1
|
Wang S, Zhang M, Li T, Chen X, Wu Q, Tian D, Granot Z, Xu H, Hao J, Zhang H. A comprehensively prognostic and immunological analysis of PARP11 in pan-cancer. J Leukoc Biol 2024; 117:qiae030. [PMID: 38334307 DOI: 10.1093/jleuko/qiae030] [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: 12/12/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Poly (ADP-ribose) polymerase family member 11 (PARP11) has important immune regulatory functions in viral infection and tumor immune response. Particularly, PARP11 showed protumor activities in multiple preclinical murine models. However, no systematic pan-cancer analysis has been conducted to explore PARP11 function. In this study, we used multiple databases to assess PARP11 expression, which is associated with clinical outcomes, immune checkpoint factors, prognostic significance, genomic characteristics, and immunological aspects. The analysis revealed varying expression levels of PARP11 across different cancer types and a significant correlation between its expression and immune cell infiltration. Insights from the CellMiner database suggest a strong link between PARP11 expression and sensitivity to anticancer drugs, highlighting its potential as a therapeutic target. Moreover, PARP11 expression correlates with patient survival during anti-PD1 and anti-CTLA4 treatments, suggesting that PARP11 would be a predictor of immune checkpoint inhibitor treatment. In summary, PARP11 would be a potential immunoregulatory target and a diagnosis and prognosis marker for certain types of cancers. The detailed mechanisms of PARP11 in tumor immune responses need to be further investigated.
Collapse
Affiliation(s)
- Shengli Wang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Mingyue Zhang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Tao Li
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Xinru Chen
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Qinhan Wu
- College of Life Sciences, Nankai University, 94 Weijin Rd, Tianjin, 300071, P.R. China
| | - Dan Tian
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshaner St, Guangzhou, 510080, Guangdong, P.R. China
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Hadassah Medical School, Ein Kerem, 9112102, Jerusalem, Israel
| | - Hongbiao Xu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshaner St, Guangzhou, 510275, Guangdong, P.R. China
| | - Jianlei Hao
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, P.R. China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, P.R. China
| | - Hongru Zhang
- College of Life Sciences, Nankai University, 94 Weijin Rd, Tianjin, 300071, P.R. China
| |
Collapse
|
2
|
Chernyshova I, Vasil'eva I, Moor N, Ivanisenko N, Kutuzov M, Abramova T, Zakharenko A, Lavrik O. Aminomethylmorpholino Nucleosides as Novel Inhibitors of PARP1 and PARP2: Experimental and Molecular Modeling Analyses of Their Selectivity and Mechanism of Action. Int J Mol Sci 2024; 25:12526. [PMID: 39684238 DOI: 10.3390/ijms252312526] [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: 10/18/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 12/18/2024] Open
Abstract
Poly(ADP-ribose) polymerases 1 and 2 (PARP1 and PARP2) play a key role in DNA repair. As major sensors of DNA damage, they are activated to produce poly(ADP-ribose). PARP1/PARP2 inhibitors have emerged as effective drugs for the treatment of cancers with BRCA deficiencies. Here, we explored aminomethylmorpholino and aminomethylmorpholino glycine nucleosides as inhibitors of PARP1 and PARP2, using different enzymatic assays. The compounds bearing thymine or 5-Br(I)-uracil bases displayed the highest inhibition potency, with all of them being more selective toward PARP1. Interaction of the inhibitors with the NAD+ binding cavity of PARP1 (PARP2) suggested by the mixed-type inhibition was demonstrated by molecular docking and the RoseTTAFold All-Atom AI-model. The best PARP1 inhibitors characterized by the inhibition constants in the range of 12-15 µM potentiate the cytotoxicity of hydrogen peroxide by displaying strong synergism. The inhibitors revealed no impact on PARP1/PARP2 affinity for DNA, while they reduced the dissociation rate of the enzyme-DNA complex upon the autopoly(ADP-ribosyl)ation reaction, thus providing evidence that their mechanism of action for PARP trapping is due primarily to catalytic inhibition. The most active compounds were shown to retain selectivity toward PARP1, despite the reduced inhibition potency in the presence of histone PARylation factor 1 (HPF1) capable of regulating PARP1/PARP2 catalytic activity and ADP-ribosylation reaction specificity. The inhibitors obtained seem to be promising for further research as potential drugs.
Collapse
Affiliation(s)
- Irina Chernyshova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Inna Vasil'eva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Nina Moor
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Nikita Ivanisenko
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- AIRI, 123112 Moscow, Russia
| | - Mikhail Kutuzov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Tatyana Abramova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexandra Zakharenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Olga Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
3
|
Sakr TM, Elsabagh MF, Fayez H, Sarhan MO, Syam YM, Anwar MM, Motaleb MA, Zaghary WA. Multi-functionalization of reduced graphene oxide nanosheets for tumor theragnosis: Synthesis, characterization, enzyme assay, in-silico study, radiolabeling and in vivo targeting evaluation. Daru 2024; 32:77-95. [PMID: 38072913 PMCID: PMC11087444 DOI: 10.1007/s40199-023-00487-7] [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: 02/17/2023] [Accepted: 10/10/2023] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND In this study, a combination of nanotechnology, organic synthesis and radiochemistry were utilized in order to design an efficient nano-system conjugated with a suitable radionuclide and an antitumor agent for possible application as tumor theragnostic agent. METHOD Four novel compounds (3 and 4a-c) bearing tetrahydroquinazoline-7-sulfonohydrazide or 1,2,3,4-tetrahydroquinazoline-7-sulfonamide scaffold were designed. Then, docking study predicted that the compounds can be considered as potential inhibitors for PARP-1. Following that; the four compounds were synthesized and properly characterized using 1HNMR, 13CNMR, IR and Mass spectroscopy. The cytotoxic effect of the four compounds was evaluated against breast cancer cell line (MDA-MB-436), where compound 3 showed the most promising cytotoxic effect. The inhibitory effect of the four compounds was evaluated in vitro against PARP-1. RESULT Carboxylated graphene oxide nanosheets (NGO-COOH) were synthesized by a modified Hummer's method and has size of range 40 nm. The NGO-COOH nanosheets were proven to be safe and biocompatible when tested in vitro against normal human lung fibroblast cells (MRC-5). The prepared NGO-COOH nanosheets were conjugated with compound 3 then radiolabeled with 99mTc to yield 99mTc-NGO-COOH-3 with a radiochemical yield of 98.5.0 ± 0.5%. 99mTc-NGO-COOH-3 was injected intravenously in solid tumor bearing mice to study the degree of localization of the nano-system at tumor tissue. The results of the study revealed, excellent localization and retention of the designed nano-system at tumor tissues with targeting ratio of 9.0. CONCLUSION Stirred a new candidate tumor theragnostic agent that is safe, selective and stable.
Collapse
Affiliation(s)
- Tamer M Sakr
- Radioisotopes Production Facility, Second Egyptian Research Reactor Complex, Egyptian Atomic Energy Authority, Cairo, 13759, Egypt
- Radioactive Isotopes and Generator Department, Hot Labs Center, Egyptian Atomic Energy Authority, Cairo, 13759, Egypt
| | - Mohammed F Elsabagh
- Radioisotopes Production Facility, Second Egyptian Research Reactor Complex, Egyptian Atomic Energy Authority, Cairo, 13759, Egypt.
- Labeled Compounds Department, Hot Labs Center, Egyptian Atomic Energy Authority, Cairo, 13759, Egypt.
| | - Hend Fayez
- Labeled Compounds Department, Hot Labs Center, Egyptian Atomic Energy Authority, Cairo, 13759, Egypt
| | - Mona O Sarhan
- Labeled Compounds Department, Hot Labs Center, Egyptian Atomic Energy Authority, Cairo, 13759, Egypt
| | - Yasmin M Syam
- Department of Therapeutic Chemistry/ National Research Centre, Cairo, Egypt
| | - Manal M Anwar
- Department of Therapeutic Chemistry/ National Research Centre, Cairo, Egypt
| | - Mohammed A Motaleb
- Labeled Compounds Department, Hot Labs Center, Egyptian Atomic Energy Authority, Cairo, 13759, Egypt
| | - Wafaa A Zaghary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
4
|
Ghiandoni GM, Flanagan SR, Bodkin MJ, Nizi MG, Galera‐Prat A, Brai A, Chen B, Wallace JEA, Hristozov D, Webster J, Manfroni G, Lehtiö L, Tabarrini O, Gillet VJ. Synthetically accessible de novo design using reaction vectors: Application to PARP1 inhibitors. Mol Inform 2024; 43:e202300183. [PMID: 38258328 PMCID: PMC11475289 DOI: 10.1002/minf.202300183] [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: 08/21/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 01/24/2024]
Abstract
De novo design has been a hotly pursued topic for many years. Most recent developments have involved the use of deep learning methods for generative molecular design. Despite increasing levels of algorithmic sophistication, the design of molecules that are synthetically accessible remains a major challenge. Reaction-based de novo design takes a conceptually simpler approach and aims to address synthesisability directly by mimicking synthetic chemistry and driving structural transformations by known reactions that are applied in a stepwise manner. However, the use of a small number of hand-coded transformations restricts the chemical space that can be accessed and there are few examples in the literature where molecules and their synthetic routes have been designed and executed successfully. Here we describe the application of reaction-based de novo design to the design of synthetically accessible and biologically active compounds as proof-of-concept of our reaction vector-based software. Reaction vectors are derived automatically from known reactions and allow access to a wide region of synthetically accessible chemical space. The design was aimed at producing molecules that are active against PARP1 and which have improved brain penetration properties compared to existing PARP1 inhibitors. We synthesised a selection of the designed molecules according to the provided synthetic routes and tested them experimentally. The results demonstrate that reaction vectors can be applied to the design of novel molecules of biological relevance that are also synthetically accessible.
Collapse
Affiliation(s)
- Gian Marco Ghiandoni
- Information SchoolUniversity of SheffieldRegent Court, 211 PortobelloSheffieldS1 4DPUK
| | | | - Michael J. Bodkin
- Evotec (U.K.) Ltd114 Innovation Drive, Milton ParkAbingdonOX14 4RZUK
| | - Maria Giulia Nizi
- Department of Pharmaceutical SciencesUniversity of Perugia06123PerugiaItaly
| | - Albert Galera‐Prat
- Faculty of Biochemistry and Molecular Medicine & Biocenter OuluUniversity of OuluOuluFI-90014Finland
| | - Annalaura Brai
- Department of Biotechnology, Chemistry and PharmacyUniversity of SienaI-53100SienaItaly
| | - Beining Chen
- Department of ChemistryUniversity of SheffieldDainton Building, Brook HillSheffieldS3 7HFUK
| | | | - Dimitar Hristozov
- Evotec (U.K.) Ltd114 Innovation Drive, Milton ParkAbingdonOX14 4RZUK
| | - James Webster
- Information SchoolUniversity of SheffieldRegent Court, 211 PortobelloSheffieldS1 4DPUK
| | - Giuseppe Manfroni
- Department of Pharmaceutical SciencesUniversity of Perugia06123PerugiaItaly
| | - Lari Lehtiö
- Faculty of Biochemistry and Molecular Medicine & Biocenter OuluUniversity of OuluOuluFI-90014Finland
| | - Oriana Tabarrini
- Department of Pharmaceutical SciencesUniversity of Perugia06123PerugiaItaly
| | - Valerie J. Gillet
- Information SchoolUniversity of SheffieldRegent Court, 211 PortobelloSheffieldS1 4DPUK
| |
Collapse
|
5
|
Rahman A, Belur Ningegowda N, Kammathalli Siddappa M, Kumar Jain S, Malleshappa Kumaraswamy H, Achur R, Devappa Satyanarayan N, Malavalli Mahadevan K. Palladium‐Catalysed C−C Bond Forming 4‐Cyanophenyl‐nicotinamide Conjugates; Anti‐Pancreatic Cancer Screening on Capan‐1 Cell Line. ChemistrySelect 2023; 8. [DOI: 10.1002/slct.202204309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/22/2023] [Indexed: 01/04/2025]
Abstract
AbstractPancreatic cancer is the most severe, as a consequence of asymptomatic nature and ineffective therapies among all malignancies. Nicotinamides are effective ring systems in the treatment of pancreatic cancer with their wide range of applications. In the present investigation, nicotinamide and 4‐cyanophenyl ring systems are brought together to obtain greater potency. For the process of investigation, PARP1 protein is targeted and evaluated by docking at the active site to determine the protein‐ligand interaction, revealed the potential with the binding affinity of −9.0 to −11.0 Kcal/mol to inhibit the poly ADP‐ribose polymerase 1 (PARP1) pathway. The MTT‐assay assessment of a synthesized series has been performed against Capan‐1 pancreatic cancer cell line. The nicotinamide compounds demonstrated a significant inhibitory effect over Capan‐1 cell line, and 6‐(4‐cyanophenyl)‐N‐(3‐phenylpropyl)nicotinamide exhibited as a potential lead for the development of novel chemotherapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Abdul Rahman
- Department of Pharmaceutical Chemistry Kuvempu University Post Graduate Centre, Kadur 577548 Chikkamagaluru Karnataka India
| | - Nippu Belur Ningegowda
- Department of Pharmaceutical Chemistry Kuvempu University Post Graduate Centre, Kadur 577548 Chikkamagaluru Karnataka India
| | | | - Sandeep Kumar Jain
- Laboratory of Experimental Medicine Department of Biotechnology Kuvempu University 577451 Shankaragatta Karnataka India
| | | | - Rajeshwara Achur
- Department of Biochemistry Kuvempu University Shankargatta 577451 Shimoga Karnataka India
| | - Nayak Devappa Satyanarayan
- Department of Pharmaceutical Chemistry Kuvempu University Post Graduate Centre, Kadur 577548 Chikkamagaluru Karnataka India
| | - Kittappa Malavalli Mahadevan
- Kittappa Malavalli Mahadevand Department of Chemistry Kuvempu University Post Graduate Centre, Kadur 577548 Chikkamagaluru Karnataka India
| |
Collapse
|
6
|
Thein KZ, Thawani R, Kummar S. Combining Poly (ADP-Ribose) Polymerase (PARP) Inhibitors with Chemotherapeutic Agents: Promise and Challenges. Cancer Treat Res 2023; 186:143-170. [PMID: 37978135 DOI: 10.1007/978-3-031-30065-3_9] [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] [Indexed: 11/19/2023]
Abstract
Better understanding of molecular drivers and dysregulated pathways has furthered the concept of precision oncology and rational drug development. The role of DNA damage response (DDR) pathways has been extensively studied in carcinogenesis and as potential therapeutic targets to improve response to chemotherapy or overcome resistance. Treatment with small molecule inhibitors of PARP has resulted in clinical response and conferred survival benefit to patients with ovarian cancer, BRCA-mutant breast cancer, HRD-deficient prostate cancer and BRCA-mutant pancreatic cancer, leading to US Food and Drug Administration (FDA) approvals. However, the observed clinical benefit with single agent PARP inhibitors is limited to few tumor types within the relevant genetic context. Since DDR pathways are essential for repair of damage caused by cytotoxic agents, PARP inhibitors have been evaluated in combination with various chemotherapeutic agents to broaden the therapeutic application of this class of drugs. In this chapter, we discuss the combination of PARP inhibitors with different chemotherapeutics agents, clinical experience to date, lessons learnt, and future directions for this approach.
Collapse
Affiliation(s)
- Kyaw Zin Thein
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| | - Rajat Thawani
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| | - Shivaani Kummar
- DeArmond Endowed Chair of Cancer Research, Division of Hematology and Medical Oncology, Clinical and Translational Research, Knight Cancer Institute (KCI), Center for Experimental Therapeutics (KCI), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, OC14HO, Portland, OR, 97239, USA.
| |
Collapse
|
7
|
Wu J, Crowe DL. PARP5B is required for nonhomologous end joining during tumorigenesis in vivo. Mol Carcinog 2021; 61:85-98. [PMID: 34710250 DOI: 10.1002/mc.23363] [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: 05/29/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/27/2022]
Abstract
Poly(ADP-ribose) polymerases (PARP) act as DNA damage sensors that produce poly(ADP-ribose) (PAR) chains at double-strand breaks, facilitating the recruitment of repair factors. Cancers with homologous recombination defects are sensitive to small molecule PARP inhibitors. Despite PARP5B gene copy number changes in many cancers, the effects of this genetic alteration on tumor phenotype are largely unknown. To better understand this clinical finding, we characterized a PARP5B null mutation in a carcinogen-induced in vivo head and neck squamous cell carcinoma (SCC) model. Reduced PARP5B expression inhibited tumor growth, induced primary tumor differentiation and apoptosis, and inhibited cell proliferation and metastasis. Loss of PARP5B expression-induced ataxia telangiectasia and Rad3 related (ATR) activation and depleted the cancer stem cell fraction. PARP5B null tumor cells lacked 53BP1+ double-strand break foci, ATM activation, and p53 induction compared to PARP5B+/+ cancers. PARP5B null SCC expresses a multiprotein complex containing PML, pRPA, Rad50, Rad51, XRCC1, proliferating cell nuclear antigen (PCNA), and Mcm2, suggesting an HR-mediated repair mechanism at DNA replication foci. Low doses of etoposide combined with the PARP5B inhibitor XAV939 induced senescence and apoptosis in human SCC lines. NBS1 overexpression in these cells inhibited the effects of low-dose etoposide/XAV939 treatment. Our results indicate that PARP5B inhibition is new targeted cancer therapy.
Collapse
Affiliation(s)
- Jianchun Wu
- Department of Diagnostic Sciences, University of Illinois Cancer Center, Chicago, Illinois, USA
| | - David L Crowe
- Department of Diagnostic Sciences, University of Illinois Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
8
|
Johannes JW, Balazs A, Barratt D, Bista M, Chuba MD, Cosulich S, Critchlow SE, Degorce SL, Di Fruscia P, Edmondson SD, Embrey K, Fawell S, Ghosh A, Gill SJ, Gunnarsson A, Hande SM, Heightman TD, Hemsley P, Illuzzi G, Lane J, Larner C, Leo E, Liu L, Madin A, Martin S, McWilliams L, O'Connor MJ, Orme JP, Pachl F, Packer MJ, Pei X, Pike A, Schimpl M, She H, Staniszewska AD, Talbot V, Underwood E, Varnes JG, Xue L, Yao T, Zhang K, Zhang AX, Zheng X. Discovery of 5-{4-[(7-Ethyl-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl]piperazin-1-yl}- N-methylpyridine-2-carboxamide (AZD5305): A PARP1-DNA Trapper with High Selectivity for PARP1 over PARP2 and Other PARPs. J Med Chem 2021; 64:14498-14512. [PMID: 34570508 DOI: 10.1021/acs.jmedchem.1c01012] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Poly-ADP-ribose-polymerase (PARP) inhibitors have achieved regulatory approval in oncology for homologous recombination repair deficient tumors including BRCA mutation. However, some have failed in combination with first-line chemotherapies, usually due to overlapping hematological toxicities. Currently approved PARP inhibitors lack selectivity for PARP1 over PARP2 and some other 16 PARP family members, and we hypothesized that this could contribute to toxicity. Recent literature has demonstrated that PARP1 inhibition and PARP1-DNA trapping are key for driving efficacy in a BRCA mutant background. Herein, we describe the structure- and property-based design of 25 (AZD5305), a potent and selective PARP1 inhibitor and PARP1-DNA trapper with excellent in vivo efficacy in a BRCA mutant HBCx-17 PDX model. Compound 25 is highly selective for PARP1 over other PARP family members, with good secondary pharmacology and physicochemical properties and excellent pharmacokinetics in preclinical species, with reduced effects on human bone marrow progenitor cells in vitro.
Collapse
Affiliation(s)
- Jeffrey W Johannes
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Amber Balazs
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Derek Barratt
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Michal Bista
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Matthew D Chuba
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Sabina Cosulich
- Oncology Projects, Oncology R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | | | - Sébastien L Degorce
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | | | - Scott D Edmondson
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Kevin Embrey
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Stephen Fawell
- Oncology Discovery, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Avipsa Ghosh
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Sonja J Gill
- Oncology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Anders Gunnarsson
- Discovery Sciences, R&D Gothenburg, AstraZeneca, KJ2, Pepparedsleden 1, SE-431 83 Mölndal, Sweden
| | - Sudhir M Hande
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Tom D Heightman
- Chemistry, Oncology R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Paul Hemsley
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | | | - Jordan Lane
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Carrie Larner
- Oncology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Elisabetta Leo
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Lina Liu
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Andrew Madin
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Scott Martin
- DMPK, Oncology R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Lisa McWilliams
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Mark J O'Connor
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Jonathan P Orme
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Fiona Pachl
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Martin J Packer
- Computational Chemistry, Oncology R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | - Xiaohui Pei
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Andrew Pike
- DMPK, Oncology R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | | | - Hongyao She
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | | | - Verity Talbot
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 OWG, U.K
| | | | - Jeffrey G Varnes
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Lin Xue
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Tieguang Yao
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Ke Zhang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Andrew X Zhang
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Xiaolan Zheng
- Chemistry, Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
9
|
Huang M, Ren J, Wang Y, Chen X, Yang J, Tang T, Yang Z, Li X, Ji M, Cai J. Design, Synthesis and Activity Evaluation of New Phthalazinone PARP Inhibitors. Chem Pharm Bull (Tokyo) 2021; 69:620-629. [PMID: 34193711 DOI: 10.1248/cpb.c20-01018] [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: 11/22/2022]
Abstract
Poly(ADP-ribose)polymerase (PARP) is a significant therapeutic target for the treatment of numerous human diseases. Olaparib has been approved as a PARP inhibitor. In this paper, a series of new compounds were designed and synthesized with Olaparib as the lead compound. In order to evaluate the inhibitory activities against PARP1 of the synthesized compounds, in vitro PARP1 inhibition assay and intracellular PARylation assay were conducted. The results showed that the inhibitory activities of the derivatives were related to the type of substituent and the length of alkyl chain connecting the aromatic ring. 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT)-based assay also proved that these compounds demonstrating strong inhibition to PARP1 also have high anti-proliferative activities against BRCA2-deficient cell line (Capan-1). Analysis of the entire results suggest that compound 23 with desirable inhibitory efficiency may hold promise for further in vivo exploration of PARP inhibition.
Collapse
Affiliation(s)
- Mingqi Huang
- School of Chemistry and Chemical Engineering, Southeast University
| | - Jinghui Ren
- School of Chemistry and Chemical Engineering, Southeast University
| | - Yuhong Wang
- School of Chemistry and Chemical Engineering, Southeast University
| | - Xixi Chen
- School of Chemistry and Chemical Engineering, Southeast University
| | - Jia Yang
- School of Chemistry and Chemical Engineering, Southeast University
| | - Tu Tang
- School of Chemistry and Chemical Engineering, Southeast University
| | - Zhenyong Yang
- School of Chemistry and Chemical Engineering, Southeast University
| | - Xiaojing Li
- School of Chemistry and Chemical Engineering, Southeast University
| | - Min Ji
- School of Biological Science & Medical Engineering, Southeast University
| | - Jin Cai
- School of Chemistry and Chemical Engineering, Southeast University
| |
Collapse
|
10
|
Li Y, Liu CF, Rao GW. A Review on Poly (ADP-ribose) Polymerase (PARP) Inhibitors and Synthetic Methodologies. Curr Med Chem 2021; 28:1565-1584. [PMID: 32164505 DOI: 10.2174/0929867327666200312113011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 11/22/2022]
Abstract
Poly (ADP-ribose) polymerase (PARP) acts as an essential DNA repair enzyme. PARP inhibitors are novel small molecule targeted drugs based on the principle of "Synthetic Lethality", which affect DNA repair process by competitively inhibiting the activity of PARP enzyme and thereby kill cancer cells. Currently, four PARP inhibitors including olaparib, rucaparib, niraparib, and talazoparib have been approved by FDA for cancer treatment and have achieved great success in the treatment of ovarian cancer, breast cancer, and pancreatic cancer, etc. This paper provides a general overview of the research progress of PARP inhibitors including the major structure types, structure-activity relationship (SAR), and synthetic routes, with the aim of providing ideas for the discovery and synthesis of novel PARP inhibitors.
Collapse
Affiliation(s)
- Ying Li
- College of Pharmaceutical Science, Zhejiang University of Technology and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chen-Fu Liu
- School of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Zhejiang University of Technology and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
11
|
Bioinformatic Analysis of the Nicotinamide Binding Site in Poly(ADP-Ribose) Polymerase Family Proteins. Cancers (Basel) 2021; 13:cancers13061201. [PMID: 33801950 PMCID: PMC8002165 DOI: 10.3390/cancers13061201] [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: 02/02/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary The PARP family consists of 17 proteins, and some of them are responsible for cancer cells’ viability. Much attention is therefore given to the search for chemical compounds with the ability to suppress distinct PARP family members (for example, PARP-5a and 5b). Here, we present the results of a family-wide bioinformatic analysis of an important functional region in the PARP structure and describe factors that can guide the design of highly selective compounds. Abstract The PARP family consists of 17 members with diverse functions, including those related to cancer cells’ viability. Several PARP inhibitors are of great interest as innovative anticancer drugs, but they have low selectivity towards distinct PARP family members and exert serious adverse effects. We describe a family-wide study of the nicotinamide (NA) binding site, an important functional region in the PARP structure, using comparative bioinformatic analysis and molecular modeling. Mutations in the NA site and D-loop mobility around the NA site were identified as factors that can guide the design of selective PARP inhibitors. Our findings are of particular importance for the development of novel tankyrase (PARPs 5a and 5b) inhibitors for cancer therapy.
Collapse
|
12
|
Poltronieri P, Celetti A, Palazzo L. Mono(ADP-ribosyl)ation Enzymes and NAD + Metabolism: A Focus on Diseases and Therapeutic Perspectives. Cells 2021; 10:cells10010128. [PMID: 33440786 PMCID: PMC7827148 DOI: 10.3390/cells10010128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
Mono(ADP-ribose) transferases and mono(ADP-ribosyl)ating sirtuins use NAD+ to perform the mono(ADP-ribosyl)ation, a simple form of post-translational modification of proteins and, in some cases, of nucleic acids. The availability of NAD+ is a limiting step and an essential requisite for NAD+ consuming enzymes. The synthesis and degradation of NAD+, as well as the transport of its key intermediates among cell compartments, play a vital role in the maintenance of optimal NAD+ levels, which are essential for the regulation of NAD+-utilizing enzymes. In this review, we provide an overview of the current knowledge of NAD+ metabolism, highlighting the functional liaison with mono(ADP-ribosyl)ating enzymes, such as the well-known ARTD10 (also named PARP10), SIRT6, and SIRT7. To this aim, we discuss the link of these enzymes with NAD+ metabolism and chronic diseases, such as cancer, degenerative disorders and aging.
Collapse
Affiliation(s)
- Palmiro Poltronieri
- Institute of Sciences of Food Productions, National Research Council of Italy, via Monteroni 7, 73100 Lecce, Italy
- Correspondence: (P.P.); (A.C.); (L.P.)
| | - Angela Celetti
- Institute for the Experimental Endocrinology and Oncology, National Research Council of Italy, Via Sergio Pansini 5, 80131 Naples, Italy
- Correspondence: (P.P.); (A.C.); (L.P.)
| | - Luca Palazzo
- Institute for the Experimental Endocrinology and Oncology, National Research Council of Italy, Via Tommaso de Amicis 95, 80145 Naples, Italy
- Correspondence: (P.P.); (A.C.); (L.P.)
| |
Collapse
|
13
|
Ferreira MT, Berger L, Rouleau M, Poirier GG. Assessment of PARP-1 Distribution in Tissues of Cynomolgus Monkeys. J Histochem Cytochem 2020; 68:413-435. [PMID: 32436762 DOI: 10.1369/0022155420926022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The poly(ADP-ribose) polymerase 1 (PARP-1) enzyme has received much attention in the last decade due to its promising role in cancer therapeutics. Despite the expanding use of PARP inhibitors in cancer therapy, little is known about PARP-1 tissue distribution. Our study provides a detailed survey of PARP-1 tissue and cellular distribution using well-preserved cynomolgus monkey organs and a well-characterized, highly specific monoclonal PARP-1 antibody. Overall, PARP-1 was detected in most organs, but its distribution was restricted to specific cells within each tissue, suggesting that PARP-1 expression is tightly regulated. The strongest expression was in the pituitary, the ovary, the male adrenal gland, and the thymus. One of the key findings of this study was the stronger expression of PARP-1 in proliferating cells rather than mature cells. This observation not only provides clues to the importance of PARP-1 in processes such as DNA replication and transcription in these cell types, but it also provides the basis for further investigation into the effects of its inhibition in the context of malignancy. Overall, this study greatly expands the current knowledge of PARP-1 tissue expression, enabling the identification of tissues where PARP inhibition may be most efficacious.
Collapse
Affiliation(s)
- Maria Tedim Ferreira
- Department of Molecular Biology, Medical Biochemistry and Pathology, Centre de Recherche sur le Cancer, Laval University, Québec City, QC, Canada.,CHU de Québec Research Center, Oncology Axis, Québec City, QC, Canada
| | - Louise Berger
- CHU de Québec, St-François d'Assise Hospital, Québec City, QC, Canada
| | - Michèle Rouleau
- CHU de Québec Research Center, Oncology Axis, Québec City, QC, Canada
| | - Guy G Poirier
- Department of Molecular Biology, Medical Biochemistry and Pathology, Centre de Recherche sur le Cancer, Laval University, Québec City, QC, Canada.,CHU de Québec Research Center, Oncology Axis, Québec City, QC, Canada
| |
Collapse
|
14
|
Makhov P, Uzzo RG, Tulin AV, Kolenko VM. Histone-dependent PARP-1 inhibitors: A novel therapeutic modality for the treatment of prostate and renal cancers. Urol Oncol 2020; 39:312-315. [PMID: 32402770 DOI: 10.1016/j.urolonc.2020.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/02/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022]
Abstract
Clinical interest in poly(ADP-ribose) polymerase 1 (PARP-1) has increased over the past decade with the recognition of its roles in transcription regulation, DNA repair, epigenetic bookmarking, and chromatin restructuring. A number of PARP-1 inhibitors demonstrating clinical efficacy against tumors of various origins have emerged in recent years. These inhibitors have been essentially designed as nicotinamide adenine dinucleotide (NAD+) mimetics. However, because NAD+ is utilized by many enzymes other than PARP-1, NAD+ competitors tend to produce certain off-target effects. To overcome the limitation of NAD-like PARP-1 inhibitors, we have developed a new class of PARP-1 inhibitors that specifically targets the histone-dependent route of PARP-1 activation, a mechanism of activation that is unique to PARP-1. Novel histone-dependent inhibitors are highly specific for PARP-1 and demonstrate promising in vitro and in vivo efficacy against prostate and renal tumors. Our findings suggest that novel PARP-1 inhibitors have strong therapeutic potential for the treatment of urological tumors.
Collapse
Affiliation(s)
- Peter Makhov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Robert G Uzzo
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | | | | |
Collapse
|
15
|
Tomassi S, Pfahler J, Mautone N, Rovere A, Esposito C, Passeri D, Pellicciari R, Novellino E, Pannek M, Steegborn C, Paiardini A, Mai A, Rotili D. From PARP1 to TNKS2 Inhibition: A Structure-Based Approach. ACS Med Chem Lett 2020; 11:862-868. [PMID: 32435397 DOI: 10.1021/acsmedchemlett.9b00654] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/03/2020] [Indexed: 11/28/2022] Open
Abstract
Tankyrases (TNKSs) have recently gained great consideration as potential targets in Wnt/β-catenin pathway-dependent solid tumors. Previously, we reported the 2-mercaptoquinazolin-4-one MC2050 as a micromolar PARP1 inhibitor. Here we show how the resolution of the X-ray structure of PARP1 in complex with MC2050, combined with the computational investigation of the structural differences between TNKSs and PARP1/2 active sites, provided the rationale for a structure-based drug design campaign that with a limited synthetic effort led to the discovery of the bis-quinazolinone 5 as a picomolar and selective TNKS2 inhibitor, endowed with antiproliferative effects in a colorectal cancer cell line (DLD-1) where the Wnt pathway is constitutively activated.
Collapse
Affiliation(s)
- Stefano Tomassi
- Department of Pharmacy, University of Naples, “Federico II”, 80131 Naples, Italy
| | - Julian Pfahler
- Department of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany
| | - Nicola Mautone
- Department of Chemistry and Technology of Drugs, ″Sapienza” University of Rome, 00185 Rome, Italy
| | - Annarita Rovere
- Department of Chemistry and Technology of Drugs, ″Sapienza” University of Rome, 00185 Rome, Italy
| | - Chiara Esposito
- Department of Biochemical Sciences “A. Rossi Fanelli″, ″Sapienza” University of Rome, 00185 Rome, Italy
| | | | | | - Ettore Novellino
- Department of Pharmacy, University of Naples, “Federico II”, 80131 Naples, Italy
| | - Martin Pannek
- Department of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany
| | - Clemens Steegborn
- Department of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany
| | - Alessandro Paiardini
- Department of Biochemical Sciences “A. Rossi Fanelli″, ″Sapienza” University of Rome, 00185 Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technology of Drugs, ″Sapienza” University of Rome, 00185 Rome, Italy
| | - Dante Rotili
- Department of Chemistry and Technology of Drugs, ″Sapienza” University of Rome, 00185 Rome, Italy
| |
Collapse
|
16
|
Sahin K, Durdagi S. Identifying new piperazine-based PARP1 inhibitors using text mining and integrated molecular modeling approaches. J Biomol Struct Dyn 2020; 39:681-690. [PMID: 32048546 DOI: 10.1080/07391102.2020.1715262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
One of the important molecular targets for antitumor drug discovery is the polyadenosine diphosphate-ribose polymerase-1 (PARP1) enzyme. It is linked with various biological functions including DNA repair and apoptosis. It is primarily a nuclear enzyme linked to chromatin, which is activated by DNA damage. Improved expression of PARP1 in melanomas, breast cancer, lung cancer and other neoplastic diseases is often observed. A tremendous PARP research concerning cancer and ischemia is progressing very rapidly. There are currently four PARP1 inhibitors approved by the FDA on the market, namely Olaparib, Rucaparib, Niraparib and Talazoparib. All of these molecules are non-selective inhibitors of PARP1. Currently there is an urgent need for novel and selective PARP1 inhibitors. In this work, asmall molecule database (Specs SC) were used to identify the new selective lead inhibitors of PARP1. Piperazine scaffold is an important fragment that is used in many currently used FDA approved drugs in different diseases including PARP1 inhibitor Olaparib. Thus, based on text mining studies, 4674 compounds thatinclude piperazine fragments were identified and virtually screened at the binding pocket of target protein PARP1. Compounds that have high docking scores were used in molecular dynamics (MD) simulations. Free energy calculations were also performed to compare the predicted binding energies with known PARP1 inhibitors. The critical amino acid interactions of these newly identified hits in the binding pocket were also investigated in detail for better understanding of the structural features required for next generation PARP1 inhibitors. Thus, here together with combination of text-mining and integrated molecular modeling approaches, we identified novel piperazine-based hits against PARP1 enzyme.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kader Sahin
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| |
Collapse
|
17
|
Design, Synthesis and Molecular Modeling Study of Conjugates of ADP and Morpholino Nucleosides as A Novel Class of Inhibitors of PARP-1, PARP-2 and PARP-3. Int J Mol Sci 2019; 21:ijms21010214. [PMID: 31892271 PMCID: PMC6982223 DOI: 10.3390/ijms21010214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
We report on the design, synthesis and molecular modeling study of conjugates of adenosine diphosphate (ADP) and morpholino nucleosides as potential selective inhibitors of poly(ADP-ribose)polymerases-1, 2 and 3. Sixteen dinucleoside pyrophosphates containing natural heterocyclic bases as well as 5-haloganeted pyrimidines, and mimicking a main substrate of these enzymes, nicotinamide adenine dinucleotide (NAD+)-molecule, have been synthesized in a high yield. Morpholino nucleosides have been tethered to the β-phosphate of ADP via a phosphoester or phosphoramide bond. Screening of the inhibiting properties of these derivatives on the autopoly(ADP-ribosyl)ation of PARP-1 and PARP-2 has shown that the effect depends upon the type of nucleobase as well as on the linkage between ADP and morpholino nucleoside. The 5-iodination of uracil and the introduction of the P–N bond in NAD+-mimetics have shown to increase inhibition properties. Structural modeling suggested that the P–N bond can stabilize the pyrophosphate group in active conformation due to the formation of an intramolecular hydrogen bond. The most active NAD+ analog against PARP-1 contained 5-iodouracil 2ʹ-aminomethylmorpholino nucleoside with IC50 126 ± 6 μM, while in the case of PARP-2 it was adenine 2ʹ-aminomethylmorpholino nucleoside (IC50 63 ± 10 μM). In silico analysis revealed that thymine and uracil-based NAD+ analogs were recognized as the NAD+-analog that targets the nicotinamide binding site. On the contrary, the adenine 2ʹ-aminomethylmorpholino nucleoside-based NAD+ analogs were predicted to identify as PAR-analogs that target the acceptor binding site of PARP-2, representing a novel molecular mechanism for selective PARP inhibition. This discovery opens a new avenue for the rational design of PARP-1/2 specific inhibitors.
Collapse
|
18
|
Karpova Y, Wu C, Divan A, McDonnell ME, Hewlett E, Makhov P, Gordon J, Ye M, Reitz AB, Childers WE, Skorski T, Kolenko V, Tulin AV. Non-NAD-like PARP-1 inhibitors in prostate cancer treatment. Biochem Pharmacol 2019; 167:149-162. [PMID: 30880062 PMCID: PMC6702078 DOI: 10.1016/j.bcp.2019.03.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/13/2019] [Indexed: 01/03/2023]
Abstract
In our previous studies of the molecular mechanisms of poly(ADP-ribose) polymerase 1 (PARP-1)-mediated transcriptional regulation we identified a novel class of PARP-1 inhibitors targeting the histone-dependent route of PARP-1 activation. Because histone-dependent activation is unique to PARP-1, non-NAD-like PARP-1 inhibitors have the potential to bypass the off-target effects of classical NAD-dependent PARP-1 inhibitors, such as olaparib, veliparib, and rucaparib. Furthermore, our recently published studies demonstrate that, compared to NAD-like PARP-1 inhibitors that are used clinically, the non-NAD-like PARP-1 inhibitor 5F02 exhibited superior antitumor activity in cell and animal models of human prostate cancer (PC). In this study, we further evaluated the antitumor activity of 5F02 and several of its novel analogues against PC cells. In contrast to NAD-like PARP-1 inhibitors, non-NAD-like PARP-1 inhibitors demonstrated efficacy against androgen-dependent and -independent routes of androgen receptor signaling activation. Our experiments reveal that methylation of the quaternary ammonium salt and the presence of esters were critical for the antitumor activity of 5F02 against PC cells. In addition, we examined the role of a related regulatory protein of PARP-1, called Poly(ADP-ribose) glycohydrolase (PARG), in prostate carcinogenesis. Our study reveals that PARG expression is severely disrupted in PC cells, which is associated with decreased integrity and localization of Cajal bodies (CB). Overall, the results of our study strengthen the justification for using non-NAD-like PARP-1 inhibitors as a novel therapeutic strategy for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
| | - Chao Wu
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Ali Divan
- University of North Dakota, Grand Forks, ND, United States
| | - Mark E McDonnell
- Fox Chase Chemical Diversity Center, Inc., Philadelphia, PA, United States
| | - Elizabeth Hewlett
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, United States
| | - Peter Makhov
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | - John Gordon
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, United States
| | - Min Ye
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, United States
| | - Allen B Reitz
- Fox Chase Chemical Diversity Center, Inc., Philadelphia, PA, United States
| | - Wayne E Childers
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, United States
| | - Tomasz Skorski
- Department of Microbiology and Immunology and Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | | | - Alexei V Tulin
- University of North Dakota, Grand Forks, ND, United States.
| |
Collapse
|
19
|
Kumar C, P.T.V. L, Arunachalam A. Structure based pharmacophore study to identify possible natural selective PARP-1 trapper as anti-cancer agent. Comput Biol Chem 2019; 80:314-323. [DOI: 10.1016/j.compbiolchem.2019.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
|
20
|
Guo T, Zuo Y, Qian L, Liu J, Yuan Y, Xu K, Miao Y, Feng Q, Chen X, Jin L, Zhang L, Dong C, Xiong S, Zheng H. ADP-ribosyltransferase PARP11 modulates the interferon antiviral response by mono-ADP-ribosylating the ubiquitin E3 ligase β-TrCP. Nat Microbiol 2019; 4:1872-1884. [DOI: 10.1038/s41564-019-0428-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 03/02/2019] [Indexed: 12/22/2022]
|
21
|
Jain PG, Patel BD. Medicinal chemistry approaches of poly ADP-Ribose polymerase 1 (PARP1) inhibitors as anticancer agents - A recent update. Eur J Med Chem 2019; 165:198-215. [PMID: 30684797 DOI: 10.1016/j.ejmech.2019.01.024] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/28/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022]
Abstract
Poly (ADP-ribose) Polymerase1 (PARP1) is a member of 17 membered PARP family having diversified biological functions such as synthetic lethality, DNA repair, apoptosis, necrosis, histone binding etc. It is primarily a chromatin-bound nuclear enzyme that gets activated by DNA damage. It binds to DNA signal- and double-strand breaks, does parylation of target proteins (using NAD+ as a substrate) like histones and other DNA repair proteins and modifies them as a part of DNA repair mechanism. Inhibition of PARP1 prevents the DNA repair and leads to cell death. Clinically, PARP1 Inhibitors have shown their potential in treating BRCAm breast and ovarian cancers and trials are going on for the treatment of other solid tumors like pancreatic, prostate, colorectal etc. as a single agent or in combination. There are currently three FDA approved PARP1 inhibitors namely Olaparib, Rucaparib and Niraparib in the market while Veliparib and Talazoparib are in the late stage of clinical development. All these molecules are nonselective PARP1 inhibitors with concurrent inhibition of PARP2 with similar potency. In addition, resistance to marketed PARP1 inhibitors has been reported. Overall, looking at the success rate of PARP1 inhibitors into various solid tumors, there is an urge of a novel and selective PARP1 inhibitors. This review provides an update on various newer heterocyclic PARP1 inhibitors reported in last three years along with their structural design strategies. We classified them into two main chemical classes; NAD analogues and non-NAD analogues and discussed the medicinal chemistry approaches of each class. To understand the structural features required for in-silico designing of next-generation PARP1 inhibitors, we also reported the crucial amino acid interactions of these inhibitors at the target site. Thus, present review provides the insight on recent development on new lead structures as PARP1 inhibitors, their SAR, an overview of in-vitro and in-vivo screening methods, current challenges and opinion on future designing of more selective and safe PARP1 inhibitors.
Collapse
Affiliation(s)
- Priyancy G Jain
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Bhumika D Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India.
| |
Collapse
|
22
|
Prakash A, Garcia-Moreno JF, Brown JAL, Bourke E. Clinically Applicable Inhibitors Impacting Genome Stability. Molecules 2018; 23:E1166. [PMID: 29757235 PMCID: PMC6100577 DOI: 10.3390/molecules23051166] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 12/14/2022] Open
Abstract
Advances in technology have facilitated the molecular profiling (genomic and transcriptomic) of tumours, and has led to improved stratification of patients and the individualisation of treatment regimes. To fully realize the potential of truly personalised treatment options, we need targeted therapies that precisely disrupt the compensatory pathways identified by profiling which allow tumours to survive or gain resistance to treatments. Here, we discuss recent advances in novel therapies that impact the genome (chromosomes and chromatin), pathways targeted and the stage of the pathways targeted. The current state of research will be discussed, with a focus on compounds that have advanced into trials (clinical and pre-clinical). We will discuss inhibitors of specific DNA damage responses and other genome stability pathways, including those in development, which are likely to synergistically combine with current therapeutic options. Tumour profiling data, combined with the knowledge of new treatments that affect the regulation of essential tumour signalling pathways, is revealing fundamental insights into cancer progression and resistance mechanisms. This is the forefront of the next evolution of advanced oncology medicine that will ultimately lead to improved survival and may, one day, result in many cancers becoming chronic conditions, rather than fatal diseases.
Collapse
Affiliation(s)
- Anu Prakash
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| | - Juan F Garcia-Moreno
- Discipline of Surgery, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| | - James A L Brown
- Discipline of Surgery, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| | - Emer Bourke
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| |
Collapse
|
23
|
Identification of Protein Substrates of Specific PARP Enzymes Using Analog-Sensitive PARP Mutants and a "Clickable" NAD + Analog. Methods Mol Biol 2018; 1608:111-135. [PMID: 28695507 DOI: 10.1007/978-1-4939-6993-7_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The PARP family of ADP-ribosyl transferases contains 17 members in human cells, most of which catalyze the transfer of the ADP-ribose moiety of NAD+ onto their target proteins. This posttranslational modification plays important roles in cellular signaling, especially during cellular stresses, such as heat shock, inflammation, unfolded protein responses, and DNA damage. Knowing the specific proteins that are substrates for individual PARPs, as well as the specific amino acid residues in a given target protein that are ADP-ribosylated, is a key step in understanding the biology of individual PARPs. Recently, we developed a robust NAD+ analog-sensitive approach for PARPs, which allows PARP-specific ADP-ribosylation of substrates that is suitable for subsequent copper-catalyzed azide-alkyne cycloaddition ("click chemistry") reactions. When coupled with proteomics and mass spectrometry, the analog-sensitive PARP approach can be used to identify the specific amino acids that are ADP-ribosylated by individual PARP proteins. In this chapter, we describe the key facets of the experimental design and application of the analog-sensitive PARP methodology to identify site-specific modification of PARP target proteins.
Collapse
|
24
|
Lu Y, Liu Y, Pang Y, Pacak K, Yang C. Double-barreled gun: Combination of PARP inhibitor with conventional chemotherapy. Pharmacol Ther 2018; 188:168-175. [PMID: 29621593 DOI: 10.1016/j.pharmthera.2018.03.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
DNA repair pathways are evolutionarily conserved molecular mechanisms that maintain the integrity of genomic DNA. In cancer therapies, the integrity and activity of DNA repair pathways predict therapy resistance and disease outcome. Members of the poly (ADP-ribose) polymerase (PARP) family initiate and organize the biologic process of DNA repair, which counteracts many types of chemotherapies. Since the first development in approximately 3 decades ago, PARP inhibitors have greatly changed the concept of cancer therapy, leading to encouraging improvements in tumor suppression and disease outcomes. Here we summaries both pre-clinical and clinical findings of PARP inhibitors applications, particularly for combination therapies.
Collapse
Affiliation(s)
- Yanxin Lu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; Basic Medical Science Department, Zunyi Medical College-Zhuhai Campus, Zhuhai, Guangdong 519041, PR China
| | - Yang Liu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Ying Pang
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
He M, Yuan H, Tan B, Bai R, Kim HS, Bae S, Che L, Kim JS, Gao SJ. SIRT1-mediated downregulation of p27Kip1 is essential for overcoming contact inhibition of Kaposi's sarcoma-associated herpesvirus transformed cells. Oncotarget 2018; 7:75698-75711. [PMID: 27708228 PMCID: PMC5342771 DOI: 10.18632/oncotarget.12359] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/21/2016] [Indexed: 11/25/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus associated with Kaposi's sarcoma (KS), a malignancy commonly found in AIDS patients. Despite intensive studies in the last two decades, the mechanism of KSHV-induced cellular transformation and tumorigenesis remains unclear. In this study, we found that the expression of SIRT1, a metabolic sensor, was upregulated in a variety of KSHV-infected cells. In a model of KSHV-induced cellular transformation, SIRT1 knockdown with shRNAs or knockout by CRISPR/Cas9 gene editing dramatically suppressed cell proliferation and colony formation in soft agar of KSHV-transformed cells by inducing cell cycle arrest and contact inhibition. SIRT1 knockdown or knockout induced the expression of cyclin-dependent kinase inhibitor 1B (p27Kip1). Consequently, p27 knockdown rescued the inhibitory effect of SIRT1 knockdown or knockout on cell proliferation and colony formation. Furthermore, treatment of KSHV-transformed cells with a SIRT1 inhibitor, nicotinamide (NAM), had the same effect as SIRT1 knockdown and knockout. NAM significantly inhibited cell proliferation in culture and colony formation in soft agar, and induced cell cycle arrest. Significantly, NAM inhibited the progression of tumors and extended the survival of mice in a KSHV-induced tumor model. Collectively, these results demonstrate that SIRT1 suppression of p27 is required for KSHV-induced tumorigenesis and identify a potential therapeutic target for KS.
Collapse
Affiliation(s)
- Meilan He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California Los Angeles, Los Angeles, CA, USA
| | - Hongfeng Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California Los Angeles, Los Angeles, CA, USA
| | - Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California Los Angeles, Los Angeles, CA, USA
| | - Rosemary Bai
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California Los Angeles, Los Angeles, CA, USA
| | - Heon Seok Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul, South Korea.,Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Sangsu Bae
- Center for Genome Engineering, Institute for Basic Science, Seoul, South Korea.,Present address: Department of Chemistry, Hanyang University, Seoul, South Korea
| | - Lu Che
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California Los Angeles, Los Angeles, CA, USA
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul, South Korea.,Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
26
|
Ding S, Bierbach U. Linker design for the modular assembly of multifunctional and targeted platinum(ii)-containing anticancer agents. Dalton Trans 2018; 45:13104-13. [PMID: 27251881 DOI: 10.1039/c6dt01399f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A versatile and efficient modular synthetic platform was developed for assembling multifunctional conjugates and targeted forms of platinum-(benz)acridines, a class of highly cytotoxic DNA-targeted hybrid agents. The synthetic strategy involved amide coupling between succinyl ester-modified platinum compounds (P1, P2) and a set of 11 biologically relevant primary and secondary amines (N1-N11). To demonstrate the feasibility and versatility of the approach, a structurally and functionally diverse range of amines was introduced. These include biologically active molecules, such as rucaparib (a PARP inhibitor), E/Z-endoxifen (an estrogen receptor antagonist), and a quinazoline-based tyrosine kinase inhibitor. Micro-scale reactions in Eppendorf tubes or on 96-well plates were used to screen for optimal coupling conditions in DMF solution with carbodiimide-, uronium-, and phosphonium-based compounds, as well as other common coupling reagents. Reactions with the phosphonium-based coupling reagent PyBOP produced the highest yields and gave the cleanest conversions. Furthermore, it was demonstrated that the chemistry can also be performed in aqueous media and is amenable to parallel synthesis based on multiple consecutive reactions in DMF in a "one-tube" format. In-line LC-MS was used to assess the stability of the conjugates in physiologically relevant buffers. Hydrolysis of the conjugates occurs at the ester moiety and is facilitated by the aquated metal moiety under low-chloride ion conditions. The rate of ester cleavage greatly depends on the nature of the amine component. Potential applications of the linker technology are discussed.
Collapse
Affiliation(s)
- S Ding
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina 27109, USA.
| | - U Bierbach
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina 27109, USA. and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA
| |
Collapse
|
27
|
Discovery of potent 2,4-difluoro-linker poly(ADP-ribose) polymerase 1 inhibitors with enhanced water solubility and in vivo anticancer efficacy. Acta Pharmacol Sin 2017; 38:1521-1532. [PMID: 28770827 DOI: 10.1038/aps.2017.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/04/2017] [Indexed: 12/11/2022]
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is overexpressed in a variety of cancers, especially in breast and ovarian cancers; tumor cells that are deficient in breast cancer gene 1/2 (BRCA1/2) are highly sensitive to PARP1 inhibition. In this study, we identified a series of 2,4-difluorophenyl-linker analogs (15-55) derived from olaparib as novel PARP1 inhibitors. Four potent analogs 17, 43, 47, and 50 (IC50=2.2-4.4 nmol/L) effectively inhibited the proliferation of Chinese hamster lung fibroblast V-C8 cells (IC50=3.2-37.6 nmol/L) in vitro, and showed specificity toward BRCA-deficient cells (SI=40-510). The corresponding hydrochloride salts 56 and 57 (based on 43 and 47) were highly water soluble in pH=1.0 buffered salt solutions (1628.2 μg/mL, 2652.5 μg/mL). In a BRCA1-mutated xenograft model, oral administration of compound 56 (30 mg·kg-1·d-1, for 21 d) exhibited more prominent tumor growth inhibition (96.6%) compared with the same dose of olaparib (56.3%); in a BRCA2-mutated xenograft model, oral administration of analog 43 (10 mg·kg-1·d-1, for 28 d) significantly inhibited tumor growth (69.0%) and had no negative effects on the body weights. Additionally, compound 56 exhibited good oral bioavailability (F=32.2%), similar to that of olaparib (F=45.4%). Furthermore, the free base 43 of the hydrochloride salt 56 exhibited minimal hERG inhibition activity (IC50=6.64 μmol/L). Collectively, these data demonstrate that compound 56 may be an excellent drug candidate for the treatment of cancer, particularly BRCA-deficient tumors.
Collapse
|
28
|
Chen W, Guo N, Qi M, Dai H, Hong M, Guan L, Huan X, Song S, He J, Wang Y, Xi Y, Yang X, Shen Y, Su Y, Sun Y, Gao Y, Chen Y, Ding J, Tang Y, Ren G, Miao Z, Li J. Discovery, mechanism and metabolism studies of 2,3-difluorophenyl-linker-containing PARP1 inhibitors with enhanced in vivo efficacy for cancer therapy. Eur J Med Chem 2017; 138:514-531. [DOI: 10.1016/j.ejmech.2017.06.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/30/2017] [Accepted: 06/25/2017] [Indexed: 10/19/2022]
|
29
|
|
30
|
Sherstyuk YV, Zakharenko AL, Kutuzov MM, Sukhanova MV, Lavrik OI, Silnikov VN, Abramova TV. Synthesis of a series of NAD+ analogues, potential inhibitors of PARP 1, using ADP conjugates functionalized at the terminal phosphate group. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2017. [DOI: 10.1134/s1068162017010095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Thorsell AG, Ekblad T, Karlberg T, Löw M, Pinto AF, Trésaugues L, Moche M, Cohen MS, Schüler H. Structural Basis for Potency and Promiscuity in Poly(ADP-ribose) Polymerase (PARP) and Tankyrase Inhibitors. J Med Chem 2016; 60:1262-1271. [PMID: 28001384 DOI: 10.1021/acs.jmedchem.6b00990] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Selective inhibitors could help unveil the mechanisms by which inhibition of poly(ADP-ribose) polymerases (PARPs) elicits clinical benefits in cancer therapy. We profiled 10 clinical PARP inhibitors and commonly used research tools for their inhibition of multiple PARP enzymes. We also determined crystal structures of these compounds bound to PARP1 or PARP2. Veliparib and niraparib are selective inhibitors of PARP1 and PARP2; olaparib, rucaparib, and talazoparib are more potent inhibitors of PARP1 but are less selective. PJ34 and UPF1069 are broad PARP inhibitors; PJ34 inserts a flexible moiety into hydrophobic subpockets in various ADP-ribosyltransferases. XAV939 is a promiscuous tankyrase inhibitor and a potent inhibitor of PARP1 in vitro and in cells, whereas IWR1 and AZ-6102 are tankyrase selective. Our biochemical and structural analysis of PARP inhibitor potencies establishes a molecular basis for either selectivity or promiscuity and provides a benchmark for experimental design in assessment of PARP inhibitor effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michael S Cohen
- Program in Chemical Biology and Department of Physiology and Pharmacology, Health & Science University , Portland, Oregon 97210, United States
| | | |
Collapse
|
32
|
Chenette EJ. The FEBS Journalpast … and present. FEBS J 2016; 283:4408-4411. [DOI: 10.1111/febs.13974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
33
|
McMahon M, Frangova TG, Henderson CJ, Wolf CR. Olaparib, Monotherapy or with Ionizing Radiation, Exacerbates DNA Damage in Normal Tissues: Insights from a New p21 Reporter Mouse. Mol Cancer Res 2016; 14:1195-1203. [PMID: 27604276 PMCID: PMC5136472 DOI: 10.1158/1541-7786.mcr-16-0108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/01/2016] [Accepted: 08/16/2016] [Indexed: 11/16/2022]
Abstract
Many drugs targeting the DNA damage response are being developed as anticancer therapies, either as single agents or in combination with ionizing radiation (IR) or other cytotoxic agents. Numerous clinical trials in this area are either in progress or planned. However, concerns remain about the potential of such treatments to increase toxicity to normal tissues. In order to address this issue, a novel reporter mouse line was created through the simultaneous incorporation of multiple reporters, β-galactosidase, and firefly luciferase, into the DNA damage-inducible p21 (CDKN1A) locus. The data demonstrate that in situ β-galactosidase staining facilitates high fidelity mapping of p21 expression across multiple organs and tissues at single-cell resolution, whereas the luciferase reporter permits noninvasive bioluminescent imaging of p21 expression. This model was used to determine the capacity of a number of DNA-damaging agents, including IR, cisplatin, and etoposide to induce p21 expression in normal tissues. In addition, the PARP inhibitor olaparib was examined alone or in combination with IR as well as cisplatin. A single exposure to olaparib alone caused DNA damage to cells in the mucosal layer lining mouse large intestine. It also exacerbated DNA damage induced in this organ and the kidney by coadministered IR. These studies suggest that olaparib has carcinogenic potential and illustrate the power of this new model to evaluate the safety of new therapeutic regimens involving combination therapies. IMPLICATIONS Olaparib causes DNA damage to normal tissues and might be a carcinogen. Mol Cancer Res; 14(12); 1195-203. ©2016 AACR.
Collapse
Affiliation(s)
- Michael McMahon
- University of Dundee, School of Medicine, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | - Tania G Frangova
- University of Dundee, School of Medicine, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | - Colin J Henderson
- University of Dundee, School of Medicine, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | - C Roland Wolf
- University of Dundee, School of Medicine, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
34
|
A versatile strategy for the design and synthesis of novel ADP conjugates and their evaluation as potential poly(ADP-ribose) polymerase 1 inhibitors. Mol Divers 2016; 21:101-113. [PMID: 27677737 DOI: 10.1007/s11030-016-9703-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/12/2016] [Indexed: 12/24/2022]
Abstract
A versatile strategy for the synthesis of [Formula: see text] mimetics was developed, involving an efficient pyrophosphate linkage formation in key conjugates containing a functional amino group which acts as useful reactive anchor for further derivatization. These [Formula: see text] mimetics consist of ADP conjugated through a diphosphate chain to an extended aliphatic linker bearing an aromatic acid residue. A number of conjugates containing aromatic carboxylic acids were found to inhibit poly(ADP-ribose) synthesis catalyzed by poly(ADP-ribose) polymerase-1 (PARP-1). A new class of potential PARP-1 inhibitors mimicking [Formula: see text], a substrate in the PARP-1 catalyzed reaction, was proposed.
Collapse
|
35
|
Kashinath K, Jadhav PD, Reddy DS. Total synthesis of an anticancer norsesquiterpene alkaloid isolated from the fungus Flammulina velutipes. Org Biomol Chem 2016; 12:4098-103. [PMID: 24769797 DOI: 10.1039/c4ob00300d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The first total synthesis of a norsesquiterpene alkaloid (R)-8-hydroxy-4,7,7-trimethyl-7,8-dihydrocyclopenta[e]isoindole-1,3(2H,6H)-dione, isolated from the mushroom-forming fungus Flammulina velutipes, in both racemic and enantiomeric pure forms, is reported. The (-)-enantiomer of the natural product has been synthesized from the D-(-)-pantolactone chiral pool. The synthesis features a one-pot, three-step reaction sequence comprising an enyne RCM/Diels-Alder/aromatization to construct the desired indane skeleton present in the natural product. Our synthesis further confirms the assigned structure and absolute configuration of the natural product.
Collapse
Affiliation(s)
- K Kashinath
- CSIR-National Chemical Laboratory, Division of Organic Chemistry, Dr. HomiBhabha Road, Pune, 411008, India.
| | | | | |
Collapse
|
36
|
Buchfellner A, Yurlova L, Nüske S, Scholz AM, Bogner J, Ruf B, Zolghadr K, Drexler SE, Drexler GA, Girst S, Greubel C, Reindl J, Siebenwirth C, Romer T, Friedl AA, Rothbauer U. A New Nanobody-Based Biosensor to Study Endogenous PARP1 In Vitro and in Live Human Cells. PLoS One 2016; 11:e0151041. [PMID: 26950694 PMCID: PMC4780744 DOI: 10.1371/journal.pone.0151041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 02/23/2016] [Indexed: 11/22/2022] Open
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is a key player in DNA repair, genomic stability and cell survival and it emerges as a highly relevant target for cancer therapies. To deepen our understanding of PARP biology and mechanisms of action of PARP1-targeting anti-cancer compounds, we generated a novel PARP1-affinity reagent, active both in vitro and in live cells. This PARP1-biosensor is based on a PARP1-specific single-domain antibody fragment (~ 15 kDa), termed nanobody, which recognizes the N-terminus of human PARP1 with nanomolar affinity. In proteomic approaches, immobilized PARP1 nanobody facilitates quantitative immunoprecipitation of functional, endogenous PARP1 from cellular lysates. For cellular studies, we engineered an intracellularly functional PARP1 chromobody by combining the nanobody coding sequence with a fluorescent protein sequence. By following the chromobody signal, we were for the first time able to monitor the recruitment of endogenous PARP1 to DNA damage sites in live cells. Moreover, tracing of the sub-nuclear translocation of the chromobody signal upon treatment of human cells with chemical substances enables real-time profiling of active compounds in high content imaging. Due to its ability to perform as a biosensor at the endogenous level of the PARP1 enzyme, the novel PARP1 nanobody is a unique and versatile tool for basic and applied studies of PARP1 biology and DNA repair.
Collapse
Affiliation(s)
| | | | - Stefan Nüske
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University, Munich, Germany
| | - Armin M. Scholz
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilians University, Munich, Germany
| | | | - Benjamin Ruf
- ChromoTek GmbH, IZB, Planegg, Martinsried, Germany
| | | | - Sophie E. Drexler
- Department of Radiation Oncology, Ludwig Maximilians University, Munich, Germany
| | - Guido A. Drexler
- Department of Radiation Oncology, Ludwig Maximilians University, Munich, Germany
| | - Stefanie Girst
- Institute for Applied Physics and Metrology, Bundeswehr University Munich, Neubiberg, Germany
| | - Christoph Greubel
- Institute for Applied Physics and Metrology, Bundeswehr University Munich, Neubiberg, Germany
| | - Judith Reindl
- Institute for Applied Physics and Metrology, Bundeswehr University Munich, Neubiberg, Germany
| | - Christian Siebenwirth
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Tina Romer
- ChromoTek GmbH, IZB, Planegg, Martinsried, Germany
- * E-mail:
| | - Anna A. Friedl
- Department of Radiation Oncology, Ludwig Maximilians University, Munich, Germany
- Clinical Cooperation Group ‘Personalized Radiotherapy of Head and Neck Cancer’, Helmholtz Center Munich, Neuherberg, Germany
| | - Ulrich Rothbauer
- Natural and Medical Institute at the University of Tuebingen, Reutlingen, Germany
- Pharmaceutical Biotechnology, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
37
|
Fang Y, Tranmer GK. Continuous flow photochemistry as an enabling synthetic technology: synthesis of substituted-6(5H)-phenanthridinones for use as poly(ADP-ribose) polymerase inhibitors. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00552c] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Methods utilizing continuous flow photochemistry have been developed for the synthesis of phenanthridinones via an intramolecular photochemical cyclization reaction in yields up to 99%, as a means to generate poly(ADP-ribose) polymerase inhibitors.
Collapse
Affiliation(s)
- Y. Fang
- College of Pharmacy, Faculty of Health Sciences
- University of Manitoba
- Winnipeg
- Canada
| | - G. K. Tranmer
- College of Pharmacy, Faculty of Health Sciences
- University of Manitoba
- Winnipeg
- Canada
- Department of Chemistry
| |
Collapse
|
38
|
Wang B, Chu D, Feng Y, Shen Y, Aoyagi-Scharber M, Post LE. Discovery and Characterization of (8S,9R)-5-Fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-2,7,8,9-tetrahydro-3H-pyrido[4,3,2-de]phthalazin-3-one (BMN 673, Talazoparib), a Novel, Highly Potent, and Orally Efficacious Poly(ADP-ribose) Polymerase-1/2 Inhibitor, as an Anticancer Agent. J Med Chem 2015; 59:335-57. [DOI: 10.1021/acs.jmedchem.5b01498] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Bing Wang
- BioMarin Pharmaceutical Inc., 105 Digital Drive, Novato, California 94949, United States
| | - Daniel Chu
- BioMarin Pharmaceutical Inc., 105 Digital Drive, Novato, California 94949, United States
| | - Ying Feng
- BioMarin Pharmaceutical Inc., 105 Digital Drive, Novato, California 94949, United States
| | - Yuqiao Shen
- BioMarin Pharmaceutical Inc., 105 Digital Drive, Novato, California 94949, United States
| | - Mika Aoyagi-Scharber
- BioMarin Pharmaceutical Inc., 105 Digital Drive, Novato, California 94949, United States
| | - Leonard E. Post
- BioMarin Pharmaceutical Inc., 105 Digital Drive, Novato, California 94949, United States
| |
Collapse
|
39
|
Salameh A, Dhein S. Strategies for Pharmacological Organoprotection during Extracorporeal Circulation Targeting Ischemia-Reperfusion Injury. Front Pharmacol 2015; 6:296. [PMID: 26733868 PMCID: PMC4686733 DOI: 10.3389/fphar.2015.00296] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/02/2015] [Indexed: 01/28/2023] Open
Abstract
Surgical correction of congenital cardiac malformations or aortocoronary bypass surgery in many cases implies the use of cardiopulmonary-bypass (CPB). However, a possible negative impact of CPB on internal organs such as brain, kidney, lung and liver cannot be neglected. In general, CPB initiates a systemic inflammatory response (SIRS) which is presumably caused by contact of blood components with the surface of CPB tubing. Moreover, during CPB the heart typically undergoes a period of cold ischemia, and the other peripheral organs a global low flow hypoperfusion. As a result, a plethora of pro-inflammatory mediators and cytokines is released activating different biochemical pathways, which finally may result in the occurrence of microthrombosis, microemboli, in depletion of coagulation factors and haemorrhagic diathesis besides typical ischemia-reperfusion injuries. In our review we will focus on possible pharmacological interventions in patients to decrease negative effects of CPB and to improve post-operative outcome with regard to heart and other organs like brain, kidney, or lung.
Collapse
Affiliation(s)
- Aida Salameh
- Clinic for Pediatric Cardiology, Heart Centre University of Leipzig Leipzig, Germany
| | - Stefan Dhein
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig Leipzig, Germany
| |
Collapse
|
40
|
Drenichev MS, Mikhailov SN. Poly(ADP-ribose)--a unique natural polymer structural features, biological role and approaches to the chemical synthesis. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2015; 34:258-76. [PMID: 25774719 DOI: 10.1080/15257770.2014.984073] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Poly(ADP-ribose) (PAR) is a natural polymer, taking part in numerous important cellular processes. Several enzymes are involved in biosynthesis and degradation of PAR. One of them, poly(ADP-ribose)polymerase-1 (PARP-1) is considered to be a perspective target for the design of new drugs, affecting PAR metabolism. The structure of PAR was established by enzymatic hydrolysis and further analysis of the products, but total chemical synthesis of PAR hasn't been described yet. Several approaches have been developed on the way to chemical synthesis of this unique biopolymer.
Collapse
Affiliation(s)
- Mikhail S Drenichev
- a Engelhardt Institute of Molecular Biology, Russian Academy of Sciences , Moscow , Russian Federation
| | | |
Collapse
|
41
|
Ekblad T, Schüler H. Sirtuins are Unaffected by PARP Inhibitors Containing Planar Nicotinamide Bioisosteres. Chem Biol Drug Des 2015; 87:478-82. [DOI: 10.1111/cbdd.12680] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 10/20/2015] [Accepted: 10/22/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Torun Ekblad
- Department of Medical Biochemistry and Biophysics; Karolinska Institutet; 17177 Stockholm Sweden
| | - Herwig Schüler
- Department of Medical Biochemistry and Biophysics; Karolinska Institutet; 17177 Stockholm Sweden
| |
Collapse
|
42
|
DNA Damage Signalling and Repair Inhibitors: The Long-Sought-After Achilles' Heel of Cancer. Biomolecules 2015; 5:3204-59. [PMID: 26610585 PMCID: PMC4693276 DOI: 10.3390/biom5043204] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
For decades, radiotherapy and chemotherapy were the two only approaches exploiting DNA repair processes to fight against cancer. Nowadays, cancer therapeutics can be a major challenge when it comes to seeking personalized targeted medicine that is both effective and selective to the malignancy. Over the last decade, the discovery of new targeted therapies against DNA damage signalling and repair has offered the possibility of therapeutic improvements in oncology. In this review, we summarize the current knowledge of DNA damage signalling and repair inhibitors, their molecular and cellular effects, and future therapeutic use.
Collapse
|
43
|
Rustandi RR, Hamm M, Loughney JW, Ha S. Detection of ADP ribosylation in PARP-1 and bacterial toxins using a capillary-based western system. Electrophoresis 2015; 36:2798-2804. [DOI: 10.1002/elps.201500173] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 11/10/2022]
Affiliation(s)
| | - Melissa Hamm
- Vaccine Analytical Development; Merck Research Laboratories; PA USA
| | - John W. Loughney
- Vaccine Analytical Development; Merck Research Laboratories; PA USA
| | - Sha Ha
- Vaccine Analytical Development; Merck Research Laboratories; PA USA
| |
Collapse
|
44
|
Passeri D, Camaioni E, Liscio P, Sabbatini P, Ferri M, Carotti A, Giacchè N, Pellicciari R, Gioiello A, Macchiarulo A. Concepts and Molecular Aspects in the Polypharmacology of PARP-1 Inhibitors. ChemMedChem 2015; 11:1219-26. [PMID: 26424664 DOI: 10.1002/cmdc.201500391] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Indexed: 11/08/2022]
Abstract
Recent years have witnessed a renewed interest in PARP-1 inhibitors as promising anticancer agents with multifaceted functions. Particularly exciting developments include the approval of olaparib (Lynparza) for the treatment of refractory ovarian cancer in patients with BRCA1/2 mutations, and the increasing understanding of the polypharmacology of PARP-1 inhibitors. The aim of this review article is to provide the reader with a comprehensive overview of the distinct levels of the polypharmacology of PARP-1 inhibitors, including 1) inter-family polypharmacology, 2) intra-family polypharmacology, and 3) multi-signaling polypharmacology. Progress made in gaining insight into the molecular basis of these multiple target-independent and target-dependent activities of PARP-1 inhibitors are discussed, with an outlook on the potential impact that a better understanding of polypharmacology may have in aiding the explanation as to why some drug candidates work better than others in clinical settings, albeit acting on the same target with similar inhibitory potency.
Collapse
Affiliation(s)
- Daniela Passeri
- TES Pharma S.r.l., via Palmiro Togliatti 20, 06073 Corciano, Perugia, Italy
| | - Emidio Camaioni
- Dipartimento di Scienze Farmaceutiche, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Paride Liscio
- TES Pharma S.r.l., via Palmiro Togliatti 20, 06073 Corciano, Perugia, Italy
| | - Paola Sabbatini
- TES Pharma S.r.l., via Palmiro Togliatti 20, 06073 Corciano, Perugia, Italy
| | - Martina Ferri
- Dipartimento di Scienze Farmaceutiche, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Andrea Carotti
- Dipartimento di Scienze Farmaceutiche, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Nicola Giacchè
- TES Pharma S.r.l., via Palmiro Togliatti 20, 06073 Corciano, Perugia, Italy
| | | | - Antimo Gioiello
- Dipartimento di Scienze Farmaceutiche, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Antonio Macchiarulo
- Dipartimento di Scienze Farmaceutiche, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy.
| |
Collapse
|
45
|
Schreiber V, Illuzzi G, Héberlé E, Dantzer F. De la découverte du poly(ADP-ribose) aux inhibiteurs PARP en thérapie du cancer. Bull Cancer 2015; 102:863-73. [DOI: 10.1016/j.bulcan.2015.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 07/28/2015] [Accepted: 07/28/2015] [Indexed: 02/05/2023]
|
46
|
Papeo G, Posteri H, Borghi D, Busel AA, Caprera F, Casale E, Ciomei M, Cirla A, Corti E, D'Anello M, Fasolini M, Forte B, Galvani A, Isacchi A, Khvat A, Krasavin MY, Lupi R, Orsini P, Perego R, Pesenti E, Pezzetta D, Rainoldi S, Riccardi-Sirtori F, Scolaro A, Sola F, Zuccotto F, Felder ER, Donati D, Montagnoli A. Discovery of 2-[1-(4,4-Difluorocyclohexyl)piperidin-4-yl]-6-fluoro-3-oxo-2,3-dihydro-1H-isoindole-4-carboxamide (NMS-P118): A Potent, Orally Available, and Highly Selective PARP-1 Inhibitor for Cancer Therapy. J Med Chem 2015. [PMID: 26222319 DOI: 10.1021/acs.jmedchem.5b00680] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The nuclear protein poly(ADP-ribose) polymerase-1 (PARP-1) has a well-established role in the signaling and repair of DNA and is a prominent target in oncology, as testified by the number of candidates in clinical testing that unselectively target both PARP-1 and its closest isoform PARP-2. The goal of our program was to find a PARP-1 selective inhibitor that would potentially mitigate toxicities arising from cross-inhibition of PARP-2. Thus, an HTS campaign on the proprietary Nerviano Medical Sciences (NMS) chemical collection, followed by SAR optimization, allowed us to discover 2-[1-(4,4-difluorocyclohexyl)piperidin-4-yl]-6-fluoro-3-oxo-2,3-dihydro-1H-isoindole-4-carboxamide (NMS-P118, 20by). NMS-P118 proved to be a potent, orally available, and highly selective PARP-1 inhibitor endowed with excellent ADME and pharmacokinetic profiles and high efficacy in vivo both as a single agent and in combination with Temozolomide in MDA-MB-436 and Capan-1 xenograft models, respectively. Cocrystal structures of 20by with both PARP-1 and PARP-2 catalytic domain proteins allowed rationalization of the observed selectivity.
Collapse
Affiliation(s)
- Gianluca Papeo
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Helena Posteri
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Daniela Borghi
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Alina A Busel
- Chemical Diversity Research Institute , Rabochaya St. 2 Khimki, Moscow Region 114401, Russia
| | - Francesco Caprera
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Elena Casale
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Marina Ciomei
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Alessandra Cirla
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Emiliana Corti
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Matteo D'Anello
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Marina Fasolini
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Barbara Forte
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Arturo Galvani
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Antonella Isacchi
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Alexander Khvat
- Chemical Diversity Research Institute , Rabochaya St. 2 Khimki, Moscow Region 114401, Russia
| | - Mikhail Y Krasavin
- Chemical Diversity Research Institute , Rabochaya St. 2 Khimki, Moscow Region 114401, Russia
| | - Rosita Lupi
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Paolo Orsini
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Rita Perego
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Enrico Pesenti
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | | | - Sonia Rainoldi
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | | | - Alessandra Scolaro
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Francesco Sola
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Fabio Zuccotto
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Eduard R Felder
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Daniele Donati
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| | - Alessia Montagnoli
- Oncology, Nerviano Medical Sciences Srl , Viale Pasteur 10, 20014 Nerviano, Milan, Italy
| |
Collapse
|
47
|
Shen Y, Aoyagi-Scharber M, Wang B. Trapping Poly(ADP-Ribose) Polymerase. J Pharmacol Exp Ther 2015; 353:446-57. [PMID: 25758918 DOI: 10.1124/jpet.114.222448] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/09/2015] [Indexed: 03/08/2025] Open
Abstract
Recent findings indicate that a major mechanism by which poly(ADP-ribose) polymerase (PARP) inhibitors kill cancer cells is by trapping PARP1 and PARP2 to the sites of DNA damage. The PARP enzyme-inhibitor complex "locks" onto damaged DNA and prevents DNA repair, replication, and transcription, leading to cell death. Several clinical-stage PARP inhibitors, including veliparib, rucaparib, olaparib, niraparib, and talazoparib, have been evaluated for their PARP-trapping activity. Although they display similar capacity to inhibit PARP catalytic activity, their relative abilities to trap PARP differ by several orders of magnitude, with the ability to trap PARP closely correlating with each drug's ability to kill cancer cells. In this article, we review the available data on molecular interactions between these clinical-stage PARP inhibitors and PARP proteins, and discuss how their biologic differences might be explained by the trapping mechanism. We also discuss how to use the PARP-trapping mechanism to guide the development of PARP inhibitors as a new class of cancer therapy, both for single-agent and combination treatments.
Collapse
Affiliation(s)
- Yuqiao Shen
- BioMarin Pharmaceutical Inc., Novato, California
| | | | - Bing Wang
- BioMarin Pharmaceutical Inc., Novato, California
| |
Collapse
|
48
|
Dal Piaz F, Ferro P, Vassallo A, Vasaturo M, Forte G, Chini MG, Bifulco G, Tosco A, De Tommasi N. Identification and mechanism of action analysis of the new PARP-1 inhibitor 2″-hydroxygenkwanol A. Biochim Biophys Acta Gen Subj 2015; 1850:1806-14. [PMID: 25999161 DOI: 10.1016/j.bbagen.2015.05.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/27/2015] [Accepted: 05/12/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Poly(ADP-ribose) polymerase 1 (PARP-1) activity has been implicated in the pathogenesis of numerous diseases as cancer, inflammation, diabetes and neurodegenerative disorders, therefore the research for new PARP-1 inhibitors is still an active area. METHODS To identify new potential PARP-1 inhibitors, we performed a screening of a small-molecule library consisting of polyphenols isolated from plants used in the traditional medicine, by Surface Plasmon Resonance (SPR). Biochemical and cellular assays were performed to confirm SPR results and select the promising candidate(s). Finally, limited proteolysis and ligand docking analyses allowed defining the protein region involved in the interaction with the putative inhibitor(s). RESULTS The dimeric spiro-flavonoid 2″-hydroxygenkwanol A, member of a relatively recently discovered class of flavonoids containing a spirane C-atom, has been identified as possible PARP-1 inhibitor. This compound showed a high affinity for the polymerase (KD: 0.32±0.05μM); moreover PARP-1 activity in the presence of 2″-hydroxygenkwanol A was significantly affected both when using the recombinant protein and when measuring the cellular effects. Finally, our study suggests this compound to efficiently interact with the protein catalytic domain, into the nicotine binding pocket. CONCLUSION 2″-hydroxygenkwanol A efficiently binds and inhibits PARP-1 at submicromolar concentrations, thus representing a promising lead for the design of a new class of PARP-1 modulators, useful as therapeutic agents and/or biochemical tools. GENERAL SIGNIFICANCE Our study has identified an additional class of plant molecules, the spiro-biflavonoids, with known beneficial pharmacological properties but with an unknown mechanism of action, as a possible novel class of PARP-1 activity inhibitors.
Collapse
Affiliation(s)
- Fabrizio Dal Piaz
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy.
| | - Piera Ferro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Antonio Vassallo
- Department of Sciences, University of Basilicata, Via Nazario Sauro 85, 85100 Potenza, SA, Italy
| | - Michele Vasaturo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Giovanni Forte
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Maria Giovanna Chini
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy.
| | - Nunziatina De Tommasi
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| |
Collapse
|
49
|
Towards small molecule inhibitors of mono-ADP-ribosyltransferases. Eur J Med Chem 2015; 95:546-51. [DOI: 10.1016/j.ejmech.2015.03.067] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/18/2015] [Accepted: 03/31/2015] [Indexed: 12/27/2022]
|
50
|
The Role of Poly(ADP-ribose) Polymerase-1 in Rheumatoid Arthritis. Mediators Inflamm 2015; 2015:837250. [PMID: 26339143 PMCID: PMC4539103 DOI: 10.1155/2015/837250] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/02/2015] [Indexed: 12/29/2022] Open
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear enzyme with a crucial role in the maintenance of genomic stability. In addition to the role of PARP-1 in DNA repair, multiple studies have also demonstrated its involvement in several inflammatory diseases, such as septic shock, asthma, atherosclerosis, and stroke, as well as in cancer. In these diseases, the pharmacological inhibition of PARP-1 has shown a beneficial effect, suggesting that PARP-1 regulates their inflammatory processes. In recent years, we have studied the role of PARP-1 in rheumatoid arthritis, as have other researchers, and the results have shown that PARP-1 has an important function in the development of this disease. This review summarizes current knowledge on the effects of PARP-1 in rheumatoid arthritis.
Collapse
|