuso
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Gabriella Lupo
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy
| | - Vittoria Spina-Purrello
- Department of Biomedical Sciences and Biotecnology, University of Catania, Via Santa Sofia 64, 95125 Catania, Italy.
| |
Collapse
|
16
|
Graziani G, Artuso S, De Luca A, Muzi A, Rotili D, Scimeca M, Atzori MG, Ceci C, Mai A, Leonetti C, Levati L, Bonanno E, Tentori L, Caccuri AM. A new water soluble MAPK activator exerts antitumor activity in melanoma cells resistant to the BRAF inhibitor vemurafenib. Biochem Pharmacol 2015; 95:16-27. [PMID: 25795251 DOI: 10.1016/j.bcp.2015.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/10/2015] [Indexed: 12/01/2022]
Abstract
Recovery of mitogen activated protein kinase (MAPK) or activation of alternative pathways, such as the PI3K/AKT/mTOR, are involved in acquired resistance to BRAF inhibitors which represent the first-line treatment of BRAF-mutated metastatic melanoma. We recently demonstrated that 6-((7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)thio)hexan-1-ol (NBDHEX) and its water soluble analog 2-(2-(2-((7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)thio)ethoxy)ethoxy)ethanol (MC3181) trigger apoptosis in BRAF V600E mutated melanoma cells through activation of the MAPK c-Jun N-terminal kinase (JNK). Herein, we investigated whether NBDHEX and MC3181 might exert antitumor activity against BRAF V600E mutated human melanoma cells rendered resistant to the BRAF inhibitor vemurafenib. To this aim we generated a subline of A375 melanoma resistant in vitro and in vivo to vemurafenib (A375-VR8) and characterized by NRAS G13R mutation, high basal levels of CRAF protein and phospho-activation of AKT. In these cells ERK phosphorylation was not significantly down-modulated by vemurafenib concentrations capable of abrogating ERK phosphorylation in sensitive A375 cells. Both NBDHEX and MC3181 induced marked antiproliferative and apoptotic effects in A375-VR8 cells and, at equitoxic concentrations, caused a strong phosphorylation of JNK, p38, and of the downstream mediators of apoptosis ATF2 and p53. Drug treatment further increased ERK phosphorylation, which was required for the cellular response to the NBD derivatives, as apoptosis was antagonized by the ERK inhibitor FR180204. Finally, in vivo administration of MC3181 provoked JNK activation at the tumor site and markedly reduced A375-VR8 growth. These evidences strongly suggest that the activation of multiple pro-apoptotic MAPK pathways by MC3181 might represent a new strategy for the treatment of melanoma resistant to BRAF inhibitors.
Collapse
Affiliation(s)
- Grazia Graziani
- Department of Systems Medicine, University of "Tor Vergata", Rome, Italy
| | - Simona Artuso
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Anastasia De Luca
- The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of "Tor Vergata", Rome, Italy
| | - Alessia Muzi
- Department of Systems Medicine, University of "Tor Vergata", Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, "Sapienza" University, Rome, Italy
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, University of "Tor Vergata", Rome, Italy; TMALab s.r.l., Spin-off of University of "Tor Vergata", Rome, Italy
| | | | - Claudia Ceci
- Department of Systems Medicine, University of "Tor Vergata", Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, "Sapienza" University, Rome, Italy; Pasteur Institute, Cenci-Bolognetti Foundation, "Sapienza" University, Rome, Italy
| | - Carlo Leonetti
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Lauretta Levati
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata" - IRCCS, Rome, Italy
| | - Elena Bonanno
- Department of Biomedicine and Prevention, University of "Tor Vergata", Rome, Italy; TMALab s.r.l., Spin-off of University of "Tor Vergata", Rome, Italy
| | - Lucio Tentori
- Department of Systems Medicine, University of "Tor Vergata", Rome, Italy
| | - Anna Maria Caccuri
- The NAST Centre for Nanoscience & Nanotechnology & Innovative Instrumentation, University of "Tor Vergata", Rome, Italy; Department of Experimental Medicine and Surgery, University of "Tor Vergata", Rome, Italy.
| |
Collapse
|
17
|
Rodríguez MI, Majuelos-Melguizo J, Martí Martín-Consuegra JM, Ruiz de Almodóvar M, López-Rivas A, Javier Oliver F. Deciphering the insights of poly(ADP-ribosylation) in tumor progression. Med Res Rev 2015; 35:678-97. [PMID: 25604534 DOI: 10.1002/med.21339] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors are particularly efficient against tumors with defects in the homologous recombination repair pathway. Nonetheless poly(ADP-ribosylation) (PARylation) modulates prometastasic activities and adaptation of tumor to a hostile microenvironment. Modulation of metastasis-promoting traits is possible through the alteration of key transcription factors involved in the regulation of the hypoxic response, the recruitment of new vessels (or angiogenesis), and the stimulation of epithelial to mesenchymal transition (EMT). In this review, we summarized some of the findings that focalize on PARP-1's action on tumor aggressiveness, suggesting new therapeutic opportunities against an assembly of tumors not necessarily bearing DNA repair defects. Metastasis accounts for the vast majority of mortality derived from solid cancer. PARP-1 is an active player in tumor adaptation to metastasis and PARP inhibitors, recognized as promising therapeutic agents against homologous recombination deficient tumors, has novel properties responsible for the antimetastatic actions in different tumor settings.
Collapse
Affiliation(s)
- María Isabel Rodríguez
- Instituto de Parasitología y Biomedicina López Neyra (IPBLN), CSIC, Granada, Spain, 18016
| | - Jara Majuelos-Melguizo
- Instituto de Parasitología y Biomedicina López Neyra (IPBLN), CSIC, Granada, Spain, 18016
| | | | | | - Abelardo López-Rivas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Consejo Superior de Investigaciones Científicas, Sevilla, Spain, 41092
| | | |
Collapse
|
18
|
Mactier S, Kaufman KL, Wang P, Crossett B, Pupo GM, Kohnke PL, Thompson JF, Scolyer RA, Yang JY, Mann GJ, Christopherson RI. Protein signatures correspond to survival outcomes of AJCC stage III melanoma patients. Pigment Cell Melanoma Res 2014; 27:1106-16. [PMID: 24995518 PMCID: PMC4285183 DOI: 10.1111/pcmr.12290] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 07/02/2014] [Indexed: 11/28/2022]
Abstract
Summary Outcomes for melanoma patients with stage III disease differ widely even within the same subcategory. Molecular signatures that more accurately predict prognosis are needed to stratify patients according to risk. Proteomic analyses were used to identify differentially abundant proteins in extracts of surgically excised samples from patients with stage IIIc melanoma lymph node metastases. Analysis of samples from patients with poor (n = 14, <1 yr) and good (n = 19, >4 yr) survival outcomes identified 84 proteins that were differentially abundant between prognostic groups. Subsequent selected reaction monitoring analysis verified 21 proteins as potential biomarkers for survival. Poor prognosis patients are characterized by increased levels of proteins involved in protein metabolism, nucleic acid metabolism, angiogenesis, deregulation of cellular energetics and methylation processes, and decreased levels of proteins involved in apoptosis and immune response. These proteins are able to classify stage IIIc patients into prognostic subgroups (P < 0.02). This is the first report of potential prognostic markers from stage III melanoma using proteomic analyses. Validation of these protein markers in larger patient cohorts should define protein signatures that enable better stratification of stage III melanoma patients.
Collapse
Affiliation(s)
- Swetlana Mactier
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lupo B, Trusolino L. Inhibition of poly(ADP-ribosyl)ation in cancer: old and new paradigms revisited. Biochim Biophys Acta Rev Cancer 2014; 1846:201-15. [PMID: 25026313 DOI: 10.1016/j.bbcan.2014.07.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/02/2014] [Accepted: 07/08/2014] [Indexed: 01/31/2023]
Abstract
Inhibitors of poly(ADP-ribose) polymerases actualized the biological concept of synthetic lethality in the clinical practice, yielding a paradigmatic example of translational medicine. The profound sensitivity of tumors with germline BRCA mutations to PARP1/2 blockade owes to inherent defects of the BRCA-dependent homologous recombination machinery, which are unleashed by interruption of PARP DNA repair activity and lead to DNA damage overload and cell death. Conversely, aspirant BRCA-like tumors harboring somatic DNA repair dysfunctions (a vast entity of genetic and epigenetic defects known as "BRCAness") not always align with the familial counterpart and appear not to be equally sensitive to PARP inhibition. The acquisition of secondary resistance in initially responsive patients and the lack of standardized biomarkers to identify "BRCAness" pose serious threats to the clinical advance of PARP inhibitors; a feeling is also emerging that a BRCA-centered perspective might have missed the influence of additional, not negligible and DNA repair-independent PARP contributions onto therapy outcome. While regulatory approval for PARP1/2 inhibitors is still pending, novel therapeutic opportunities are sprouting from different branches of the PARP family, although they remain immature for clinical extrapolation. This review is an endeavor to provide a comprehensive appraisal of the multifaceted biology of PARPs and their evolving impact on cancer therapeutics.
Collapse
Affiliation(s)
- Barbara Lupo
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Torino, Italy; Laboratory of Molecular Pharmacology, Candiolo Cancer Institute, FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Torino, Italy; Laboratory of Molecular Pharmacology, Candiolo Cancer Institute, FPO IRCCS, 10060 Candiolo, Torino, Italy.
| |
Collapse
|
20
|
Weaver AN, Yang ES. Beyond DNA Repair: Additional Functions of PARP-1 in Cancer. Front Oncol 2013; 3:290. [PMID: 24350055 PMCID: PMC3841914 DOI: 10.3389/fonc.2013.00290] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 11/13/2013] [Indexed: 12/18/2022] Open
Abstract
Poly(ADP-ribose) polymerases (PARPs) are DNA-dependent nuclear enzymes that transfer negatively charged ADP-ribose moieties from cellular nicotinamide-adenine-dinucleotide (NAD(+)) to a variety of protein substrates, altering protein-protein and protein-DNA interactions. The most studied of these enzymes is poly(ADP-ribose) polymerase-1 (PARP-1), which is an excellent therapeutic target in cancer due to its pivotal role in the DNA damage response. Clinical studies have shown susceptibility to PARP inhibitors in DNA repair defective cancers with only mild adverse side effects. Interestingly, additional studies are emerging which demonstrate a role for this therapy in DNA repair proficient tumors through a variety of mechanisms. In this review, we will discuss additional functions of PARP-1 - including regulation of inflammatory mediators, cellular energetics and death pathways, gene transcription, sex hormone- and ERK-mediated signaling, and mitosis - and the role these PARP-1-mediated processes play in oncogenesis, cancer progression, and the development of therapeutic resistance. As PARP-1 can act in both a pro- and anti-tumor manner depending on the context, it is important to consider the global effects of this protein in determining when, and how, to best use PARP inhibitors in anticancer therapy.
Collapse
Affiliation(s)
- Alice N. Weaver
- Department of Radiation Oncology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eddy S. Yang
- Department of Radiation Oncology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pharmacology and Toxicology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
21
|
Shah GM, Robu M, Purohit NK, Rajawat J, Tentori L, Graziani G. PARP Inhibitors in Cancer Therapy: Magic Bullets but Moving Targets. Front Oncol 2013; 3:279. [PMID: 24294592 PMCID: PMC3827545 DOI: 10.3389/fonc.2013.00279] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 10/29/2013] [Indexed: 02/03/2023] Open
Affiliation(s)
- Girish M Shah
- Laboratory for Skin Cancer Research, CHU-Q (CHUL) Research Centre, Laval University , Quebec City, QC , Canada
| | | | | | | | | | | |
Collapse
|
22
|
Rodríguez MI, Peralta-Leal A, O'Valle F, Rodriguez-Vargas JM, Gonzalez-Flores A, Majuelos-Melguizo J, López L, Serrano S, de Herreros AG, Rodríguez-Manzaneque JC, Fernández R, del Moral RG, de Almodóvar JM, Oliver FJ. PARP-1 regulates metastatic melanoma through modulation of vimentin-induced malignant transformation. PLoS Genet 2013; 9:e1003531. [PMID: 23785295 PMCID: PMC3681683 DOI: 10.1371/journal.pgen.1003531] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 04/12/2013] [Indexed: 12/30/2022] Open
Abstract
PARP inhibition can induce anti-neoplastic effects when used as monotherapy or in combination with chemo- or radiotherapy in various tumor settings; however, the basis for the anti-metastasic activities resulting from PARP inhibition remains unknown. PARP inhibitors may also act as modulators of tumor angiogenesis. Proteomic analysis of endothelial cells revealed that vimentin, an intermediary filament involved in angiogenesis and a specific hallmark of EndoMT (endothelial to mesenchymal transition) transformation, was down-regulated following loss of PARP-1 function in endothelial cells. VE-cadherin, an endothelial marker of vascular normalization, was up-regulated in HUVEC treated with PARP inhibitors or following PARP-1 silencing; vimentin over-expression was sufficient to drive to an EndoMT phenotype. In melanoma cells, PARP inhibition reduced pro-metastatic markers, including vasculogenic mimicry. We also demonstrated that vimentin expression was sufficient to induce increased mesenchymal/pro-metastasic phenotypic changes in melanoma cells, including ILK/GSK3-β-dependent E-cadherin down-regulation, Snail1 activation and increased cell motility and migration. In a murine model of metastatic melanoma, PARP inhibition counteracted the ability of melanoma cells to metastasize to the lung. These results suggest that inhibition of PARP interferes with key metastasis-promoting processes, leading to suppression of invasion and colonization of distal organs by aggressive metastatic cells. Metastasis is the spread of malignant tumor cells from their original site to other parts of the body and is responsible for the vast majority of solid cancer-related mortality. PARP inhibitors are emerging as promising anticancer therapeutics and are currently undergoing clinical trials. It is therefore important to elucidate the mechanisms underlying the anti-tumor actions of these drugs. In our current study, we elucidated novel anti-neoplastic properties of PARP inhibitors that are responsible for the anti-metastatic effect of these drugs in the context of malignant melanoma. These effects appear to be the result of PARP-1's ability to regulate the expression of key factors, such as vimentin and VE-cadherin, involved in vascular cell dynamics and to limit pro-malignant processes such as vasculogenic mimicry and EMT.
Collapse
Affiliation(s)
- María Isabel Rodríguez
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
- IBIMER, Centro de Investigaciones Biomédicas, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular I, Universidad de Granada, Granada, Spain
- * E-mail: (MIR); (FJO)
| | | | - Francisco O'Valle
- Departamento de Anatomía Patológica, Universidad de Granada, Granada, Spain
| | | | | | | | - Laura López
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
| | - Santiago Serrano
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
| | | | | | - Rubén Fernández
- Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Raimundo G. del Moral
- Unidad de Anatomía Patológica, Complejo Hospitalario y Áreas Sur y Noreste de Granada, Granada, Spain
| | | | - F. Javier Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
- * E-mail: (MIR); (FJO)
| |
Collapse
|
23
|
Masutani M, Fujimori H. Poly(ADP-ribosyl)ation in carcinogenesis. Mol Aspects Med 2013; 34:1202-16. [PMID: 23714734 DOI: 10.1016/j.mam.2013.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 05/14/2013] [Accepted: 05/19/2013] [Indexed: 12/18/2022]
Abstract
Cancer develops through diverse genetic, epigenetic and other changes, so-called 'multi-step carcinogenesis', and each cancer harbors different alterations and properties. Here in this article we review how poly(ADP-ribosyl)ation is involved in multi-step and diverse pathways of carcinogenesis. Involvement of poly- and mono-ADP-ribosylation in carcinogenesis has been studied at molecular and cellular levels, and further by animal models and human genetic approaches. PolyADP-ribosylation acts in DNA damage repair response and maintenance mechanisms of genomic stability. Several DNA repair pathways, including base-excision repair and double strand break repair pathways, involve PARP and PARG functions. These care-taker functions of poly(ADP-ribosyl)ation suggest that polyADP-ribosyation may mainly act in a tumor suppressive manner because genomic instability caused by defective DNA repair response could serve as a driving force for tumor progression, leading to invasion, metastasis and relapse of cancer. On the other hand, the new concept of 'synthetic lethality by PARP inhibition' suggests the significance of PARP activities for survival of cancer cells that harbor defects in DNA repair. Accumulating evidence has revealed that some PARP family molecules are involved in various signaling cascades other than DNA repair, including epigenetic and transcriptional regulations, inflammation/immune response and epithelial-mesenchymal transition, suggesting that poly(ADP-ribosyl)ation both promotes and suppresses carcinogenic processes depending on the conditions. Expanding understanding of poly(ADP-ribosyl)ation suggests that strategies to achieve cancer prevention targeting poly(ADP-ribosyl)ation for genome protection against life-long exposure to environmental carcinogens and endogenous carcinogenic stimuli.
Collapse
Affiliation(s)
- Mitsuko Masutani
- Division of Genome Stability Research, National Cancer Center Research Institute, Japan.
| | | |
Collapse
|
24
|
Peña-Chilet M, Blanquer-Maceiras M, Ibarrola-Villava M, Martinez-Cadenas C, Martin-Gonzalez M, Gomez-Fernandez C, Mayor M, Aviles JA, Lluch A, Ribas G. Genetic variants in PARP1 (rs3219090) and IRF4 (rs12203592) genes associated with melanoma susceptibility in a Spanish population. BMC Cancer 2013; 13:160. [PMID: 23537197 PMCID: PMC3704782 DOI: 10.1186/1471-2407-13-160] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 03/20/2013] [Indexed: 12/24/2022] Open
Abstract
Background Few high penetrance genes are known in Malignant Melanoma (MM), however, the involvement of low-penetrance genes such as MC1R, OCA2, ASIP, SLC45A2 and TYR has been observed. Lately, genome-wide association studies (GWAS) have been the ideal strategy to identify new common, low-penetrance susceptibility loci. In this case–control study, we try to validate in our population nine melanoma associated markers selected from published GWAS in melanoma predisposition. Methods We genotyped the 9 markers corresponding to 8 genes (PARP1, MX2, ATM, CCND1, NADSYN1, CASP8, IRF4 and CYP2R1) in 566 cases and 347 controls from a Spanish population using KASPar probes. Genotypes were analyzed by logistic regression and adjusted by phenotypic characteristics. Results We confirm the protective role in MM of the rs3219090 located on the PARP1 gene (p-value 0.027). Additionally, this SNP was also associated with eye color (p-value 0.002). A second polymorphism, rs12203592, located on the IRF4 gene was associated with protection to develop MM for the dominant model (p-value 0.037). We have also observed an association of this SNP with both lentigines (p-value 0.014) and light eye color (p-value 3.76 × 10-4). Furthermore, we detected a novel association with rs1485993, located on the CCND1 gene, and dark eye color (p-value 4.96 × 10-4). Finally, rs1801516, located on the ATM gene, showed a trend towards a protective role in MM similar to the one firstly described in a GWAS study. Conclusions To our knowledge, this is the first time that these SNPs have been associated with MM in a Spanish population. We confirmed the proposed role of rs3219090, located on the PARP1 gene, and rs12203592, located on the IRF4 gene, as protective to MM along the same lines as have previous genome-wide associated works. Finally, we have seen associations between IRF4, PARP1, and CCND1 and phenotypic characteristics, confirming previous results for the IRF4 gene and presenting novel data for the last two, suggesting that pigmentation characteristics correlated with eye color are potential mediators between PARP1 and MM protection.
Collapse
|
25
|
|
26
|
van der Weyden L, Adams DJ. Using mice to unveil the genetics of cancer resistance. Biochim Biophys Acta Rev Cancer 2012; 1826:312-30. [PMID: 22613679 DOI: 10.1016/j.bbcan.2012.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/10/2012] [Accepted: 05/13/2012] [Indexed: 11/28/2022]
Abstract
In the UK, four in ten people will develop some form of cancer during their lifetime, with an individual's relative risk depending on many factors, including age, lifestyle and genetic make-up. Much research has gone into identifying the genes that are mutated in tumorigenesis with the overwhelming majority of genetically-modified (GM) mice in cancer research showing accelerated tumorigenesis or recapitulating key aspects of the tumorigenic process. Yet if six out of ten people will not develop some form of cancer during their lifetime, together with the fact that some cancer patients experience spontaneous regression/remission, it suggests there are ways of 'resisting' cancer. Indeed, there are wildtype, spontaneously-arising mutants and GM mice that show some form of 'resistance' to cancer. Identification of mice with increased resistance to cancer is a novel aspect of cancer research that is important in terms of providing both chemopreventative and therapeutic options. In this review we describe the different mouse lines that display a 'cancer resistance' phenotype and discuss the molecular basis of their resistance.
Collapse
Affiliation(s)
- Louise van der Weyden
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.
| | | |
Collapse
|
27
|
Rojo F, García-Parra J, Zazo S, Tusquets I, Ferrer-Lozano J, Menendez S, Eroles P, Chamizo C, Servitja S, Ramírez-Merino N, Lobo F, Bellosillo B, Corominas JM, Yelamos J, Serrano S, Lluch A, Rovira A, Albanell J. Nuclear PARP-1 protein overexpression is associated with poor overall survival in early breast cancer. Ann Oncol 2012; 23:1156-1164. [PMID: 21908496 DOI: 10.1093/annonc/mdr361] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Poly(ADP-ribose)polymerase-1 (PARP-1) is a highly promising novel target in breast cancer. However, the expression of PARP-1 protein in breast cancer and its associations with outcome are yet poorly characterized. PATIENTS AND METHODS Quantitative expression of PARP-1 protein was assayed by a specific immunohistochemical signal intensity scanning assay in a range of normal to malignant breast lesions, including a series of patients (N = 330) with operable breast cancer to correlate with clinicopathological factors and long-term outcome. RESULTS PARP-1 was overexpressed in about a third of ductal carcinoma in situ and infiltrating breast carcinomas. PARP-1 protein overexpression was associated to higher tumor grade (P = 0.01), estrogen-negative tumors (P < 0.001) and triple-negative phenotype (P < 0.001). The hazard ratio (HR) for death in patients with PARP-1 overexpressing tumors was 7.24 (95% CI; 3.56-14.75). In a multivariate analysis, PARP-1 overexpression was an independent prognostic factor for both disease-free (HR 10.05; 95% CI 5.42-10.66) and overall survival (HR 1.82; 95% CI 1.32-2.52). CONCLUSIONS Nuclear PARP-1 is overexpressed during the malignant transformation of the breast, particularly in triple-negative tumors, and independently predicts poor prognosis in operable invasive breast cancer.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/diagnosis
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Cell Nucleus/metabolism
- Cell Nucleus/pathology
- Cells, Cultured
- Disease Progression
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mice
- Mice, Knockout
- Middle Aged
- Neoplasm Invasiveness
- Neoplasm Staging
- Poly (ADP-Ribose) Polymerase-1
- Poly(ADP-ribose) Polymerase Inhibitors
- Poly(ADP-ribose) Polymerases/genetics
- Poly(ADP-ribose) Polymerases/metabolism
- Prognosis
- RNA, Small Interfering/pharmacology
- Survival Analysis
- Up-Regulation/drug effects
- Up-Regulation/genetics
Collapse
Affiliation(s)
- F Rojo
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona; Department of Pathology, Hospital del Mar, Barcelona; Department of Pathology, IIS-Fundación Jiménez Díaz, Madrid
| | - J García-Parra
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona; Medical Oncology Department, Hospital del Mar, Barcelona
| | - S Zazo
- Department of Pathology, IIS-Fundación Jiménez Díaz, Madrid
| | - I Tusquets
- Medical Oncology Department, Hospital del Mar, Barcelona
| | - J Ferrer-Lozano
- Department of Oncology and Hematology, Hospital Clinico Universitario, Valencia
| | - S Menendez
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona; Medical Oncology Department, Hospital del Mar, Barcelona
| | - P Eroles
- Department of Oncology and Hematology, Hospital Clinico Universitario, Valencia
| | - C Chamizo
- Department of Pathology, IIS-Fundación Jiménez Díaz, Madrid
| | - S Servitja
- Medical Oncology Department, Hospital del Mar, Barcelona
| | | | - F Lobo
- Department of Oncology, IIS-Fundación Jiménez Díaz, Madrid
| | - B Bellosillo
- Department of Pathology, Hospital del Mar, Barcelona
| | - J M Corominas
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona; Department of Pathology, Hospital del Mar, Barcelona; Department of Medicine, Autonomous University of Barcelona, Barcelona
| | - J Yelamos
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona; Immunology Department, Hospital del Mar, Barcelona
| | - S Serrano
- Department of Pathology, Hospital del Mar, Barcelona; Department of Medicine, Autonomous University of Barcelona, Barcelona
| | - A Lluch
- Department of Oncology and Hematology, Hospital Clinico Universitario, Valencia; Department of Medicine, Valencia Central University, Valencia, Spain
| | - A Rovira
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona; Medical Oncology Department, Hospital del Mar, Barcelona
| | - J Albanell
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barcelona; Medical Oncology Department, Hospital del Mar, Barcelona; Department of Medicine, Autonomous University of Barcelona, Barcelona.
| |
Collapse
|
28
|
Javle M, Curtin NJ. The potential for poly (ADP-ribose) polymerase inhibitors in cancer therapy. Ther Adv Med Oncol 2011; 3:257-67. [PMID: 22084640 DOI: 10.1177/1758834011417039] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The modulation of DNA repair pathways for therapeutic benefit in cancer has now become a reality with the development of poly (ADP-ribose) polymerase inhibitors (PARPi). PARP is involved in single-strand DNA breaks, which in the presence of defective homologous recombination repair lead to double-strand DNA breaks, the most lethal form of DNA damage. These agents therefore may be the drugs of choice for BRCA mutant breast and ovarian cancers. PARPi result in synergistic antitumor effects when combined with cisplatin, temozolomide, topoisomerase inhibitors and ionizing radiation. The indications for PARPi lie beyond BRCA mutations and may include genomic and functional defects in DNA repair and damage response pathways. Several PARPi are in the clinical development phase at this time and, given the recent failure of a phase III clinical trial of iniparib in triple-negative breast cancer, the identification of structural and functional differences between these inhibitors becomes critical. Acquired resistance to PARPi is being noted and represents an important limitation in this field. A concise review of the literature in this field is presented.
Collapse
Affiliation(s)
- M Javle
- Associate Professor, UT-MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
29
|
The glutathione transferase inhibitor 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio)hexanol (NBDHEX) increases temozolomide efficacy against malignant melanoma. Eur J Cancer 2011; 47:1219-30. [DOI: 10.1016/j.ejca.2010.12.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 12/01/2010] [Accepted: 12/14/2010] [Indexed: 11/23/2022]
|
30
|
Mangerich A, Bürkle A. How to kill tumor cells with inhibitors of poly(ADP-ribosyl)ation. Int J Cancer 2010; 128:251-65. [DOI: 10.1002/ijc.25683] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2010] [Accepted: 08/19/2010] [Indexed: 02/07/2023]
|
31
|
Pizem J, Popovic M, Cör A. Expression of Gli1 and PARP1 in medulloblastoma: an immunohistochemical study of 65 cases. J Neurooncol 2010; 103:459-67. [PMID: 20953661 DOI: 10.1007/s11060-010-0431-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 09/20/2010] [Indexed: 12/29/2022]
Abstract
Activation of the sonic hedgehog (SHH) signalling pathway, which is involved in the formation of a significant proportion of medulloblastomas, is characterised by up-regulation and nuclear localisation of downstream transcription factor Gli1. Our aim was to analyse Gli1 expression by immunohistochemistry in a large group of medulloblastomas, to assess possible correlations with WNT (wingless) pathway activation and poly(ADP-ribose) polymerase-1 (PARP1) expression, previously shown to be associated with SHH pathway activation in a mouse model of medulloblastoma. We analysed expression and localisation of Gli1, β-catenin and PARP1 by immunohistochemistry in a series of 65 consecutive medulloblastomas. Gli1 was positive in 40 (61.5%) medulloblastomas, as revealed by either strong (21 cases) or mild (19 cases) nuclear reaction in more than 50% of tumour cells. Nuclear positivity for PARP1 was noted in all 65 cases, ranging from 46% to 100% (mean 80%) but was not correlated with Gli1 positivity. Gli1 was positive in 9 of 11 cases with nuclear localisation of β-catenin, signifying concurrent activation of SHH and WNT pathways. Overall survival of patients with strong nuclear reaction to Gli1 was better compared with patients with Gli1-negative medulloblastomas. Immunohistochemical detection of Gli1 could be useful in identifying medulloblastomas with SHH pathway activation. As revealed by nuclear reaction to Gli1, the SHH pathway is activated in approximately 60% of medulloblastomas. In some medulloblastomas, both SHH and WNT appear to be activated. PARP1 is highly expressed in medulloblastomas. It might be useful as a target to increase the effectiveness of current treatment modalities.
Collapse
Affiliation(s)
- Joze Pizem
- Medical Faculty, Institute of Pathology, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | | | | |
Collapse
|
32
|
Nicolás L, Martínez C, Baró C, Rodríguez M, Baroja-Mazo A, Sole F, Flores JM, Ampurdanés C, Dantzer F, Martin-Caballero J, Aparicio P, Yelamos J. Loss of poly(ADP-ribose) polymerase-2 leads to rapid development of spontaneous T-cell lymphomas in p53-deficient mice. Oncogene 2010; 29:2877-83. [PMID: 20154718 DOI: 10.1038/onc.2010.11] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Poly(ADP-ribose) polymerase-2 (Parp-2) belongs to a family of enzymes that catalyse poly(ADP-ribosyl)ation of proteins. Parp-2 deficiency in mice (Parp-2(-/-)) results in reduced thymic cellularity associated with increased apoptosis in thymocytes, defining Parp-2 as an important mediator of T-cell survival during thymopoiesis. To determine whether there is a link between Parp-2 and the p53 DNA-damage-dependent apoptotic response, we have generated Parp-2/p53-double-null mutant mice. We found that p53(-/-) backgrounds completely restored the survival and development of Parp-2(-/-) thymocytes. However, Parp-2-deficient thymocytes accumulated high levels of DNA double-strand breaks (DSB), independently of the p53 status, in line with a function of Parp-2 as a caretaker promoting genomic stability during thymocytes development. Although Parp-2(-/-) mice do not have spontaneous tumours, Parp-2 deficiency accelerated spontaneous tumour development in p53-null mice, mainly T-cell lymphomas. These data suggest a synergistic interaction between Parp-2 and p53 in tumour suppression through the role of Parp-2 in DNA-damage response and genome integrity surveillance, and point to the potential importance of examining human tumours for the status of both genes.
Collapse
Affiliation(s)
- L Nicolás
- Department of Immunology, IMIM-Hospital del Mar, Barcelona Biomedical Research Park, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Peralta-Leal A, Rodríguez-Vargas JM, Aguilar-Quesada R, Rodríguez MI, Linares JL, de Almodóvar MR, Oliver FJ. PARP inhibitors: new partners in the therapy of cancer and inflammatory diseases. Free Radic Biol Med 2009; 47:13-26. [PMID: 19362586 DOI: 10.1016/j.freeradbiomed.2009.04.008] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 04/07/2009] [Accepted: 04/08/2009] [Indexed: 12/21/2022]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are defined as cell signaling enzymes that catalyze the transfer of ADP-ribose units from NAD(+) to a number of acceptor proteins. PARP-1, the best characterized member of the PARP family, which currently comprises 18 members, is an abundant nuclear enzyme implicated in cellular responses to DNA injury provoked by genotoxic stress. PARP is involved in DNA repair and transcriptional regulation and is now recognized as a key regulator of cell survival and cell death as well as a master component of a number of transcription factors involved in tumor development and inflammation. PARP-1 is essential to the repair of DNA single-strand breaks via the base excision repair pathway. Inhibitors of PARP-1 have been shown to enhance the cytotoxic effects of ionizing radiation and DNA-damaging chemotherapy agents, such as the methylating agents and topoisomerase I inhibitors. There are currently at least five PARP inhibitors in clinical trial development. Recent in vitro and in vivo evidence suggests that PARP inhibitors could be used not only as chemo/radiotherapy sensitizers, but also as single agents to selectively kill cancers defective in DNA repair, specifically cancers with mutations in the breast cancer-associated genes (BRCA1 and BRCA2). PARP becomes activated in response to oxidative DNA damage and depletes cellular energy pools, thus leading to cellular dysfunction in various tissues. The activation of PARP may also induce various cell death processes and promotes an inflammatory response associated with multiple organ failure. Inhibition of PARP activity is protective in a wide range of inflammatory and ischemia-reperfusion-associated diseases, including cardiovascular diseases, diabetes, rheumatoid arthritis, endotoxic shock, and stroke. The aim of this review is to overview the emerging data in the literature showing the role of PARP in the pathogenesis of cancer and inflammatory diseases and unravel the solid body of literature that supports the view that PARP is an important target for therapeutic intervention in critical illness.
Collapse
Affiliation(s)
- Andreína Peralta-Leal
- Instituto de Parasitología y Biomedicina López Neyra, Consejo Superior de Investigaciones Cientificas (CSIC), Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
34
|
Ghodgaonkar MM, Shah RG, Kandan-Kulangara F, Affar EB, Qi HH, Wiemer E, Shah GM. Abrogation of DNA vector-based RNAi during apoptosis in mammalian cells due to caspase-mediated cleavage and inactivation of Dicer-1. Cell Death Differ 2009; 16:858-68. [PMID: 19229243 DOI: 10.1038/cdd.2009.15] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
RNA interference (RNAi) is used as a reverse-genetic tool to examine functions of a gene in different cellular processes including apoptosis. As key cellular proteins are inactivated during apoptosis, and as RNAi requires cooperation of many cellular proteins, we examined whether DNA vector-based RNAi would continue to function during apoptosis. The short hairpin RNA transcribed from the DNA vector is processed by Dicer-1 to form small interfering RNA that is incorporated in the RNA-induced silencing complex (RISC) to guide a sequence-specific silencing of the target mRNA. We report here that DNA vector-based RNAi of three different genes, namely poly(ADP-ribose) polymerase-1, p14(ARF) and lamin A/C are abrogated during apoptosis. The failure of DNA vector-based RNAi was not at the level of Ago-2 or RISC-mediated step of RNAi but due to catalytic inactivation of Dicer-1 on specific cleavage at the STTD(1476) and CGVD(1538) sites within its RNase IIIa domain. Using multiple approaches, caspase-3 was identified as the major caspase responsible for the cleavage and inactivation of Dicer-1. As Dicer-1 is also the common endonuclease required for formation of microRNA (miRNA) in mammalian cells, we observed decreased levels of mature forms of miR-16, miR-21 and let-7a. Our results suggest a role for apoptotic cleavage and inactivation of Dicer-1 in controlling apoptotic events through altered availability of miRNA.
Collapse
Affiliation(s)
- M M Ghodgaonkar
- Laboratory for Skin Cancer Research, Faculty of Medicine, CHUL Research Center (CHUQ), Laval University, QC, Canada
| | | | | | | | | | | | | |
Collapse
|