101
|
Sahin ID, Jönsson JM, Hedenfalk I. Crizotinib and PARP inhibitors act synergistically by triggering apoptosis in high-grade serous ovarian cancer. Oncotarget 2019; 10:6981-6996. [PMID: 31857852 PMCID: PMC6916751 DOI: 10.18632/oncotarget.27363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the predominant and most lethal histological type of epithelial ovarian cancer. During the last few years, several new treatment options with PARP inhibitors have emerged. The FDA has approved the PARP inhibitor olaparib (Lynparza™) as maintenance treatment after first-line platinum-containing chemotherapy and olaparib, niraparib (Zejula™) and rucaparib (Rubraca™) are approved as maintenance therapies in the recurrent, platinum-sensitive setting; nevertheless, development of resistance limits their efficacy. In this study, new combinatorial treatment strategies targeting key signaling pathways were explored to enhance the activity of PARP inhibitors in HGSOC. Carboplatin, olaparib, niraparib, the PI3K inhibitor LY294002 and the c-Met inhibitor crizotinib were used for this investigation. PARP inhibitors and carboplatin alone and in combination caused accumulation of DNA double-strand breaks and G2/M cell cycle arrest. In contrast, crizotinib alone or in combination with PARP inhibitors induced accumulation of cells in sub-G1. Crizotinib together with either of the PARP inhibitors was more strongly synergistic than combinations with a PARP inhibitor and carboplatin or the PI3K inhibitor. Sequential combination of crizotinib and a PARP inhibitor resulted in activation of ATM/CHK2 and inhibition of c-Met pathways, contributing to a decrease in RAD51 levels and induction of caspase-3 dependent apoptotic cell death and suggesting that the combination of crizotinib with a PARP inhibitor may be considered and further explored as a new therapeutic strategy in HGSOC.
Collapse
Affiliation(s)
- Irem Durmaz Sahin
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
- School of Medicine, Koç University, Istanbul, Turkey
| | - Jenny-Maria Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| |
Collapse
|
102
|
Tomao F, Bardhi E, Di Pinto A, Sassu CM, Biagioli E, Petrella MC, Palaia I, Muzii L, Colombo N, Panici PB. Parp inhibitors as maintenance treatment in platinum sensitive recurrent ovarian cancer: An updated meta-analysis of randomized clinical trials according to BRCA mutational status. Cancer Treat Rev 2019; 80:101909. [DOI: 10.1016/j.ctrv.2019.101909] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 02/01/2023]
|
103
|
Wickstroem K, Karlsson J, Ellingsen C, Cruciani V, Kristian A, Hagemann UB, Bjerke RM, Ryan OB, Linden L, Mumberg D, Brands M, Cuthbertson A. Synergistic Effect of a HER2 Targeted Thorium-227 Conjugate in Combination with Olaparib in a BRCA2 Deficient Xenograft Model. Pharmaceuticals (Basel) 2019; 12:ph12040155. [PMID: 31618864 PMCID: PMC6958469 DOI: 10.3390/ph12040155] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/11/2019] [Accepted: 10/12/2019] [Indexed: 01/07/2023] Open
Abstract
Targeted thorium-227 conjugates (TTCs) represent a novel class of therapeutic radiopharmaceuticals for the treatment of cancer. TTCs consist of the alpha particle emitter thorium-227 complexed to a 3,2-hydroxypyridinone chelator conjugated to a tumor-targeting monoclonal antibody. The high energy and short range of the alpha particles induce potent and selective anti-tumor activity driven by the induction of DNA damage in the target cell. Methods: The efficacy of human epidermal growth factor receptor 2 (HER2)-TTC was tested in combination in vitro and in vivo with the poly ADP ribose polymerase (PARP) inhibitor (PARPi), olaparib, in the human colorectal adenocarcinoma isogenic cell line pair DLD-1 and the knockout variant DLD-1 BRCA2 -/- Results: The in vitro combination effects were determined to be synergistic in DLD-1 BRCA2 -/- and additive in DLD-1 parental cell lines. Similarly, the in vivo efficacy of the combination was determined to be synergistic only in the DLD-1 BRCA2 -/- xenograft model, with statistically significant tumor growth inhibition at a single TTC dose of 120 kBq/kg body weight (bw) and 50 mg/kg bw olaparib (daily, i.p. for 4 weeks), demonstrating comparable tumor growth inhibition to a single TTC dose of 600 kBq/kg bw. Conclusions: This study supports the further investigation of DNA damage response inhibitors in combination with TTCs as a new strategy for the effective treatment of mutation-associated cancers.
Collapse
Affiliation(s)
| | - Jenny Karlsson
- Thorium Conjugate Research, Bayer AS, Oslo 0283, Norway.
| | | | | | | | - Urs B Hagemann
- Bayer AG, Pharmaceuticals Division, Berlin 13353, Germany.
| | - Roger M Bjerke
- Thorium Conjugate Research, Bayer AS, Oslo 0283, Norway.
| | - Olav B Ryan
- Thorium Conjugate Research, Bayer AS, Oslo 0283, Norway.
| | - Lars Linden
- Bayer AG, Pharmaceuticals Division, Wuppertal 42113, Germany.
| | | | - Michael Brands
- Bayer AG, Pharmaceuticals Division, Berlin 13353, Germany.
| | | |
Collapse
|
104
|
Abstract
DNA topoisomerases are enzymes that catalyze changes in the torsional and flexural strain of DNA molecules. Earlier studies implicated these enzymes in a variety of processes in both prokaryotes and eukaryotes, including DNA replication, transcription, recombination, and chromosome segregation. Studies performed over the past 3 years have provided new insight into the roles of various topoisomerases in maintaining eukaryotic chromosome structure and facilitating the decatenation of daughter chromosomes at cell division. In addition, recent studies have demonstrated that the incorporation of ribonucleotides into DNA results in trapping of topoisomerase I (TOP1)–DNA covalent complexes during aborted ribonucleotide removal. Importantly, such trapped TOP1–DNA covalent complexes, formed either during ribonucleotide removal or as a consequence of drug action, activate several repair processes, including processes involving the recently described nuclear proteases SPARTAN and GCNA-1. A variety of new TOP1 inhibitors and formulations, including antibody–drug conjugates and PEGylated complexes, exert their anticancer effects by also trapping these TOP1–DNA covalent complexes. Here we review recent developments and identify further questions raised by these new findings.
Collapse
Affiliation(s)
- Mary-Ann Bjornsti
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294-0019, USA
| | - Scott H Kaufmann
- Departments of Oncology and Molecular Pharmacolgy & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
105
|
A Novel Tanshinone Analog Exerts Anti-Cancer Effects in Prostate Cancer by Inducing Cell Apoptosis, Arresting Cell Cycle at G2 Phase and Blocking Metastatic Ability. Int J Mol Sci 2019; 20:ijms20184459. [PMID: 31510010 PMCID: PMC6770861 DOI: 10.3390/ijms20184459] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/31/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PCa), an epithelial malignant tumor, is the second common cause of cancer death among males in western countries. Thus, the development of new strategies is urgently needed. Tanshinones isolated from Salvia miltiorrhiza and its synthetic analogs show various biological activities including anticancer effects. Among them, the tanshinone analog 2-((Glycine methyl ester)methyl)-naphtho (TC7) is the most effective, with better selectivity and lower toxicity. Therefore, in this work, the effect of TC7 against PCa was investigated through assessing the molecular mechanisms regulating the growth, metastasis, and invasion of PCa cells. Human PCa cells, PC3 and LNCAP, were used to evaluate TC7 mechanisms of action in vitro, while male BALB/c nude mice were used for in vivo experiments by subjecting each mouse to a subcutaneous injection of PC3 cells into the right flank to evaluate TC7 effects on tumor volume. Our in vitro results showed that TC7 inhibited cell proliferation by arresting the cell cycle at G2/M through the regulation of cyclin b1, p53, GADD45A, PLK1, and CDC2/cyclin b1. In addition, TC7 induced cell apoptosis by regulating apoptosis-associated genes such as p53, ERK1, BAX, p38, BCL-2, caspase-8, cleaved-caspase-8, PARP1, and the phosphorylation level of ERK1 and p38. Furthermore, it decreased DNA synthesis and inhibited the migration and invasion ability by regulating VEGF-1 and MMP-9 protein expression. Our in vivo evidence supports the conclusion that TC7 could be considered as a potential promising chemotherapeutic candidate in the treatment of PCa.
Collapse
|
106
|
Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in Ovarian Cancer: Are We There Yet? J Clin Oncol 2019; 37:2460-2471. [PMID: 31403857 DOI: 10.1200/jco.19.00508] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| | - Kunle Odunsi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - George Coukos
- Ludwig Institute for Cancer Research and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
107
|
Paller CJ, Antonarakis ES, Beer TM, Borno HT, Carlo MI, George DJ, Graff JN, Gupta S, Heath EI, Higano CS, McKay RR, Morgans AK, Patnaik A, Petrylak DP, Rettig MB, Ryan CJ, Taplin ME, Whang YE, Vinson J, Cheng HH, Giri VN. Germline Genetic Testing in Advanced Prostate Cancer; Practices and Barriers: Survey Results from the Germline Genetics Working Group of the Prostate Cancer Clinical Trials Consortium. Clin Genitourin Cancer 2019; 17:275-282.e1. [PMID: 31171481 PMCID: PMC6662206 DOI: 10.1016/j.clgc.2019.04.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/11/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Germline genetic testing increasingly identifies advanced prostate cancer (PCa) patients who are candidates for precision therapies. The Prostate Cancer Clinical Trials Consortium (PCCTC) established the Germline Genetics Working Group to provide guidance and resources to expand effective use of germline genetic testing. MATERIALS AND METHODS A 14-item questionnaire was e-mailed to academic oncologists at 43 PCCTC sites to collect information on germline genetic testing patterns, including patients considered, choice of assays, barriers slowing adoption, and actions to overcome barriers. RESULTS Twenty-six genitourinary oncologists from 19 institutions responded. Less than 40% (10 of 26) reported referring patients to a genetics department, whereas the remainder take personal responsibility for genetic testing and counseling; 16 (62%) consider testing all metastatic PCa patients, whereas 3 (12%) consider testing all patients with high-risk local disease; and 7 (27%) use multigene comprehensive pan-cancer panels, and 14 (54%) use smaller or targeted cancer gene panels. Barriers to widespread use are: (1) delayed or limited access to genetic counseling; (2) no insurance coverage; (3) lack of effective workflows; (4) insufficient educational materials; and (5) time and space constraints in busy clinics. The primary limitation was the <50% (19 of 43) response from PCCTC sites and no coverage of nonacademic cancer treatment facilities. CONCLUSION Joint efforts by urologists, oncologists, genetics counselors, insurers, and cancer centers can accelerate implementation of integrated germline genetic services for personalized treatment and clinical trial eligibility for PCa patients.
Collapse
Affiliation(s)
- Channing J Paller
- Sidney Kimmel Comprehensive Cancer Center, Department of Urology, Johns Hopkins University, Baltimore, MD
| | - Emmanuel S Antonarakis
- Sidney Kimmel Comprehensive Cancer Center, Department of Urology, Johns Hopkins University, Baltimore, MD
| | - Tomasz M Beer
- Division of Hematology/Medical Oncology School of Medicine, Oregon Health and Science University Knight Cancer Institute, Portland, OR
| | - Hala T Borno
- Department of Medicine, University of California San Francisco/Helen Diller Family Cancer, San Francisco, CA
| | - Maria I Carlo
- Sidney Kimmel Center for Prostate and Urologic Cancers, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, NC
| | - Julie N Graff
- Division of Hematology and Medical Oncology, VA Portland Health Care System/Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Shilpa Gupta
- Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis, MN
| | - Elisabeth I Heath
- Karmanos Cancer Institute and Department of Oncology, Wayne State University, Detroit, MI
| | - Celestia S Higano
- Fred Hutchinson Cancer Research Center and Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Rana R McKay
- Department of Medicine, University of California at San Diego Moores Cancer Center, La Jolla, CA
| | - Alicia K Morgans
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine at Northwestern University, Chicago, IL
| | - Akash Patnaik
- Department of Medicine, University of Chicago Comprehensive Cancer Center, Chicago, IL
| | | | - Matthew B Rettig
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Charles J Ryan
- Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis, MN
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA
| | - Young E Whang
- Department of Medicine, Hematology/Oncology, University of North Carolina Lineberger Cancer Center, Chapel Hill, NC
| | - Jacob Vinson
- Prostate Cancer Clinical Trials Consortium, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heather H Cheng
- Fred Hutchinson Cancer Research Center and Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA.
| | - Veda N Giri
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA.
| |
Collapse
|
108
|
Boussios S, Karihtala P, Moschetta M, Karathanasi A, Sadauskaite A, Rassy E, Pavlidis N. Combined Strategies with Poly (ADP-Ribose) Polymerase (PARP) Inhibitors for the Treatment of Ovarian Cancer: A Literature Review. Diagnostics (Basel) 2019; 9:E87. [PMID: 31374917 PMCID: PMC6787707 DOI: 10.3390/diagnostics9030087] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors are the first clinically approved drugs designed to exploit synthetic lethality, and were first introduced as a cancer-targeting strategy in 2005. They have led to a major change in the treatment of advanced ovarian cancer, and altered the natural history of a disease with extreme genetic complexity and defective DNA repair via homologous recombination (HR) pathway. Furthermore, additional mechanisms apart from breast related cancer antigens 1 and 2 (BRCA1/2) mutations can also result in HR pathway alterations and consequently lead to a clinical benefit from PARP inhibitors. Novel combinations of PARP inhibitors with other anticancer therapies are challenging, and better understanding of PARP biology, DNA repair mechanisms, and PARP inhibitor mechanisms of action is crucial. It seems that PARP inhibitor and biologic agent combinations appear well tolerated and clinically effective in both BRCA-mutated and wild-type cancers. They target differing aberrant and exploitable pathways in ovarian cancer, and may induce greater DNA damage and HR deficiency. The input of immunotherapy in ovarian cancer is based on the observation that immunosuppressive microenvironments can affect tumour growth, metastasis, and even treatment resistance. Several biologic agents have been studied in combination with PARP inhibitors, including inhibitors of vascular endothelial growth factor (VEGF; bevacizumab, cediranib), and PD-1 or PD-L1 (durvalumab, pembrolizumab, nivolumab), anti-CTLA4 monoclonal antibodies (tremelimumab), mTOR-(vistusertib), AKT-(capivasertib), and PI3K inhibitors (buparlisib, alpelisib), as well as MEK 1/2, and WEE1 inhibitors (selumetinib and adavosertib, respectively). Olaparib and veliparib have also been combined with chemotherapy with the rationale of disrupting base excision repair via PARP inhibition. Olaparib has been investigated with carboplatin and paclitaxel, whereas veliparib has been tested additionally in combination with temozolomide vs. pegylated liposomal doxorubicin, as well as with oral cyclophosphamide, and topoisomerase inhibitors. However, overlapping myelosuppression observed with PARP inhibitor and chemotherapy combinations requires further investigation with dose escalation studies. In this review, we discuss multiple clinical trials that are underway examining the antitumor activity of such combination strategies.
Collapse
Affiliation(s)
- Stergios Boussios
- Acute Oncology Assessment Unit, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK.
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece.
| | - Peeter Karihtala
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 22, 90029 Oulu, Finland
| | - Michele Moschetta
- Drug Development Unit, Sarah Cannon Research Institute, 93 Harley Street, London W1G 6AD, UK
| | - Afroditi Karathanasi
- Acute Oncology Assessment Unit, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK
| | - Agne Sadauskaite
- Department of Pharmacy, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, Kent, UK
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy Institut, 94805 Villejuif, France
- Department of Hematology-Oncology, Hotel Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, 166830 Beirut, Lebanon
| | - Nicholas Pavlidis
- Medical School, University of Ioannina, Stavros Niarchou Avenue, 45110 Ioannina, Greece
| |
Collapse
|
109
|
Dok R, Bamps M, Glorieux M, Zhao P, Sablina A, Nuyts S. Radiosensitization approaches for HPV-positive and HPV-negative head and neck squamous carcinomas. Int J Cancer 2019; 146:1075-1085. [PMID: 31283004 PMCID: PMC6973261 DOI: 10.1002/ijc.32558] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/12/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Radiotherapy is one of the most used treatment approaches for head and neck squamous cell carcinoma (HNSCC). Targeted inhibition of DNA repair machinery has the potential to improve treatment response by tailoring treatment to cancer cells lacking specific DNA repair pathways. Human papillomavirus (HPV)‐negative and HPV‐positive HNSCCs respond differently to radiotherapy treatment, suggesting that different approaches of DNA repair inhibition should be employed for these HNSCC groups. Here, we searched for optimal radiosensitization approaches for HPV‐positive and HPV‐negative HNSCCs by performing a targeted CRISPR‐Cas9 screen. We found that inhibition of base excision repair resulted in a better radiotherapy response in HPV‐positive HNSCC, which is correlated with upregulation of genes involved in base excision repair. In contrast, inhibition of nonhomologous end‐joining and mismatch repair showed strong effects in both HNSCC groups. We validated the screen results by combining radiotherapy with targeted inhibition of DNA repair in several preclinical models including primary and recurrent patient‐derived HNSCC xenografts. These findings underline the importance of stratifying HNSCC patients for combination treatments. What's new? The combination of radiotherapy and targeted inhibition of DNA repair pathways can potentially improve therapeutic response in patients with head and neck squamous cell carcinoma (HNSCC). Here, a targeted CRISPR‐Cas9 screen was used to identify optimal radiosensitization approaches for human papillomavirus (HPV)‐positive and HPV‐negative HNSCC. Inhibition of base excision repair was associated with improved radiotherapy response in HPV‐positive HNSCC cells. By comparison, inhibition of non‐homologous end‐joining and mismatch repair was effective in both HPV‐positive and HPV‐negative cells. The screen results were validated in patient‐derived xenograft models, suggesting that stratification of HNSCC patients by HPV status may benefit therapeutic outcome.
Collapse
Affiliation(s)
- Rüveyda Dok
- Department of OncologyKU Leuven, University of LeuvenLeuvenBelgium
| | - Marieke Bamps
- Department of OncologyKU Leuven, University of LeuvenLeuvenBelgium
| | - Mary Glorieux
- Department of OncologyKU Leuven, University of LeuvenLeuvenBelgium
| | - Peihua Zhao
- Department of OncologyKU Leuven, University of LeuvenLeuvenBelgium
- VIB‐KU Leuven Center for Cancer Biology, VIBLeuvenBelgium
| | - Anna Sablina
- Department of OncologyKU Leuven, University of LeuvenLeuvenBelgium
- VIB‐KU Leuven Center for Cancer Biology, VIBLeuvenBelgium
| | - Sandra Nuyts
- Department of OncologyKU Leuven, University of LeuvenLeuvenBelgium
- Department of Radiation OncologyUZ LeuvenLeuvenBelgium
| |
Collapse
|
110
|
Activation of PARP-1 by snoRNAs Controls Ribosome Biogenesis and Cell Growth via the RNA Helicase DDX21. Mol Cell 2019; 75:1270-1285.e14. [PMID: 31351877 DOI: 10.1016/j.molcel.2019.06.020] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 04/16/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022]
Abstract
PARP inhibitors (PARPi) prevent cancer cell growth by inducing synthetic lethality with DNA repair defects (e.g., in BRCA1/2 mutant cells). We have identified an alternative pathway for PARPi-mediated growth control in BRCA1/2-intact breast cancer cells involving rDNA transcription and ribosome biogenesis. PARP-1 binds to snoRNAs, which stimulate PARP-1 catalytic activity in the nucleolus independent of DNA damage. Activated PARP-1 ADP-ribosylates DDX21, an RNA helicase that localizes to nucleoli and promotes rDNA transcription when ADP-ribosylated. Treatment with PARPi or mutation of the ADP-ribosylation sites reduces DDX21 nucleolar localization, rDNA transcription, ribosome biogenesis, protein translation, and cell growth. The salient features of this pathway are evident in xenografts in mice and human breast cancer patient samples. Elevated levels of PARP-1 and nucleolar DDX21 are associated with cancer-related outcomes. Our studies provide a mechanistic rationale for efficacy of PARPi in cancer cells lacking defects in DNA repair whose growth is inhibited by PARPi.
Collapse
|
111
|
Liu Y, Song H, Song H, Feng X, Zhou C, Huo Z. Targeting autophagy potentiates the anti-tumor effect of PARP inhibitor in pediatric chronic myeloid leukemia. AMB Express 2019; 9:108. [PMID: 31309361 PMCID: PMC6629728 DOI: 10.1186/s13568-019-0836-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/08/2019] [Indexed: 01/23/2023] Open
Abstract
Due to its potent cytotoxicity in BRCA-mutated tumors, synthetic lethality elicited by poly (ADP-ribose) polymerase (PARP) inhibitor gives renewed enthusiasm to researching and developing anti-cancer therapies. Chronic myeloid leukemia (CML) is a type of cancers that starts in certain blood-forming cells of the bone marrow. Here, we showed that poly (ADP-ribose) polymerase (PARP) inhibitor talazoparib could induce a concentration-dependent cytotoxicity in CML cells derived from pediatric patients. During talazoparib treatment, autophagy was markedly activated, which was confirmed by the accumulation of autophagosomes, decrease of SQSTM1 and up-regulation of LC3-II. Inhibition of autophagy by pharmaceutical inhibitor chloroquine or small-interfering RNA siATG5 significantly increased the cytotoxicity of talazoparib in pediatric CML cells and elicited synergistic anti-tumor effect in patient-derived xenograft model. Our data demonstrated that autophagy played a cyto-protective role in talazoparib-treated pediatric CML and co-treatment with talazoparib and autophagy inhibitor could induce synergetic anti-tumor effect, providing novel insights for pediatric CML treatment.
Collapse
|
112
|
Lheureux S, Braunstein M, Oza AM. Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J Clin 2019; 69:280-304. [PMID: 31099893 DOI: 10.3322/caac.21559] [Citation(s) in RCA: 630] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is the second most common cause of gynecologic cancer death in women around the world. The outcomes are complicated, because the disease is often diagnosed late and composed of several subtypes with distinct biological and molecular properties (even within the same histological subtype), and there is inconsistency in availability of and access to treatment. Upfront treatment largely relies on debulking surgery to no residual disease and platinum-based chemotherapy, with the addition of antiangiogenic agents in patients who have suboptimally debulked and stage IV disease. Major improvement in maintenance therapy has been seen by incorporating inhibitors against poly (ADP-ribose) polymerase (PARP) molecules involved in the DNA damage-repair process, which have been approved in a recurrent setting and recently in a first-line setting among women with BRCA1/BRCA2 mutations. In recognizing the challenges facing the treatment of ovarian cancer, current investigations are enlaced with deep molecular and cellular profiling. To improve survival in this aggressive disease, access to appropriate evidence-based care is requisite. In concert, realizing individualized precision medicine will require prioritizing clinical trials of innovative treatments and refining predictive biomarkers that will enable selection of patients who would benefit from chemotherapy, targeted agents, or immunotherapy. Together, a coordinated and structured approach will accelerate significant clinical and academic advancements in ovarian cancer and meaningfully change the paradigm of care.
Collapse
Affiliation(s)
- Stephanie Lheureux
- Clinician Investigator, Bras Drug Development Program; and Staff Medical Oncologist and Gynecology Site Leader, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Assistant Professor, University of Toronto, Toronto, ON, Canada
| | - Marsela Braunstein
- Scientific Associate, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Amit M Oza
- Chief, Division of Medical Oncology and Hematology; Director, Cancer Clinical Research Unit; and Director, Bras Drug Development Program, Princess Margaret Cancer Centre, University Health Network and Mt. Sinai Health System, Toronto, ON, Canada
- Professor of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
113
|
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: 1.0] [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]
|
114
|
Miller AL, Fehling SC, Garcia PL, Gamblin TL, Council LN, van Waardenburg RCAM, Yang ES, Bradner JE, Yoon KJ. The BET inhibitor JQ1 attenuates double-strand break repair and sensitizes models of pancreatic ductal adenocarcinoma to PARP inhibitors. EBioMedicine 2019; 44:419-430. [PMID: 31126889 PMCID: PMC6604668 DOI: 10.1016/j.ebiom.2019.05.035] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND DNA repair deficiency accumulates DNA damage and sensitizes tumor cells to PARP inhibitors (PARPi). Based on our observation that the BET inhibitor JQ1 increases levels of DNA damage, we evaluated the efficacy of JQ1 + the PARPi olaparib in preclinical models of pancreatic ductal adenocarcinoma (PDAC). We also addressed the mechanism by which JQ1 increased DNA damage. METHODS The effect of JQ1 + olaparib on in vivo tumor growth was assessed with patient-derived xenograft (PDX) models of PDAC. Changes in protein expression were detected by immunohistochemistry and immunoblot. In vitro growth inhibition and mechanistic studies were done using alamarBlue, qRT-PCR, immunoblot, immunofluorescence, ChIP, and shRNA knockdown assays. FINDINGS Tumors exposed in vivo to JQ1 had higher levels of the DNA damage marker γH2AX than tumors exposed to vehicle only. Increases in γH2AX was concomitant with decreased expression of DNA repair proteins Ku80 and RAD51. JQ1 + olaparib inhibited the growth of PDX tumors greater than either drug alone. Mechanistically, ChIP assays demonstrated that JQ1 decreased the association of BRD4 and BRD2 with promoter loci of Ku80 and RAD51, and shRNA data showed that expression of Ku80 and RAD51 was BRD4- and BRD2-dependent in PDAC cell lines. INTERPRETATION The data are consistent with the hypothesis that JQ1 confers a repair deficient phenotype and the consequent accumulation of DNA damage sensitizes PDAC cells to PARPi. Combinations of BET inhibitors with PARPi may provide a novel strategy for treating PDAC. FUND: NIH grants R01CA208272 and R21CA205501; UAB CMB T32 predoctoral training grant.
Collapse
Affiliation(s)
- Aubrey L Miller
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, USA
| | - Samuel C Fehling
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, USA
| | - Patrick L Garcia
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, USA
| | - Tracy L Gamblin
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, USA
| | - Leona N Council
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham, NP3551 North Pavilion UAB Hospital, Birmingham, AL, USA; The Birmingham Veterans Administration Medical Center, 700 19(th) St S, Birmingham, AL, USA
| | - Robert C A M van Waardenburg
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, USA
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham, Hazelrig Salter Radiation Oncology Center, 1700 6(th) Avenue S, Birmingham, AL, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Karina J Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, USA.
| |
Collapse
|
115
|
Corrado G, Palluzzi E, Bottoni C, Pietragalla A, Salutari V, Ghizzoni V, Distefano M, Scambia G, Ferrandina G. New medical approaches in advanced ovarian cancer. Minerva Med 2019; 110:367-384. [PMID: 31124637 DOI: 10.23736/s0026-4806.19.06139-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer death among women and the most lethal gynecologic malignancy. Most women with advanced epithelial ovarian cancer will experience many episodes of recurrent disease with progressively shorter disease-free intervals. For women whose disease continues to respond to platinum-based drugs, the disease can often be controlled for 5 years or more. Enormous progress has been made in the management of this disease, and new targeted treatments such as antiangiogenic drugs, poly(adenosine diphosphate-ribose) polymerase inhibitors, and immune checkpoint inhibitors offer potential for improved survival. A variety of combination strategies are being evaluated to leverage these agents. The objective of this review is to summarize results from clinical trials that tested cytotoxic drugs and target strategies for the treatment of ovarian cancer with particular attention to Phase III and ongoing trials.
Collapse
Affiliation(s)
- Giacomo Corrado
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy -
| | - Eleonora Palluzzi
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy
| | - Carolina Bottoni
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy
| | - Antonella Pietragalla
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy
| | - Vanda Salutari
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy
| | - Viola Ghizzoni
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy
| | - Mariagrazia Distefano
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy
| | - Giovanni Scambia
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy.,Division of Gynecologic Oncology, Sacred Heart Catholic University, Policlinico A. Gemelli Foundation, Rome, Italy
| | - Gabriella Ferrandina
- Division of Gynecologic Oncology, Department of Women and Children's Health, A. Gemelli University Hospital and Institute for Research and Care, Rome, Italy.,Division of Gynecologic Oncology, Sacred Heart Catholic University, Policlinico A. Gemelli Foundation, Rome, Italy
| |
Collapse
|
116
|
Inherited predisposition to malignant mesothelioma and overall survival following platinum chemotherapy. Proc Natl Acad Sci U S A 2019; 116:9008-9013. [PMID: 30975761 DOI: 10.1073/pnas.1821510116] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Survival from malignant mesothelioma, particularly pleural mesothelioma, is very poor. For patients with breast, ovarian, or prostate cancers, overall survival is associated with increased sensitivity to platinum chemotherapy due to loss-of-function mutations in DNA repair genes. The goal of this project was to evaluate, in patients with malignant mesothelioma, the relationship between inherited loss-of-function mutations in DNA repair and other tumor suppressor genes and overall survival following platinum chemotherapy. Patients with histologically confirmed malignant mesothelioma were evaluated for inherited mutations in tumor suppressor genes. Survival was evaluated with respect to genotype and site of mesothelioma. Among 385 patients treated with platinum chemotherapy, median overall survival was significantly longer for patients with loss-of-function mutations in any of the targeted genes compared with patients with no such mutation (P = 0.0006). The effect of genotype was highly significant for patients with pleural mesothelioma (median survival 7.9 y versus 2.4 y, P = 0.0012), but not for patients with peritoneal mesothelioma (median survival 8.2 y versus 5.4 y, P = 0.47). Effect of patient genotype on overall survival, measured at 3 y, remained independently significant after adjusting for gender and age at diagnosis, two other known prognostic factors. Patients with pleural mesothelioma with inherited mutations in DNA repair and other tumor suppressor genes appear to particularly benefit from platinum chemotherapy compared with patients without inherited mutations. These patients may also benefit from other DNA repair targeted therapies such as poly-ADP ribose polymerase (PARP) inhibitors.
Collapse
|
117
|
Want MY, Konstorum A, Huang RY, Jain V, Matsueda S, Tsuji T, Lugade A, Odunsi K, Koya R, Battaglia S. Neoantigens retention in patient derived xenograft models mediates autologous T cells activation in ovarian cancer. Oncoimmunology 2019; 8:e1586042. [PMID: 31069153 PMCID: PMC6492964 DOI: 10.1080/2162402x.2019.1586042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/17/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) has an overall modest number of mutations that facilitate a functional immune infiltrate able to recognize tumor mutated antigens, or neoantigens. Although patient-derived xenografts (PDXs) can partially model the tumor mutational load and mimic response to chemotherapy, no study profiled a neoantigen-driven response in OC PDXs. Here we demonstrate that the genomic status of the primary tumor from an OC patient can be recapitulated in vivo in a PDX model, with the goal of defining autologous T cells activation by neoantigens using in silico, in vitro and in vivo approaches. By profiling the PDX mutanome we discovered three main clusters of mutations defining the expansion, retraction or conservation of tumor clones based on their variant allele frequencies (VAF). RNASeq analyses revealed a strong functional conservation between the primary tumor and PDXs, highlighted by the upregulation of antigen presenting pathways. We tested in vitro a set of 30 neoantigens for recognition by autologous T cells and identified a core of six neoantigens that define a potent T cell activation able to slow tumor growth in vivo. The pattern of recognition of these six neoantigens indicates the pre-existence of anti-tumor immunity in the patient. To evaluate the breadth of T cell activation, we performed single cell sequencing profiling the TCR repertoire upon stimulation with neoantigenic moieties and identified sequence motifs that define an oligoclonal and autologous T cell response. Overall, these results indicate that OC PDXs can be a valid tool to model OC response to immunotherapy.
Collapse
Affiliation(s)
| | - Anna Konstorum
- Center for Quantitative Medicine, UConn Health, Farmington, CT, USA
| | - Ruea-Yea Huang
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Vaibhav Jain
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Satoko Matsueda
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Takemasa Tsuji
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Amit Lugade
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kunle Odunsi
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Richard Koya
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sebastiano Battaglia
- Center For Immunotherapy, Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Cancer Genetics and Genomics, Roswell Park, Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
118
|
Lheureux S, Gourley C, Vergote I, Oza AM. Epithelial ovarian cancer. Lancet 2019; 393:1240-1253. [PMID: 30910306 DOI: 10.1016/s0140-6736(18)32552-2] [Citation(s) in RCA: 999] [Impact Index Per Article: 199.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/02/2018] [Accepted: 10/10/2018] [Indexed: 12/17/2022]
Abstract
Epithelial ovarian cancer generally presents at an advanced stage and is the most common cause of gynaecological cancer death. Treatment requires expert multidisciplinary care. Population-based screening has been ineffective, but new approaches for early diagnosis and prevention that leverage molecular genomics are in development. Initial therapy includes surgery and adjuvant therapy. Epithelial ovarian cancer is composed of distinct histological subtypes with unique genomic characteristics, which are improving the precision and effectiveness of therapy, allowing discovery of predictors of response such as mutations in breast cancer susceptibility genes BRCA1 and BRCA2, and homologous recombination deficiency for DNA damage response pathway inhibitors or resistance (cyclin E1). Rapidly evolving techniques to measure genomic changes in tumour and blood allow for assessment of sensitivity and emergence of resistance to therapy, and might be accurate indicators of residual disease. Recurrence is usually incurable, and patient symptom control and quality of life are key considerations at this stage. Treatments for recurrence have to be designed from a patient's perspective and incorporate meaningful measures of benefit. Urgent progress is needed to develop evidence and consensus-based treatment guidelines for each subgroup, and requires close international cooperation in conducting clinical trials through academic research groups such as the Gynecologic Cancer Intergroup.
Collapse
Affiliation(s)
- Stephanie Lheureux
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Edinburgh Cancer Research UK Centre, MRC IGMM, University of Edinburgh, Western General Hospital, Edinburgh, UK; Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | - Ignace Vergote
- Division of Gynaecological Oncology, Leuven Cancer Institute, Department of Gynaecology and Obstetrics, Universitaire Ziekenhuizen Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Amit M Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
119
|
MYC status as a determinant of synergistic response to Olaparib and Palbociclib in ovarian cancer. EBioMedicine 2019; 43:225-237. [PMID: 30898650 PMCID: PMC6557734 DOI: 10.1016/j.ebiom.2019.03.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/18/2022] Open
Abstract
Background While PARP inhibitors and CDK4/6 inhibitors, the two classes of FDA-approved agents, have shown promising clinical benefits, there is an urgent need to develop new therapeutic strategies to improve clinical response. Meanwhile, extending the utility of these inhibitors beyond their respective molecularly defined cancer types is challenging and will likely require biomarkers predictive of treatment response especially when used in a combination drug development setting. Methods The effects of PARP inhibitor Olaparib and CDK4/6 inhibitor Palbociclib on ovarian cancer cells lines including those of high-grade serous histology were examined in vitro and in vivo. We investigated the molecular mechanism underlying the synergistic effects of drug combination. Findings We show for the first time that combining PARP and CDK4/6 inhibition has synergistic effects against MYC overexpressing ovarian cancer cells both in vitro and in vivo. Mechanistically, we find that Palbociclib induces homologous recombination (HR) deficiency through downregulation of MYC-regulated HR pathway genes, causing synthetic lethality with Olaparib. We further demonstrate that MYC expression determines sensitivity to combinatorial treatment with Olaparib and Palbociclib. Interpretation Our data provide a rationale for clinical evaluation of therapeutic synergy of these two classes of inhibitors in ovarian cancer patients whose tumors show high MYC expression and who do not respond to PARP inhibitors or CDK4/6 inhibitors monotherapies. Fund This work was supported by the National Natural Science Foundation of China [81672575, 81874111, 81472447 to HC; 81572586 and 81372853 to PL], and the Liaoning Provincial Key Basic Research Program for Universities [LZ2017002 to HC].
Collapse
|
120
|
Hodara E, Morrison G, Cunha A, Zainfeld D, Xu T, Xu Y, Dempsey PW, Pagano PC, Bischoff F, Khurana A, Koo S, Ting M, Cotter PD, Moore MW, Gunn S, Usher J, Rabizadeh S, Danenberg P, Danenberg K, Carpten J, Dorff T, Quinn D, Goldkorn A. Multiparametric liquid biopsy analysis in metastatic prostate cancer. JCI Insight 2019; 4:125529. [PMID: 30702443 DOI: 10.1172/jci.insight.125529] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/25/2019] [Indexed: 12/11/2022] Open
Abstract
Molecular profiling of prostate cancer with liquid biopsies, such as circulating tumor cells (CTCs) and cell-free nucleic acid analysis, yields informative yet distinct data sets. Additional insights may be gained by simultaneously interrogating multiple liquid biopsy components to construct a more comprehensive molecular disease profile. We conducted an initial proof-of-principle study aimed at piloting this multiparametric approach. Peripheral blood samples from men with metastatic castrate-resistant prostate cancer were analyzed simultaneously for CTC enumeration, single-cell copy number variations, CTC DNA and matched cell-free DNA mutations, and plasma cell-free RNA levels of androgen receptor (AR) and AR splice variant (ARV7). In addition, liquid biopsies were compared with matched tumor profiles when available, and a second liquid biopsy was drawn and analyzed at disease progression in a subset of patients. In this manner, multiparametric liquid biopsy profiles were successfully generated for each patient and time point, demonstrating the feasibility of this approach and highlighting shared as well as unique cancer-relevant alterations. With further refinement and validation in large cohorts, multiparametric liquid biopsies can optimally integrate disparate but clinically informative data sets and maximize their utility for molecularly directed, real-time patient management.
Collapse
Affiliation(s)
- Emmanuelle Hodara
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - Gareth Morrison
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - Alexander Cunha
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - Daniel Zainfeld
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - Tong Xu
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - Yucheng Xu
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - Paul W Dempsey
- Cynvenio Biosystems Inc., Westlake Village, California, USA
| | - Paul C Pagano
- Cynvenio Biosystems Inc., Westlake Village, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | - John Carpten
- Department of Translational Genomics, USC Keck School of Medicine and NCCC, Los Angeles, California, USA
| | - Tanya Dorff
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - David Quinn
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| | - Amir Goldkorn
- Department of Medicine, University of Southern California (USC) Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), Los Angeles, California, USA
| |
Collapse
|
121
|
Tan GX, Wang XN, Tang YY, Cen WJ, Li ZH, Wang GC, Jiang JW, Wang XC. PP-22 promotes autophagy and apoptosis in the nasopharyngeal carcinoma cell line CNE-2 by inducing endoplasmic reticulum stress, downregulating STAT3 signaling, and modulating the MAPK pathway. J Cell Physiol 2019; 234:2618-2630. [PMID: 30191969 DOI: 10.1002/jcp.27076] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/28/2018] [Indexed: 12/24/2022]
Abstract
Paris polyphylla var. yunnanensis, named Chong Lou, is considered an antitumor substance. In this study, we investigated the effect of PP-22, a monomer purified from P. polyphylla var. yunnanensis, on the nasopharyngeal carcinoma cell line CNE-2 in vitro. The results showed that PP-22 could inhibit the proliferation of CNE-2 cells via the induction of apoptosis, with evidence of the characteristic morphological changes in the apoptosis in the nucleus and an increase in Annexin V-positive cells. In addition, we found that PP-22 could activate the p38 mitogen-activated protein kinase (MAPK) pathway and that this activation was reversed by SB203580, a specific inhibitor of the p38 MAPK pathway. In contrast, PP-22 promoted apoptosis via an intrinsic pathway, including the endoplasmic reticulum stress pathway, in a caspase-dependent manner. A further study showed that PP-22 also induced apoptosis by downregulating the signal transducers and activators of transcription 3 (STAT3) pathway, and the inhibitory effect was also confirmed by STAT3 small interfering RNA. In addition, PP-22 could promote autophagy by inhibiting the extracellular regulated protein kinases (ERK) pathway. And autophagy plays a protective role against apoptosis. Together, these data show that PP-22 promotes autophagy and apoptosis in the nasopharyngeal carcinoma CNE-2 cell line.
Collapse
Affiliation(s)
- Gui-Xiang Tan
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Gastroenterology, People's Hospital of Qingyuan, Guangdong, China
| | - Xin-Ning Wang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yun-Yun Tang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Wan-Jing Cen
- Department of Stomatology, Guangzhou Development District Hospital, Guangzhou, China
| | - Zhen-Hua Li
- Translation Research Institute, Jinan University, Guangzhou, China
| | - Guo-Cai Wang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jian-Wei Jiang
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, China
| | - Xi-Cheng Wang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
122
|
Sengar A, Vandana J, Chambers VS, Di Antonio M, Winnerdy F, Balasubramanian S, Phan AT. Structure of a (3+1) hybrid G-quadruplex in the PARP1 promoter. Nucleic Acids Res 2019; 47:1564-1572. [PMID: 30551210 PMCID: PMC6379715 DOI: 10.1093/nar/gky1179] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/02/2018] [Accepted: 12/12/2018] [Indexed: 01/08/2023] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) has emerged as an attractive target for cancer therapy due to its key role in DNA repair processes. Inhibition of PARP1 in BRCA-mutated cancers has been observed to be clinically beneficial. Recent genome-mapping experiments have identified a non-canonical G-quadruplex-forming sequence containing bulges within the PARP1 promoter. Structural features, like bulges, provide opportunities for selective chemical targeting of the non-canonical G-quadruplex structure within the PARP1 promoter, which could serve as an alternative therapeutic approach for the regulation of PARP1 expression. Here we report the G-quadruplex structure formed by a 23-nucleotide G-rich sequence in the PARP1 promoter. Our study revealed a three-layered intramolecular (3+1) hybrid G-quadruplex scaffold, in which three strands are oriented in one direction and the fourth in the opposite direction. This structure exhibits unique structural features such as an adenine bulge and a G·G·T base triple capping structure formed between the central edgewise loop, propeller loop and 5' flanking terminal. Given the highly important role of PARP1 in DNA repair and cancer intervention, this structure presents an attractive opportunity to explore the therapeutic potential of PARP1 inhibition via G-quadruplex DNA targeting.
Collapse
Affiliation(s)
- Anjali Sengar
- School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - J Jeya Vandana
- School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Vicki S Chambers
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Marco Di Antonio
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Fernaldo Richtia Winnerdy
- School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Shankar Balasubramanian
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Anh Tuân Phan
- School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| |
Collapse
|
123
|
NADP + is an endogenous PARP inhibitor in DNA damage response and tumor suppression. Nat Commun 2019; 10:693. [PMID: 30741937 PMCID: PMC6370829 DOI: 10.1038/s41467-019-08530-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 12/05/2018] [Accepted: 12/30/2018] [Indexed: 11/15/2022] Open
Abstract
ADP-ribosylation is a unique posttranslational modification catalyzed by poly(ADP-ribose) polymerases (PARPs) using NAD+ as ADP-ribose donor. PARPs play an indispensable role in DNA damage repair and small molecule PARP inhibitors have emerged as potent anticancer drugs. However, to date, PARP inhibitor treatment has been restricted to patients with BRCA1/2 mutation-associated breast and ovarian cancer. One of the major challenges to extend the therapeutic potential of PARP inhibitors to other cancer types is the absence of predictive biomarkers. Here, we show that ovarian cancer cells with higher level of NADP+, an NAD+ derivative, are more sensitive to PARP inhibitors. We demonstrate that NADP+ acts as a negative regulator and suppresses ADP-ribosylation both in vitro and in vivo. NADP+ impairs ADP-ribosylation-dependent DNA damage repair and sensitizes tumor cell to chemically synthesized PARP inhibitors. Taken together, our study identifies NADP+ as an endogenous PARP inhibitor that may have implications in cancer treatment. Cancer cells respond differently to inhibitors of Poly (ADP-ribose) polymerase. Here the authors reveal that ovarian cancer cells with higher cellular NADP+ levels are more sensitive to clinically relevant PARP1 inhibitors and show that NADP+ act as an endogenous inhibitor of PARP enzymes.
Collapse
|
124
|
Houts AC, Olufade T, Shenolikar R, Walker MS, Schwartzberg LS. Treatment patterns, clinical outcomes, health resource utilization, and cost in patients with BRCA-mutated metastatic breast cancer treated in community oncology settings. Cancer Treat Res Commun 2019; 19:100121. [PMID: 30785027 DOI: 10.1016/j.ctarc.2019.100121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/14/2019] [Accepted: 02/05/2019] [Indexed: 06/09/2023]
Abstract
PURPOSE This retrospective study of community oncology patients with breast cancer gene (BRCA)-mutated metastatic breast cancer (MBC) examined treatment outcomes and health resource utilization (HRU) and costs for a sample of patients with human epidermal growth factor receptor 2 (HER2)-negative disease who were either hormone receptor positive (HR+) or triple negative breast cancer (TNBC). METHODS Evidence from the Vector Oncology Data Warehouse, a repository of electronic medical records/billing data and provider notes, was analyzed. Treatment outcomes were progression-free survival (PFS) and overall survival (OS) from start of first-line therapy in the metastatic setting. HRU and cost measures were collected from the time of MBC diagnosis to end of the record. HRU included hospitalizations, emergency room visits, infused/parenteral supportive care drugs, and outpatient visits. Costs were computed both as total and monthly costs. RESULTS 57 HR+ and 57 TNBC patients (2013-2015) met inclusion criteria. Eight TNBC patients did not get treatment. HR+ patients had median first line PFS of 12.1 months and TNBC patients had 6.1 months. HR+ patients had median OS from start of first line of 38.4 months, and TNBC patients had 23.4 months. Rate of use of infused/parenteral supportive care drugs was 25.5% overall and 36.7% among TNBC patients with 15.8% among HR+ patients. CONCLUSION There is an unmet need in BRCA-mutated patients with MBC, including those with HR+ and TNBC disease. The unmet need among TNBC patients was most evident in that 12% were not treated and TNBC patients appeared to have poor treatment outcomes. MICRO ABSTRACT Reviewed medical records for outcomes, resource utilization, and costs in 114 community patients with BRCA mutated metastatic breast cancer. 57 hormone positive (HP); 57 triple negative (TN). RESULTS median PFS: 12.1 months HP; 6.1 TN. HP OS was 38.4; TN 23.4. Rate of infused supportive care drugs: 25.5% HP; 36.7% TN. Patients with TN disease need better therapeutic options.
Collapse
MESH Headings
- Adult
- Aged
- Antineoplastic Combined Chemotherapy Protocols/economics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- BRCA1 Protein/genetics
- BRCA2 Protein/genetics
- Biomarkers, Tumor/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/economics
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/economics
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Lobular/drug therapy
- Carcinoma, Lobular/economics
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/secondary
- Community Health Centers
- Costs and Cost Analysis
- Female
- Follow-Up Studies
- Health Care Rationing/economics
- Health Care Rationing/statistics & numerical data
- Humans
- Inflammatory Breast Neoplasms/drug therapy
- Inflammatory Breast Neoplasms/economics
- Inflammatory Breast Neoplasms/genetics
- Inflammatory Breast Neoplasms/secondary
- Middle Aged
- Mutation
- Retrospective Studies
- Survival Rate
- Treatment Outcome
Collapse
Affiliation(s)
- Arthur C Houts
- Vector Oncology, 6555 Quince, Suite 400, Memphis, TN 38119, United States
| | - Tope Olufade
- AstraZeneca, 101 Orchard Ridge Dr. (3233D), Gaithersburg, MD 20878, United States
| | - Rahul Shenolikar
- AstraZeneca, 101 Orchard Ridge Dr. (3233D), Gaithersburg, MD 20878, United States
| | - Mark S Walker
- Vector Oncology, 6555 Quince, Suite 400, Memphis, TN 38119, United States.
| | - Lee S Schwartzberg
- West Cancer Center, 7945 Wolf River Boulevard, Germantown, TN 38138, United States
| |
Collapse
|
125
|
Ulm M, Ramesh AV, McNamara KM, Ponnusamy S, Sasano H, Narayanan R. Therapeutic advances in hormone-dependent cancers: focus on prostate, breast and ovarian cancers. Endocr Connect 2019; 8:R10-R26. [PMID: 30640710 PMCID: PMC6365668 DOI: 10.1530/ec-18-0425] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
Hormonal cancers affect over 400,000 men and women and contribute collectively to over 100,000 deaths in the United States alone. Thanks to advances in the understanding of these cancers at the molecular level and to the discovery of several disease-modifying therapeutics, the last decade has seen a plateauing or even a decreasing trend in the number of deaths from these cancers. These advanced therapeutics not only effectively slow the growth of hormonal cancers, but also provide an insight on how these cancers become refractory and evolve as an altogether distinct subset. This review summarizes the current therapeutic trends in hormonal cancers, with focus on prostate, breast and ovarian cancers. The review discusses the clinical drugs being used now, promising molecules that are going through various stages of development and makes some predictions on how the therapeutic landscape will shift in the next decade.
Collapse
Affiliation(s)
- Michael Ulm
- University of Tennessee Health Science Center, Memphis, Tennessee, USA
- West Cancer Center, Memphis, Tennessee, USA
| | | | | | - Suriyan Ponnusamy
- University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | - Ramesh Narayanan
- University of Tennessee Health Science Center, Memphis, Tennessee, USA
- West Cancer Center, Memphis, Tennessee, USA
| |
Collapse
|
126
|
Hurley RM, Wahner Hendrickson AE, Visscher DW, Ansell P, Harrell MI, Wagner JM, Negron V, Goergen KM, Maurer MJ, Oberg AL, Meng XW, Flatten KS, De Jonge MJA, Van Herpen CD, Gietema JA, Koornstra RHT, Jager A, den Hollander MW, Dudley M, Shepherd SP, Swisher EM, Kaufmann SH. 53BP1 as a potential predictor of response in PARP inhibitor-treated homologous recombination-deficient ovarian cancer. Gynecol Oncol 2019. [PMID: 30686551 DOI: 10.1016/j.ygyno.2019.01.015]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Poly(ADP-ribose) polymerase (PARP) inhibitors have shown substantial activity in homologous recombination- (HR-) deficient ovarian cancer and are undergoing testing in other HR-deficient tumors. For reasons that are incompletely understood, not all patients with HR-deficient cancers respond to these agents. Preclinical studies have demonstrated that changes in alternative DNA repair pathways affect PARP inhibitor (PARPi) sensitivity in ovarian cancer models. This has not previously been assessed in the clinical setting. METHODS Clonogenic and plasmid-based HR repair assays were performed to compare BRCA1-mutant COV362 ovarian cancer cells with or without 53BP1 gene deletion. Archival biopsies from ovarian cancer patients in the phase I, open-label clinical trial of PARPi ABT-767 were stained for PARP1, RAD51, 53BP1 and multiple components of the nonhomologous end-joining (NHEJ) DNA repair pathway. Modified histochemistry- (H-) scores were determined for each repair protein in each sample. HRD score was determined from tumor DNA. RESULTS 53BP1 deletion increased HR in BRCA1-mutant COV362 cells and decreased PARPi sensitivity in vitro. In 36 women with relapsed ovarian cancer, responses to the PARPi ABT-767 were observed exclusively in cancers with HR deficiency. In this subset, 7 of 18 patients (39%) had objective responses. The actual HRD score did not further correlate with change from baseline tumor volume (r = 0.050; p = 0.87). However, in the HR-deficient subset, decreased 53BP1 H-score was associated with decreased antitumor efficacy of ABT-767 (r = -0.69, p = 0.004). CONCLUSION Differences in complementary repair pathways, particularly 53BP1, correlate with PARPi response of HR-deficient ovarian cancers.
Collapse
Affiliation(s)
- Rachel M Hurley
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States of America
| | | | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States of America
| | - Peter Ansell
- Abbvie, North Chicago, IL, United States of America
| | - Maria I Harrell
- Department of Obstetrics and Gynecology, Univ. of Washington, Seattle, WA, United States of America
| | - Jill M Wagner
- Department of Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Vivian Negron
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States of America
| | - Krista M Goergen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Matthew J Maurer
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - X Wei Meng
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States of America
| | - Karen S Flatten
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States of America
| | | | | | | | | | - Agnes Jager
- Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | | | | | | | - Elizabeth M Swisher
- Department of Obstetrics and Gynecology, Univ. of Washington, Seattle, WA, United States of America
| | - Scott H Kaufmann
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States of America; Department of Oncology, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
127
|
53BP1 as a potential predictor of response in PARP inhibitor-treated homologous recombination-deficient ovarian cancer. Gynecol Oncol 2019. [PMID: 30686551 DOI: 10.1016/j.ygyno.2019.01.015] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Poly(ADP-ribose) polymerase (PARP) inhibitors have shown substantial activity in homologous recombination- (HR-) deficient ovarian cancer and are undergoing testing in other HR-deficient tumors. For reasons that are incompletely understood, not all patients with HR-deficient cancers respond to these agents. Preclinical studies have demonstrated that changes in alternative DNA repair pathways affect PARP inhibitor (PARPi) sensitivity in ovarian cancer models. This has not previously been assessed in the clinical setting. METHODS Clonogenic and plasmid-based HR repair assays were performed to compare BRCA1-mutant COV362 ovarian cancer cells with or without 53BP1 gene deletion. Archival biopsies from ovarian cancer patients in the phase I, open-label clinical trial of PARPi ABT-767 were stained for PARP1, RAD51, 53BP1 and multiple components of the nonhomologous end-joining (NHEJ) DNA repair pathway. Modified histochemistry- (H-) scores were determined for each repair protein in each sample. HRD score was determined from tumor DNA. RESULTS 53BP1 deletion increased HR in BRCA1-mutant COV362 cells and decreased PARPi sensitivity in vitro. In 36 women with relapsed ovarian cancer, responses to the PARPi ABT-767 were observed exclusively in cancers with HR deficiency. In this subset, 7 of 18 patients (39%) had objective responses. The actual HRD score did not further correlate with change from baseline tumor volume (r = 0.050; p = 0.87). However, in the HR-deficient subset, decreased 53BP1 H-score was associated with decreased antitumor efficacy of ABT-767 (r = -0.69, p = 0.004). CONCLUSION Differences in complementary repair pathways, particularly 53BP1, correlate with PARPi response of HR-deficient ovarian cancers.
Collapse
|
128
|
53BP1 as a potential predictor of response in PARP inhibitor-treated homologous recombination-deficient ovarian cancer. Gynecol Oncol 2019; 153:127-134. [PMID: 30686551 DOI: 10.1016/j.ygyno.2019.01.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Poly(ADP-ribose) polymerase (PARP) inhibitors have shown substantial activity in homologous recombination- (HR-) deficient ovarian cancer and are undergoing testing in other HR-deficient tumors. For reasons that are incompletely understood, not all patients with HR-deficient cancers respond to these agents. Preclinical studies have demonstrated that changes in alternative DNA repair pathways affect PARP inhibitor (PARPi) sensitivity in ovarian cancer models. This has not previously been assessed in the clinical setting. METHODS Clonogenic and plasmid-based HR repair assays were performed to compare BRCA1-mutant COV362 ovarian cancer cells with or without 53BP1 gene deletion. Archival biopsies from ovarian cancer patients in the phase I, open-label clinical trial of PARPi ABT-767 were stained for PARP1, RAD51, 53BP1 and multiple components of the nonhomologous end-joining (NHEJ) DNA repair pathway. Modified histochemistry- (H-) scores were determined for each repair protein in each sample. HRD score was determined from tumor DNA. RESULTS 53BP1 deletion increased HR in BRCA1-mutant COV362 cells and decreased PARPi sensitivity in vitro. In 36 women with relapsed ovarian cancer, responses to the PARPi ABT-767 were observed exclusively in cancers with HR deficiency. In this subset, 7 of 18 patients (39%) had objective responses. The actual HRD score did not further correlate with change from baseline tumor volume (r = 0.050; p = 0.87). However, in the HR-deficient subset, decreased 53BP1 H-score was associated with decreased antitumor efficacy of ABT-767 (r = -0.69, p = 0.004). CONCLUSION Differences in complementary repair pathways, particularly 53BP1, correlate with PARPi response of HR-deficient ovarian cancers.
Collapse
|
129
|
Alblihy A, Mesquita KA, Sadiq MT, Madhusudan S. Development and implementation of precision therapies targeting base-excision DNA repair in BRCA1-associated tumors. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1567266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Adel Alblihy
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| | - Katia A. Mesquita
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| | - Maaz T. Sadiq
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | - Srinivasan Madhusudan
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
- Department of Oncology, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| |
Collapse
|
130
|
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: 85] [Impact Index Per Article: 17.0] [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
|
131
|
Cramer T, Rosenberg T, Kisliouk T, Meiri N. PARP Inhibitor Affects Long-term Heat-stress Response via Changes in DNA Methylation. Neuroscience 2018; 399:65-76. [PMID: 30579833 DOI: 10.1016/j.neuroscience.2018.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023]
Abstract
Resilience to stress can be obtained by adjusting the stress-response set point during postnatal sensory development. Recent studies have implemented epigenetic mechanisms to play leading roles in improving resilience. We previously found that better resilience to heat stress in chicks can be achieved by conditioning them to moderate heat stress during their critical developmental period of thermal control establishment, 3 days posthatch. Furthermore, the expression level of corticotropin-releasing hormone (CRH) was found to play a direct role in determining future resilience or vulnerability to heat stress by alterations in its DNA-methylation and demethylation pattern. Here we demonstrate how intraperitoneal injection of poly (ADP-ribose) polymerase (PARP) inhibitor (PARPi) influences the DNA methylation pattern, thereby affecting the long-term heat-stress response. Single PARPi administration, induced a reduction in both 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC), without affecting body temperature. The accumulated effect of three PARPi doses brought about a long-term decrease in 5mC% and 5hmC%. These changes coincided with a reduction in body temperature in non-conditioned chicks, similar to that occurring in moderately conditioned heat-stress-resilient chicks. The observed changes in DNA methylation can be explained by decreased activity of the enzyme DNA methyltransferase as a result of the PARPi injection. Furthermore, evaluation of the DNA-methylation pattern along the CRH intron showed a reduction in 5mC% as a result of PARPi treatment, alongside a reduction in CRH mRNA expression. Thus, PARPi treatment can affect DNA methylation, which can alter hypothalamic-pituitary-adrenal (HPA) axis anchors such as CRH, thereby potentially enhancing long-term resilience to heat stress.
Collapse
Affiliation(s)
- Tomer Cramer
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeZiyyon 7528809, Israel; The Robert H. Smith Faculty of Agriculture, Food and Environment, Department of Animal Science, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Tali Rosenberg
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeZiyyon 7528809, Israel; The Robert H. Smith Faculty of Agriculture, Food and Environment, Department of Animal Science, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Tatiana Kisliouk
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeZiyyon 7528809, Israel
| | - Noam Meiri
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeZiyyon 7528809, Israel.
| |
Collapse
|
132
|
Siraj AK, Pratheeshkumar P, Parvathareddy SK, Divya SP, Al-Dayel F, Tulbah A, Ajarim D, Al-Kuraya KS. Overexpression of PARP is an independent prognostic marker for poor survival in Middle Eastern breast cancer and its inhibition can be enhanced with embelin co-treatment. Oncotarget 2018; 9:37319-37332. [PMID: 30647872 PMCID: PMC6324669 DOI: 10.18632/oncotarget.26470] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/04/2018] [Indexed: 12/29/2022] Open
Abstract
Patients with aggressive breast cancer (BC) subtypes usually don’t have favorable prognosis despite the improvement in treatment modalities. These cancers still remain a major cause of morbidity and mortality in females. This has fostered a major effort to discover actionable molecular targets to treat these patients. Poly ADP ribose polymerase (PARP) is one of these molecular targets that are under comprehensive investigation for treatment of such tumors. However, its role in the pathogenesis of BC from Middle Eastern ethnicity has not yet been explored. Therefore, we examined the expression of PARP protein in a large cohort of over 1000 Middle Eastern BC cases by immunohistochemistry. Correlation with clinico-pathological parameters were performed. Nuclear PARP overexpression was observed in 44.7% of all BC cases and was significantly associated with aggressive clinico-pathological markers. Interestingly, nuclear PARP overexpression was an independent predictor of poor prognosis. PARP overexpression was also directly associated with XIAP overexpression, with PARP and XIAP co-expression in 15.8% (159/1008) of our cases. We showed that combined inhibition of PARP by olaparib and XIAP by embelin significantly and synergistically inhibited cell growth and induced apoptosis in BC cell lines. Finally, co-treatment of olaparib and embelin regressed BC xenograft tumor growth in nude mice. Our results revealed the role of PARP in Middle Eastern BC pathogenesis and prognosis. Furthermore, our data support the potential clinical development of combined inhibition of PARP and XIAP, which eventually could extend the utility of olaparib beyond BRCA deficient cancer.
Collapse
Affiliation(s)
- Abdul Khalid Siraj
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Poyil Pratheeshkumar
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Sasidharan Padmaja Divya
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Asma Tulbah
- Department of Pathology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dahish Ajarim
- Department of Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khawla S Al-Kuraya
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
133
|
Li D, Luo Y, Chen X, Zhang L, Wang T, Zhuang Y, Fan Y, Xu J, Chen Y, Wu L. NF-κB and Poly (ADP-ribose) Polymerase 1 Form a Positive Feedback Loop that Regulates DNA Repair in Acute Myeloid Leukemia Cells. Mol Cancer Res 2018; 17:761-772. [DOI: 10.1158/1541-7786.mcr-18-0523] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/20/2018] [Accepted: 12/12/2018] [Indexed: 11/16/2022]
|
134
|
Strome A, Kossatz S, Zanoni DK, Rajadhyaksha M, Patel S, Reiner T. Current Practice and Emerging Molecular Imaging Technologies in Oral Cancer Screening. Mol Imaging 2018; 17:1536012118808644. [PMID: 32852263 PMCID: PMC6287312 DOI: 10.1177/1536012118808644] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oral cancer is one of the most common cancers globally. Survival rates for patients are directly correlated with stage of diagnosis; despite this knowledge, 60% of individuals are presenting with late-stage disease. Currently, the initial evaluation of a questionable lesion is performed by a conventional visual examination with white light. If a lesion is deemed suspicious, a biopsy is taken for diagnosis. However, not all lesions present suspicious under visual white light examination, and there is limited specificity in differentiating between benign and malignant transformations. Several vital dyes, light-based detection systems, and cytology evaluation methods have been formulated to aid in the visualization process, but their lack of specific biomarkers resulted in high false-positive rates and thus limits their reliability as screening and guidance tools. In this review, we will analyze the current methodologies and demonstrate the need for specific intraoral imaging agents to aid in screening and diagnosis to identify patients earlier. Several novel molecular imaging agents will be presented as, by result of their molecular targeting, they aim to have high specificity for tumor pathways and can support in identifying dysplastic/cancerous lesions and guiding visualization of biopsy sites. Imaging agents that are easy to use, inexpensive, noninvasive, and specific can be utilized to increase the number of patients who are screened and monitored in a variety of different environments, with the ultimate goal of increasing early detection.
Collapse
Affiliation(s)
- Arianna Strome
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Milind Rajadhyaksha
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Snehal Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Radiology, Weill-Cornell Medical College, New York, NY, USA
| |
Collapse
|
135
|
Wardi J, Ernst O, Lilja A, Aeed H, Katz S, Ben-Nachum I, Ben-Dror I, Katz D, Bernadsky O, Kandhikonda R, Avni Y, Fraser IDC, Weinstain R, Biro A, Zor T. 3-Aminobenzamide Prevents Concanavalin A-Induced Acute Hepatitis by an Anti-inflammatory and Anti-oxidative Mechanism. Dig Dis Sci 2018; 63:3382-3397. [PMID: 30196390 DOI: 10.1007/s10620-018-5267-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 08/24/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Concanavalin A is known to activate T cells and to cause liver injury and hepatitis, mediated in part by secretion of TNFα from macrophages. Poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors have been shown to prevent tissue damage in various animal models of inflammation. The objectives of this study were to evaluate the efficacy and mechanism of the PARP-1 inhibitor 3-aminobenzamide (3-AB) in preventing concanavalin A-induced liver damage. METHODS We tested the in vivo effects of 3-AB on concanavalin A-treated mice, its effects on lipopolysaccharide (LPS)-stimulated macrophages in culture, and its ability to act as a scavenger in in vitro assays. RESULTS 3-AB markedly reduced inflammation, oxidative stress, and liver tissue damage in concanavalin A-treated mice. In LPS-stimulated RAW264.7 macrophages, 3-AB inhibited NFκB transcriptional activity and subsequent expression of TNFα and iNOS and blocked NO production. In vitro, 3-AB acted as a hydrogen peroxide scavenger. The ROS scavenger N-acetylcysteine (NAC) and the ROS formation inhibitor diphenyleneiodonium (DPI) also inhibited TNFα expression in stimulated macrophages, but unlike 3-AB, NAC and DPI were unable to abolish NFκB activity. PARP-1 knockout failed to affect NFκB and TNFα suppression by 3-AB in stimulated macrophages. CONCLUSIONS Our results suggest that 3-AB has a therapeutic effect on concanavalin A-induced liver injury by inhibiting expression of the key pro-inflammatory cytokine TNFα, via PARP-1-independent NFκB suppression and via an NFκB-independent anti-oxidative mechanism.
Collapse
Affiliation(s)
- Joram Wardi
- Department of Gastroenterology, E. Wolfson Medical Center, P.O.B. 5, 58100, Holon, Israel.
| | - Orna Ernst
- Department of Biochemistry and Molecular Biology, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel.,Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anna Lilja
- Department of Biochemistry and Molecular Biology, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Hussein Aeed
- Department of Gastroenterology, E. Wolfson Medical Center, P.O.B. 5, 58100, Holon, Israel
| | - Sebastián Katz
- Department of Biochemistry and Molecular Biology, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Idan Ben-Nachum
- Department of Biochemistry and Molecular Biology, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Iris Ben-Dror
- Department of Biochemistry and Molecular Biology, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Dolev Katz
- Department of Biochemistry and Molecular Biology, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Olga Bernadsky
- Department of Pathology, E. Wolfson Medical Center, P.O.B. 5, 58100, Holon, Israel
| | - Rajendar Kandhikonda
- Department of Molecular Biology and Ecology of Plants, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Yona Avni
- Department of Gastroenterology, E. Wolfson Medical Center, P.O.B. 5, 58100, Holon, Israel
| | - Iain D C Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Roy Weinstain
- Department of Molecular Biology and Ecology of Plants, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel
| | - Alexander Biro
- Institute of Nephrology, E. Wolfson Medical Center, P.O.B. 5, 58100, Holon, Israel.
| | - Tsaffrir Zor
- Department of Biochemistry and Molecular Biology, Life Sciences Faculty, Tel-Aviv University, 69978, Tel-Aviv, Israel.
| |
Collapse
|
136
|
Fenerty KE, Padget M, Wolfson B, Gameiro SR, Su Z, Lee JH, Rabizadeh S, Soon-Shiong P, Hodge JW. Immunotherapy utilizing the combination of natural killer- and antibody dependent cellular cytotoxicity (ADCC)-mediating agents with poly (ADP-ribose) polymerase (PARP) inhibition. J Immunother Cancer 2018; 6:133. [PMID: 30486888 PMCID: PMC6264611 DOI: 10.1186/s40425-018-0445-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/08/2018] [Indexed: 01/24/2023] Open
Abstract
Background Poly (ADP-ribose) polymerase inhibitors (PARPi) prevent single-stranded DNA repair. Olaparib is a PARPi approved for the treatment of BRCA mutant ovarian and breast carcinoma. Emerging clinical data suggest a benefit of combining olaparib with immunotherapy in prostate cancer patients both with and without somatic BRCA mutations. Methods We examined if olaparib, when combined with IgG1 antibody-dependent cellular cytotoxicity (ADCC)-mediating monoclonal antibodies (mAbs) cetuximab (anti-EGFR), or avelumab (anti-PD-L1), would increase tumor cell sensitivity to killing by natural killer (NK) cells independently of BRCA status or mAb target upregulation. BRCA mutant and BRCA wildtype (WT) prostate carcinoma cell lines were pretreated with olaparib and then exposed to NK cells in the presence or absence of cetuximab or avelumab. Results NK-mediated killing was significantly increased in both cell lines and was further increased using the ADCC-mediating mAbs. Pre-exposure of NK cells to recombinant IL-15/IL-15Rα further increased the lysis of olaparib treated tumor cells. In addition, olaparib treated tumor cells were killed to a significantly greater degree by engineered high-affinity NK cells (haNK). We show here for the first time that (a) olaparib significantly increased tumor cell sensitivity to NK killing and ADCC in both BRCA WT and BRCA mutant prostate carcinoma cells, independent of PD-L1 or EGFR modulation; (b) mechanistically, treatment with olaparib upregulated death receptor TRAIL-R2; and (c) olaparib significantly enhanced NK killing of additional tumor types, including breast, non-small cell lung carcinoma, and chordoma. Conclusions These studies support the combined use of NK- and ADCC-mediating agents with correctly timed PARP inhibition. Electronic supplementary material The online version of this article (10.1186/s40425-018-0445-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kathleen E Fenerty
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD, 20892, USA
| | - Michelle Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD, 20892, USA
| | - Benjamin Wolfson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD, 20892, USA
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD, 20892, USA
| | - Zhen Su
- EMD Serono, Billerica, MA, USA
| | | | | | | | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room 8B09, Bethesda, MD, 20892, USA.
| |
Collapse
|
137
|
Césaire M, Thariat J, Candéias SM, Stefan D, Saintigny Y, Chevalier F. Combining PARP inhibition, radiation, and immunotherapy: A possible strategy to improve the treatment of cancer? Int J Mol Sci 2018; 19:ijms19123793. [PMID: 30487462 PMCID: PMC6321381 DOI: 10.3390/ijms19123793] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/20/2018] [Accepted: 11/24/2018] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy has revolutionized the practice of oncology, improving survival in certain groups of patients with cancer. Immunotherapy can synergize with radiation therapy, increase locoregional control, and have abscopal effects. Combining it with other treatments, such as targeted therapies, is a promising means of improving the efficacy of immunotherapy. Because the value of immunotherapy is amplified with the expression of tumor antigens, coupling poly(ADP-ribose) polymerase (PARP) inhibitors and immunotherapy might be a promising treatment for cancer. Further, PARP inhibitors (PARPis) are being combined with radiation therapy to inhibit DNA repair functions, thus enhancing the effects of radiation; this association might interact with the antitumor immune response. Cytotoxic T lymphocytes are central to the antitumor immune response. PARP inhibitors and ionizing radiation can enhance the infiltration of cytotoxic T lymphocytes into the tumor bed, but they can also enhance PD-1/PDL-1 expression. Thus, the addition of immune checkpoint inhibitors with PARP inhibitors and/or ionizing radiation could counterbalance such immunosuppressive effects. With the present review article, we proposed to evaluate some of these associated therapies, and we explored the biological mechanisms and medical benefits of the potential combination of radiation therapy, immunotherapy, and PARP inhibitors.
Collapse
Affiliation(s)
- Mathieu Césaire
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14076 Caen, France.
- UMR6252 CIMAP, CEA - CNRS - ENSICAEN - Université de Caen Normandie, 14076 Caen, France.
- Radiotherapy Unit, Centre François Baclesse, 14000 Caen, France.
| | - Juliette Thariat
- Radiotherapy Unit, Centre François Baclesse, 14000 Caen, France.
| | - Serge M Candéias
- ProMD, Chemistry and Biology of Metals Laboratory, Univ. Grenoble Alpes, CEA, CNRS, BIG-LCBM, 38054 Grenoble, France.
| | - Dinu Stefan
- Radiotherapy Unit, Centre François Baclesse, 14000 Caen, France.
| | - Yannick Saintigny
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14076 Caen, France.
- UMR6252 CIMAP, CEA - CNRS - ENSICAEN - Université de Caen Normandie, 14076 Caen, France.
| | - François Chevalier
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14076 Caen, France.
- UMR6252 CIMAP, CEA - CNRS - ENSICAEN - Université de Caen Normandie, 14076 Caen, France.
| |
Collapse
|
138
|
Abstract
OPINION STATEMENT Use of poly(ADP-ribose) polymerase (PARP) inhibitors has greatly increased over the past 5 years. With several new Food and Drug Administration (FDA) approvals, three PARP inhibitors have entered into standard of care treatment for epithelial ovarian cancer (including ovarian, fallopian tube, and primary peritoneal cancer). Olaparib and rucaparib currently have indications for treatment of recurrent BRCA mutant ovarian cancer. Olaparib, rucaparib, and niraparib all have indications for maintenance therapy in recurrent platinum-sensitive ovarian cancer after response to platinum-based therapy. In our practice, we use both olaparib and rucaparib in the recurrent setting, and all three PARP inhibitors in the maintenance setting. Choice of which PARP inhibitor to use in either setting is largely based upon baseline laboratory values, number of prior therapies, and presence of a BRCA mutation and/or homologous recombination deficiency (HRD). As (HRD) and other biomarker assessments continue to improve, we anticipate being able to better identify which patients might most benefit from PARP inhibitor therapy in the future. The clinically available PARP inhibitors are currently undergoing extensive investigations in clinical trials. Other newer agents such as talazoparib, veliparib, 2X-121, and CEP-9722 are in earlier stages of development. As more FDA-approved indications for PARP inhibitor therapy in ovarian cancer become available, we anticipate the decision of which PARP inhibitor to use will become increasingly complex.
Collapse
|
139
|
Gonzales J, Kossatz S, Roberts S, Pirovano G, Brand C, Pérez-Medina C, Donabedian P, de la Cruz MJ, Mulder WJM, Reiner T. Nanoemulsion-Based Delivery of Fluorescent PARP Inhibitors in Mouse Models of Small Cell Lung Cancer. Bioconjug Chem 2018; 29:3776-3782. [PMID: 30354077 DOI: 10.1021/acs.bioconjchem.8b00640] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The preclinical potential of many diagnostic and therapeutic small molecules is limited by their rapid washout kinetics and consequently modest pharmacological performances. In several cases, these could be improved by loading the small molecules into nanoparticulates, improving blood half-life, in vivo uptake and overall pharmacodynamics. In this study, we report a nanoemulsion (NE) encapsulated form of PARPi-FL. As a proof of concept, we used PARPi-FL, which is a fluorescently labeled sensor for olaparib, a FDA-approved small molecule inhibitor of the nuclear enzyme poly(ADP-ribose)polymerase 1 (PARP1). Encapsulated PARPi-FL showed increased blood half-life, and delineated subcutaneous xenografts of small cell lung cancer (SCLC), a fast-progressing disease where efficient treatment options remain an unmet clinical need. Our study demonstrates an effective method for expanding the circulation time of a fluorescent PARP inhibitor, highlighting the pharmacokinetic benefits of nanoemulsions as nanocarriers and confirming the value of PARPi-FL as an imaging agent targeting PARP1 in small cell lung cancer.
Collapse
Affiliation(s)
- Junior Gonzales
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Susanne Kossatz
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Sheryl Roberts
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Giacomo Pirovano
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Christian Brand
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Department of Radiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Patrick Donabedian
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - M Jason de la Cruz
- Structural Biology Program, Sloan Kettering Institute , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Department of Radiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems , Eindhoven University of Technology , Eindhoven , The Netherlands
| | - Thomas Reiner
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Department of Radiology , Weill Cornell Medical College , New York , New York 10065 , United States
| |
Collapse
|
140
|
Zhong Q, Hu Z, Li Q, Yi T, Li J, Yang H. Cyclin D1 silencing impairs DNA double strand break repair, sensitizes BRCA1 wildtype ovarian cancer cells to olaparib. Gynecol Oncol 2018; 152:157-165. [PMID: 30414739 DOI: 10.1016/j.ygyno.2018.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/14/2018] [Accepted: 10/22/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Poly(ADP-ribose) polymerase inhibitors (PARPi) are active in cancer cells that have impaired repair of DNA by the homologous recombination (HR) pathway. Strategies that disrupt HR may sensitize HR-proficient tumors to PARP inhibition. As a component of the core cell cycle machinery, cyclin D1 has unexpected function in DNA repair, suggesting that targeting cyclin D1 may represent a plausible strategy for expanding the utility of PARPi in ovarian cancer. METHODS BRCA1 wildtype ovarian cancer cells (A2780 and SKOV3) were treated with a combination of CCND1 siRNA and olaparib in vitro. Cell viability was assessed by MTT. The effects of the combined treatment on DNA damage repair and cell cycle progression were examined to dissect molecular mechanisms. In vivo studies were performed in an orthotopic ovarian cancer mouse model. Animals were treated with a combination of lentivirus-mediated CCND1 shRNA and olaparib or olaparib plus scrambled shRNA. Molecular downstream effects were examined by immunohistochemistry. RESULTS Silencing of cyclin D1 sensitized ovarian cancer cells to olaparib through interfering with RAD51 accumulation and inducing cell cycle G0/G1 arrest. Treatment of lentivirus-mediated CCND1-shRNA in nude mice statistically significantly augmented the olaparib response (mean tumor weight ± SD, CCND1-shRNA plus olaparib vs scrambled shRNA plus olaparib: 0.172 ± 0.070 g vs 0.324 ± 0.044 g, P< 0.05). CONCLUSIONS Silencing of cyclin D1 combined with olaparib may lead to substantial benefit for ovarian cancer management by mimicking a BRCAness phenotype, and induction of G0/G1 cell cycle arrest.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Gynecology and Obstetrics, West China Second University Hospital of Sichuan University, Chengdu, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, PR China.
| | - Zhongyi Hu
- Center for Research on Reproduction & Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qiao Li
- Physical Examination Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Tao Yi
- Department of Gynecology and Obstetrics, West China Second University Hospital of Sichuan University, Chengdu, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, PR China
| | - Jinke Li
- Department of Gynecology and Obstetrics, West China Second University Hospital of Sichuan University, Chengdu, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, PR China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China.
| |
Collapse
|
141
|
Mullen MM, Kuroki LM, Thaker PH. Novel treatment options in platinum-sensitive recurrent ovarian cancer: A review. Gynecol Oncol 2018; 152:416-425. [PMID: 30409489 DOI: 10.1016/j.ygyno.2018.10.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/18/2018] [Accepted: 10/20/2018] [Indexed: 11/26/2022]
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death due to gynecologic malignancy. The majority of advanced stage EOC patients, even those who respond well to frontline therapy, will ultimately recur and succumb to their disease. In platinum-sensitive EOC patients, or those who recur ≥6 months from initial diagnosis, treatment of recurrent disease has traditionally consisted of repeat platinum-based chemotherapy. Secondary cytoreduction remains controversial. Due to recent advances in molecularly targeted treatment options, outcomes for advanced stage EOC patients are significantly improving and hold great promise. This review discusses pivotal trials establishing platinum-based combination chemotherapy as the standard of care and addresses the utility of increasing a patient's platinum-free interval. It then discusses the role of anti-angiogenesis therapeutics, specifically bevacizumab, cediranib, and trebananib and their side effects. Lastly, it reviews key trials for the three poly-adenosine diphosphate [ADP]-ribose polymerases (PARP) inhibitors that have been FDA-approved for maintenance therapy in platinum-sensitive recurrent EOC: olaparib, rucaparib, and niraparib. This review concludes with a discussion regarding ongoing and future clinical trials.
Collapse
Affiliation(s)
- Mary M Mullen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, Alvin J. Siteman Cancer Center, St Louis, MO, United States of America
| | - Lindsay M Kuroki
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, Alvin J. Siteman Cancer Center, St Louis, MO, United States of America
| | - Premal H Thaker
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, Alvin J. Siteman Cancer Center, St Louis, MO, United States of America.
| |
Collapse
|
142
|
Ovarian carcinomas: at least five different diseases with distinct histological features and molecular genetics. Hum Pathol 2018; 80:11-27. [DOI: 10.1016/j.humpath.2018.06.018] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 11/18/2022]
|
143
|
Job A, Schmitt LM, von Wenserski L, Lankat-Buttgereit B, Gress TM, Buchholz M, Gallmeier E. Inactivation of PRIM1 Function Sensitizes Cancer Cells to ATR and CHK1 Inhibitors. Neoplasia 2018; 20:1135-1143. [PMID: 30257222 PMCID: PMC6154763 DOI: 10.1016/j.neo.2018.08.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
The phosphoinositide 3-kinase-related kinase ATR is a central regulator of the DNA damage response. Its chemical inhibition eliminates subsets of cancer cells in various tumor types. This effect is caused at least partly by the synthetically lethal relationship between ATR and certain DNA repair genes. In a previous screen using an siRNA library against DNA repair genes, we identified PRIM1, a part of the polymerase α-primase complex, as acting synthetically lethal with ATR. Applying a genetic ATR knock-in model of colorectal cancer cells, we confirmed that PRIM1 depletion inhibited proliferation of ATR-deficient cells and excluded artifacts due to clonal variation using an ATR reexpressing cell clone. We expanded these data by demonstrating in different cell lines that also chemical inhibition of ATR or its main effector kinase CHK1 reduces proliferation upon depletion of PRIM1. Mechanistically, PRIM1 depletion in ATR-deficient cells caused S-phase stasis in the absence of increased DNA damage followed by Wee1-mediated activation of caspase 8 and apoptosis. As PRIM1 inactivation sensitizes cancer cells to ATR and CHK1 inhibitors, mutations in PRIM1 or other components of the polymerase α-primase complex could represent novel targets for individualized tumor therapeutic approaches using ATR/CHK1 inhibitors, as has been previously demonstrated for POLD1, the catalytic subunit of polymerase δ.
Collapse
Affiliation(s)
- Albert Job
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Lisa-Maria Schmitt
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Lisa von Wenserski
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Brigitte Lankat-Buttgereit
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Thomas M Gress
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Malte Buchholz
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany
| | - Eike Gallmeier
- Center for Tumor Biology and Immunology, Department of Gastroenterology, Endocrinology and Metabolism, University Hospital of Marburg, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
144
|
Prati B, Marangoni B, Boccardo E. Human papillomavirus and genome instability: from productive infection to cancer. Clinics (Sao Paulo) 2018; 73:e539s. [PMID: 30208168 PMCID: PMC6113919 DOI: 10.6061/clinics/2018/e539s] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/16/2018] [Indexed: 12/29/2022] Open
Abstract
Infection with high oncogenic risk human papillomavirus types is the etiological factor of cervical cancer and a major cause of other epithelial malignancies, including vulvar, vaginal, anal, penile and head and neck carcinomas. These agents affect epithelial homeostasis through the expression of specific proteins that deregulate important cellular signaling pathways to achieve efficient viral replication. Among the major targets of viral proteins are components of the DNA damage detection and repair machinery. The activation of many of these cellular factors is critical to process viral genome replication intermediates and, consequently, to sustain faithful viral progeny production. In addition to the important role of cellular DNA repair machinery in the infective human papillomavirus cycle, alterations in the expression and activity of many of its components are observed in human papillomavirus-related tumors. Several studies from different laboratories have reported the impact of the expression of human papillomavirus oncogenes, mainly E6 and E7, on proteins in almost all the main cellular DNA repair mechanisms. This has direct consequences on cellular transformation since it causes the accumulation of point mutations, insertions and deletions of short nucleotide stretches, as well as numerical and structural chromosomal alterations characteristic of tumor cells. On the other hand, it is clear that human papillomavirus-transformed cells depend on the preservation of a basal cellular DNA repair activity level to maintain tumor cell viability. In this review, we summarize the data concerning the effect of human papillomavirus infection on DNA repair mechanisms. In addition, we discuss the potential of exploiting human papillomavirus-transformed cell dependency on DNA repair pathways as effective antitumoral therapies.
Collapse
Affiliation(s)
- Bruna Prati
- Departamento de Microbiologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Bruna Marangoni
- Departamento de Microbiologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Enrique Boccardo
- Departamento de Microbiologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
145
|
Involvement of poly(ADP-ribose) polymerase-1 in Chinese patients with glioma: a potential target for effective patient care. Int J Biol Markers 2018; 33:68-72. [PMID: 28777431 DOI: 10.5301/ijbm.5000267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE We aimed to evaluate the genetic variation of poly(ADP-ribose) polymerase-1 (PARP-1) in the development of gliomas among Chinese individuals. MATERIALS AND METHODS Patients with a confirmed diagnosis of glioma and healthy individuals with no clinical symptoms of glioma were enrolled at Liaocheng People's Hospital, China. Genetic polymorphisms were studied in plasma samples by polymerase chain reaction-restriction fragment length polymorphism assay. Cytokine levels were measured routinely in serum samples by sandwich ELISA technique. RESULTS A total of 120 Chinese patients with gliomas and 120 healthy Chinese individuals were included. We found that patients with the GG genotype (odds ratio [OR] 2.53, 95% confidence interval [CI] 1.46-4.38, p<0.001) and carriers of the G allele (OR 11.5, 95% CI 6.31-21.3, p<0.0001) were at high risk of developing glioma. A del/ins polymorphism of the NF-κB1 gene (OR 4.27, 95% CI 2.43-7.50, p<0.001) was also found to be associated with glioma. In addition, significantly increased cytokine levels were observed in patients with glioma (p<0.05). CONCLUSIONS Our findings showed that PARP-1 polymorphisms are involved in the development of glioma in Chinese individuals. Also serum cytokine levels can be considered among the potential risk factors for developing glioma.
Collapse
|
146
|
Dal Molin GZ, Westin SN, Coleman RL. Rucaparib in ovarian cancer: extending the use of PARP inhibitors in the recurrent disease. Future Oncol 2018; 14:3101-3110. [PMID: 30105925 DOI: 10.2217/fon-2018-0215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rucaparib is a potent inhibitor of poly (ADP-ribose) polymerase (PARP) PARP1, PARP2 and PARP3, and to a lesser extent, PARP4, PARP10, PARP12, PARP15 and PARP16. Study 10 and ARIEL2 evaluated the use of rucaparib as treatment in patients with recurrent high-grade ovarian carcinoma and resulting in approval of rucaparib for patients with both germline and somatic BRCA mutation. Data from the Phase III trial ARIEL3 led to approval in platinum-sensitive disease as maintenance. This article reviews the efficacy, safety, pharmacokinetics and pharmacodynamics of rucaparib as well as future and ongoing trials.
Collapse
Affiliation(s)
- Graziela Z Dal Molin
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
147
|
Mangoni M, Sottili M, Salvatore G, Meattini I, Desideri I, Greto D, Loi M, Becherini C, Garlatti P, Delli Paoli C, Dominici L, Gerini C, Scoccianti S, Bonomo P, Silvano A, Beltrami G, Campanacci D, Livi L. Enhancement of Soft Tissue Sarcoma Cell Radiosensitivity by Poly(ADP-ribose) Polymerase-1 Inhibitors. Radiat Res 2018; 190:464-472. [PMID: 30067444 DOI: 10.1667/rr15035.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Soft tissue sarcomas (STS) are aggressive tumors with a poor prognosis. Poly(ADP-ribose) polymerase (PARP)-1 inhibitors (PARPi) enhance the cytotoxic effects of radiation. In this study, we evaluated the effect of PARPi on survival and DNA damage of irradiated STS cells. For clonogenic assays, STS cell lines were irradiated with or without olaparib, iniparib or veliparib pretreatment. The effect of PARP inhibition on γ-H2AX and Rad51 foci formation, on PARP-1, phospho-ERK and cleaved caspase-3 protein expression and on cell cycle progression was evaluated on irradiated rhabdomyosarcoma cells pretreated with olaparib. The results from this work showed that PARPi induced significant radiosensitization in STS cells. Rhabdomyosarcoma cells showed the highest increase in radiosensitivity, with a radiosensitization enhancement ratio at 50% survival (ER50) of 3.41 with veliparib. All PARPi exerted a synergistic effect when combined with radiation. Fibrosarcoma cells showed an ER50 of 2.29 with olaparib. Leiomyosarcoma and liposarcoma cells showed their highest ER50 with veliparib (1.71 and 1.84, respectively). In rhabdomyosarcoma, olaparib enhanced the formation of radiation-induced γ-H2AX/Rad51 foci and PARP-1 cleavage, induced slightly increased expression of cleaved caspase-3 and reduced phospho-ERK expression. Moreover, the combination of olaparib and radiation resulted in a significantly enhanced cell cycle arrest in the G2/M phase compared to the two treatments alone. In conclusion, we have shown that PARPi are potent radiosensitizers of human STS cells. These results support the pursuit of further investigations into the effects of PARPi combined with radiation on STS.
Collapse
Affiliation(s)
- Monica Mangoni
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Mariangela Sottili
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Giulia Salvatore
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Icro Meattini
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Isacco Desideri
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Daniela Greto
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Mauro Loi
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Carlotta Becherini
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Pietro Garlatti
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Camilla Delli Paoli
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Luca Dominici
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Chiara Gerini
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Silvia Scoccianti
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Pierluigi Bonomo
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Angela Silvano
- b Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Giovanni Beltrami
- c Department of Orthopaedic Oncology, Azienda Ospedaliera Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy and.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Domenico Campanacci
- c Department of Orthopaedic Oncology, Azienda Ospedaliera Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy and.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| | - Lorenzo Livi
- Department of a Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.,d Istituto Toscano Tumori, via Taddeo Alderotti 26/N, 50139 Florence, Italy
| |
Collapse
|
148
|
Bi Y, Verginadis II, Dey S, Lin L, Guo L, Zheng Y, Koumenis C. Radiosensitization by the PARP inhibitor olaparib in BRCA1-proficient and deficient high-grade serous ovarian carcinomas. Gynecol Oncol 2018; 150:534-544. [PMID: 30025822 DOI: 10.1016/j.ygyno.2018.07.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/27/2018] [Accepted: 07/01/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Approximately 15-25% of high-grade serous ovarian carcinomas (HGSOC) harbor BRCA1/2 mutations. Inhibition of Poly (ADP-ribose) polymerase (PARP) is synthetically lethal to cells and tumors with BRCA1/2 mutation. Our goal was to investigate the radiosensitizing effects of PARP inhibitor olaparib in HGSOC with different BRCA1 status. METHODS The radiosensitizing effects of olaparib were tested on BRCA1-proficient and deficient HGSOC by clonogenic survival and tumor growth assays. The effects of olaparib and radiation on DNA damage, PARP activity, and apoptosis were determined. RESULTS BRCA1-deficient HGSOC cells were more sensitive to RT alone and exhibited significantly higher levels of olaparib-mediated radiosensitization compared to BRCA1-proficient cells. Furthermore, when combined with RT, olaparib inhibited DNA damage repair and PARP1 activity, increased apoptosis, decreased growth of HGSOC xenografts and increased overall host survival. The growth-inhibitory effects of the combined olaparib and RT treatment were more pronounced in mice bearing BRCA1-deficient tumors compared to BRCA1-proficient tumors. CONCLUSIONS These results provide a preclinical rationale for improved treatment modalities using olaparib as an effective radiosensitizer in HGSOC, particularly in tumors with BRCA1-deficiencies.
Collapse
Affiliation(s)
- Yue Bi
- Oncology Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, China; Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Souvik Dey
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lilie Lin
- MD Anderson Cancer Center, Division of Radiation Oncology, Houston, TX 77054, USA
| | - Linlang Guo
- Oncology Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yanfang Zheng
- Oncology Center, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, China.
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
149
|
O'Cearbhaill RE. Using PARP Inhibitors in Advanced Ovarian Cancer. ONCOLOGY (WILLISTON PARK, N.Y.) 2018; 32:339-343. [PMID: 30080919 PMCID: PMC6662180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Poly(ADP-ribose) polymerase (PARP) proteins are used by cells in several DNA repair processes. PARP inhibition can result in preferential death of cancer cells when another mechanism for repairing DNA is defective. Two PARP inhibitors, olaparib and rucaparib, have been approved by the US Food and Drug Administration (FDA) for the treatment of recurrent, BRCA-associated ovarian cancer. More recently, these two and a third PARP inhibitor, niraparib, were approved by the FDA as maintenance therapy following platinum-based chemotherapy for recurrent ovarian cancer. This has caused a paradigm shift in disease management and a challenge for clinicians, who must decide how best to use these agents in individualized treatment. The oral formulation is attractive to patients, but adverse effects such as nausea and fatigue can impact quality of life. As clinicians become comfortable selecting PARP inhibitors and managing associated toxicities, future steps will be to investigate how to safely administer them in combination with other therapies.
Collapse
|
150
|
Grimmett C, Brooks C, Recio-Saucedo A, Armstrong A, Cutress RI, Gareth Evans D, Copson E, Turner L, Meiser B, Wakefield CE, Eccles D, Foster C. Development of Breast Cancer Choices: a decision support tool for young women with breast cancer deciding whether to have genetic testing for BRCA1/2 mutations. Support Care Cancer 2018; 27:297-309. [DOI: 10.1007/s00520-018-4307-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 06/06/2018] [Indexed: 01/26/2023]
|