1
|
Marranci A, Maresca L, Lodovichi S, Luserna di Rorà AG, Stecca B, Poliseno L. PARP1 in melanoma: Mechanistic insights and implications for basic and clinical research. Cancer Lett 2025; 617:217599. [PMID: 40024566 DOI: 10.1016/j.canlet.2025.217599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/04/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Targeted therapies and immunotherapies have revolutionized the treatment of metastatic melanoma and have set a successful example for the treatment of other cancers. A similar breakthrough was achieved with the advent of PARP inhibitors (PARPi) in breast and ovarian cancer. Recent evidence highlights the critical role of PARP1 in melanoma initiation and progression. High PARP1 expression correlates with aggressive melanoma characteristics and poor patient outcomes. Preclinical and clinical data suggest that PARPi, alone or in combination, can effectively reduce melanoma cell viability and inhibit tumor growth. However, integrating PARPi with current treatment approaches and identifying patients who could benefit the most from such combinations remain underexplored areas of investigation. This review highlights the need for further basic and clinical research on PARP1 in melanoma, to better understand its role and to tackle major challenges in the field, such as resistance to targeted therapies and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Andrea Marranci
- Oncohematology Unit, Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, 56017, Pisa, Italy. http://www.fpscience.it/
| | - Luisa Maresca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, 50139, Florence, Italy
| | - Samuele Lodovichi
- Department of Biosciences, University of Milan, 20133, Milan, Italy; Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy
| | | | - Barbara Stecca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy; Oncogenomics Unit, Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), 56124, Pisa, Italy.
| |
Collapse
|
2
|
Joshi D, Patel J, Munshi M, Mistry Z, Prajapati A, Mukherjee A, Ramachandran AV, Parashar NC, Parashar G, Haque S, Tuli HS. Hormones as a double-edged sword: the role of hormones in cancer progression and the potential of targeted hormone therapies. Med Oncol 2024; 41:283. [PMID: 39400627 DOI: 10.1007/s12032-024-02517-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
Cancer remains a significant cause of mortality in the world, with increasing prevalence worldwide. There are numerous treatments ranging from surgery to chemotherapy and radiotherapy, but since cancer is a heterogeneous disease, only few patients possibly respond to treatments. However, it opens a huge space for the advent of targeted therapies such as hormone therapy, immunotherapy, and target-specific drugs. Hormonal therapy using hormone agonists/antagonists or hormone receptor inhibitors-called the next-generation hormonal agents-hits distinct hormonal pathways that are involved in breast, prostate and ovarian cancer. Preliminary results show that through combination of drugs, it is possible that the synergistic effects may actually lead to better survival than with the use of single drugs. With manageable adverse effects, hormonal therapy offers much hope for treatment of this rather challenging malignancy of the hormone-sensitive cancers, especially in combination with other treatments.
Collapse
Affiliation(s)
- Dixita Joshi
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India
| | - Janaki Patel
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India
| | - Muskaan Munshi
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India
| | - Zeel Mistry
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India
| | - Alok Prajapati
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India
| | - Asmi Mukherjee
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India
| | - A V Ramachandran
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India
| | - Nidarshana Chaturvedi Parashar
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India.
| | - Gaurav Parashar
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, 391410, India.
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 11022801, Lebanon
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India
| |
Collapse
|
3
|
Huang W, Kim BS, Zhang Y, Lin L, Chai G, Zhao Z. Regulatory T cells subgroups in the tumor microenvironment cannot be overlooked: Their involvement in prognosis and treatment strategy in melanoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:4512-4530. [PMID: 38530049 DOI: 10.1002/tox.24247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/03/2024] [Accepted: 03/14/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Melanoma, the most lethal form of skin cancer, presents substantial challenges despite effective surgical interventions for in situ lesions. Regulatory T cells (Tregs) wield a pivotal immunomodulatory influence within the tumor microenvironment, yet their impact on melanoma prognosis and direct molecular interactions with melanoma cells remain elusive. This investigation employs single-cell analysis to unveil the intricate nature of Tregs in human melanoma. METHODS Single-cell RNA and bulk sequencing data, alongside clinical information, were obtained from public repositories. Initially, GO and GSEA analyses were employed to delineate functional disparities among distinct cell subsets. Pseudotime and cell-cell interconnection analyses were conducted, followed by an endeavor to construct a prognostic model grounded in Treg-associated risk scores. This model's efficacy was demonstrated via PCA and K-M analyses, with multivariate Cox regression affirming its independent prognostic value in melanoma patients. Furthermore, immune infiltration analysis, immune checkpoint gene expression scrutiny, and drug sensitivity assessments were performed to ascertain the clinical relevance of this prognostic model. RESULTS Following batch effect correction, 80 025 cells partitioned into 31 clusters, encompassing B cells, plasma cells, endothelial cells, fibroblasts, melanoma cells, monocytes, macrophages, and T_NK cells. Within these, 4240 CD4+ T cells were subclassified into seven distinct types. Functional analysis underscored the immunomodulatory function of Tregs within the melanoma tumor microenvironment, elucidating disparities among Treg subpopulations. Notably, the ITGB2 signaling pathway emerged as a plausible molecular nexus linking Tregs to melanoma cells. Our prognostic signature exhibited robust predictive capacities for melanoma prognosis and potential implications in evaluating immunotherapy response. CONCLUSION Tregs exert a critical role in immune suppression within the melanoma tumor microenvironment, revealing a potential molecular-level association with melanoma cells. Our innovative Treg-centered signature introduces a promising prognostic marker for melanoma, holding potential for future clinical prognostic assessments.
Collapse
Affiliation(s)
- Wenyi Huang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Byeong Seop Kim
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yichi Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Lin
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Stomatology, First Affiliated Hospital of Soochow University, Suzhou, China
- National Center for Translational Medicine(Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Gang Chai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijie Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Thein KZ, Thawani R, Kummar S. Combining Poly (ADP-Ribose) Polymerase (PARP) Inhibitors with Chemotherapeutic Agents: Promise and Challenges. Cancer Treat Res 2023; 186:143-170. [PMID: 37978135 DOI: 10.1007/978-3-031-30065-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Better understanding of molecular drivers and dysregulated pathways has furthered the concept of precision oncology and rational drug development. The role of DNA damage response (DDR) pathways has been extensively studied in carcinogenesis and as potential therapeutic targets to improve response to chemotherapy or overcome resistance. Treatment with small molecule inhibitors of PARP has resulted in clinical response and conferred survival benefit to patients with ovarian cancer, BRCA-mutant breast cancer, HRD-deficient prostate cancer and BRCA-mutant pancreatic cancer, leading to US Food and Drug Administration (FDA) approvals. However, the observed clinical benefit with single agent PARP inhibitors is limited to few tumor types within the relevant genetic context. Since DDR pathways are essential for repair of damage caused by cytotoxic agents, PARP inhibitors have been evaluated in combination with various chemotherapeutic agents to broaden the therapeutic application of this class of drugs. In this chapter, we discuss the combination of PARP inhibitors with different chemotherapeutics agents, clinical experience to date, lessons learnt, and future directions for this approach.
Collapse
Affiliation(s)
- Kyaw Zin Thein
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| | - Rajat Thawani
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| | - Shivaani Kummar
- DeArmond Endowed Chair of Cancer Research, Division of Hematology and Medical Oncology, Clinical and Translational Research, Knight Cancer Institute (KCI), Center for Experimental Therapeutics (KCI), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, OC14HO, Portland, OR, 97239, USA.
| |
Collapse
|
5
|
Plummer R. Evolution of the Development of PARP Inhibitors. Cancer Treat Res 2023; 186:1-11. [PMID: 37978127 DOI: 10.1007/978-3-031-30065-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
PARP inhibitors first entered the clinic in 2003 in combination with DNA damaging agents in an attempt to overcome treatment resistance to established agents. A brief overview of ADP-ribosylator enzyme biology and the early preclinical development of the class is discussed, illustrating the multiple biological activities of these enzymes and potential wider clinical applicability. The chapter then documents those early years of clinical development and the evolution of the field and eventual registration of PARP inhibitors as active anticancer agents in their own right-in genetically vulnerable tumours.
Collapse
Affiliation(s)
- Ruth Plummer
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
6
|
PARP Inhibitors for Breast Cancer: Germline BRCA1/2 and Beyond. Cancers (Basel) 2022; 14:cancers14174332. [PMID: 36077867 PMCID: PMC9454726 DOI: 10.3390/cancers14174332] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/27/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Poly-adenosine diphosphate ribose polymerase (PARP) inhibitors (PARPi) are effective against tumors with mutations in DNA repair genes, most commonly in the BRCA1 and BRCA2 genes. Because these tumors are unable to repair their DNA, PARPi have been used to target DNA repair pathways and are useful in the treatment of breast cancers with some of these alterations. There are two FDA-approved PARPi for patients with breast cancer—olaparib and talazoparib. The data on olaparib and talazoparib in the treatment of breast cancer are summarized in this review, and we also explore potential future applications of PARPi beyond inherited BRCA mutations. Abstract Poly-adenosine diphosphate ribose polymerase (PARP) inhibitors (PARPi) are approved for BRCA1/2 carriers with HER2-negative breast cancer in the adjuvant setting with a high risk of recurrence as well as the metastatic setting. However, the indications for PARPi are broader for patients with other cancer types (e.g., prostate and ovarian cancer), involving additional biomarkers (e.g., ATM, PALB2, and CHEK) and genomic instability scores. Herein, we summarize the data on PARPi and breast cancer and discuss their use beyond BRCA carriers.
Collapse
|
7
|
Leal TA, Sharifi MN, Chan N, Wesolowski R, Turk AA, Bruce JY, O'Regan RM, Eickhoff J, Barroilhet LM, Malhotra J, Mehnert J, Girda E, Wiley E, Schmitz N, Andrews S, Liu G, Wisinski KB. A phase I study of talazoparib (BMN 673) combined with carboplatin and paclitaxel in patients with advanced solid tumors (NCI9782). Cancer Med 2022; 11:3969-3981. [PMID: 35396812 PMCID: PMC9636507 DOI: 10.1002/cam4.4724] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/21/2022] [Accepted: 03/13/2022] [Indexed: 11/25/2022] Open
Abstract
Background Inhibitors of poly(ADP‐ribose) polymerase (PARP) proteins potentiate antitumor activity of platinum chemotherapy. This study sought to determine the safety and tolerability of PARP inhibitor talazoparib with carboplatin and paclitaxel. Methods We conducted a phase I study of talazoparib with carboplatin AUC5‐6 and paclitaxel 80 mg/m2 days 1, 8, 15 of 21‐day cycles in patients with advanced solid tumors. Patients enrolled using a 3 + 3 design in two cohorts with talazoparib for 7 (schedule A) or 3 days (schedule B). After induction with 4–6 cycles of triplet therapy, patients received one of three maintenance options: (a) continuation of triplet (b) carboplatin/talazoparib, or (c) talazoparib monotherapy. Results Forty‐three patients were treated. The MTD for both schedules was talazoparib 250mcg daily. The main toxicity was myelosuppression including grade 3/4 hematologic treatment‐related adverse events (TRAEs). Dose modification occurred in 87% and 100% of patients for schedules A and B, respectively. Discontinuation due to TRAEs was 13% in schedule A and 10% in B. Ten out of 22 evaluable patients in schedule A and 5/16 patients in schedule B had a complete or partial response. Twelve out of 43 patients received ≥6 cycles of talazoparib after induction, with a 13‐month median duration of maintenance. Conclusion We have established the recommended phase II dose of Talazoparib at 250mcg on a 3‐ or 7‐day schedule with carboplatin AUC6 and paclitaxel 80 mg/m2 on days 1, 8, 15 of 21‐day cycles. This regimen is associated with significant myelosuppression, and in addition to maximizing supportive care, modification of the chemotherapy component would be a consideration for further development of this combination with the schedules investigated in this study.
Collapse
Affiliation(s)
| | - Marina N Sharifi
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Nancy Chan
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Robert Wesolowski
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Anita A Turk
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Justine Y Bruce
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Ruth M O'Regan
- Department of Medicine, University of Rochester, Rochester, New York, USA
| | - Jens Eickhoff
- Department of Biostatistics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Lisa M Barroilhet
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jyoti Malhotra
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Janice Mehnert
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York City, USA
| | - Eugenia Girda
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Elizabeth Wiley
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Natalie Schmitz
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Shannon Andrews
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Glenn Liu
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Kari B Wisinski
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| |
Collapse
|
8
|
In Silico Evaluation of a Promising Key Intermediate Thieno [2,3-d] Pyrimidine Derivative with Expected JAK2 Kinase Inhibitory Activity. MOLBANK 2022. [DOI: 10.3390/m1352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
This work describes the synthesis and the cytotoxic evaluation of thiophene and thienopyrimidine derivatives. The investigated compound was subjected to target prediction that indicated its high affinity to kinases and to Janus kinase 2 (JAK2) specifically. Molecular docking screening was performed on three different JAK2 proteins downloaded from the Protein Data Bank (PDB: 5AEP, 4C62 and 3ZMM). In vitro kinase inhibitory activity was evaluated and then compound cytotoxicity was performed on three different cancerous cell lines (HT-29, HepG-2, and MCF-7). Marked cytotoxic activity of the thienopyrimidine derivative against the HepG-2 cell line was demonstrated, reflected by its IC50 value of 8.001 ± 0.0445 μM, which is better than that of the reference standard (IC50 13.91 ± 2.170 μM). Pharmacokinetic studies revealed good well permeability and GI absorption with no violations against Lipinski’s rule.
Collapse
|
9
|
Sethy C, Kundu CN. PARP inhibitor BMN-673 induced apoptosis by trapping PARP-1 and inhibiting base excision repair via modulation of pol-β in chromatin of breast cancer cells. Toxicol Appl Pharmacol 2022; 436:115860. [PMID: 34998856 DOI: 10.1016/j.taap.2021.115860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 01/05/2023]
Abstract
PARP inhibitors emerged as clinically effective anti-tumor agents in combination with DNA damaging agents but the toxicity of DNA damaging agents and their off-target effects caused serious problems in cancer therapy. They confer cytotoxicity in cancer cells both by catalytic inhibition and trapping of PARP-1 at the DNA damage site. There is a lack of direct evidence to quantitatively determine the trapped PARP-1 in cellular DNA. Here, we have precisely evaluated the mechanism of PARP trapping mediated anti-cancer action of Quinacrine (QC), BMN-673, and their combination (QC + BMN-673) in breast cancer cells. We introduced a strategy to measure the cellular PARP trapping potentiality of BMN-673 in QC pretreated cells using a fluorescence-based assay system. It was found that QC+ BMN-673 induced apoptosis by triggering DNA damage in breast cancer cells. Treatment with QC + BMN-673 stimulated the expression of PARP-1 in the chromatin compared to that of PARP-2 and PARP-3. QC + BMN-673 treatment also caused a dose-dependent and time-dependent accumulation of PARP-1 and inhibition of PARylation in the chromatin. Upregulation of BER components (pol-β and FEN-1), an unchanged HR and NHEJ pathway proteins, and reduction of luciferase activity of the cells transfected with R-p21-P (LP-BER) were noted in combined drug-treated cells. Interestingly, silencing of pol-β resulted in unchanged PARP-1 trapping and PAR activity in the chromatin with increasing time after QC + BMN-673 treatment without altering APC and FEN-1 expression. Thus, our data suggested that the QC + BMN-673 augmented breast cancer cell death by pol-β mediated repair inhibition primarily through trapping of PARP-1 besides PARP-1 catalytic inhibition.
Collapse
Affiliation(s)
- Chinmayee Sethy
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
10
|
Elmongy EI, Attallah NGM, Altwaijry N, AlKahtani MM, Henidi HA. Design and Synthesis of New Thiophene/Thieno[2,3-d]pyrimidines along with Their Cytotoxic Biological Evaluation as Tyrosine Kinase Inhibitors in Addition to Their Apoptotic and Autophagic Induction. Molecules 2021; 27:molecules27010123. [PMID: 35011354 PMCID: PMC8746632 DOI: 10.3390/molecules27010123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
This work describes the synthesis and anticancer activity against kinase enzymes of newly designed thiophene and thieno[2,3-d]pyrimidine derivatives, along with their potential to activate autophagic and apoptotic cell death in cancer cells. The designed compounds were scanned for their affinity for kinases. The results were promising with affinity ranges from 46.7% to 13.3%. Molecular docking studies were performed, and the compounds were then screened for their antiproliferative effects. Interestingly, compounds 8 and 5 resulted in higher cytotoxic effects than the reference standard against MCF-7 and HepG-2. The compounds were evaluated for their induction of apoptosis and/or necrosis on HT-29 and HepG-2. Three compounds induced significant early apoptosis compared to untreated control HT-29 cells, and four derivatives were more significant compared to untreated HepG-2 cells. We further investigated the effect of four compounds on the autophagy process within HT-29, HepG-2, and MCF-7 cells with flow cytometry. Similar to the apoptosis results, compound 5 showed the highest autophagic induction among all compounds. The potential inhibitory activity of the synthesized compounds on kinases was assessed. Screened compounds showed inhibition activity ranging from 41.4% to 83.5%. Compounds recorded significant inhibition were further investigated for their specific FLT3 kinase inhibitory activity. Noticeably, Compound 5 exhibited the highest inhibitory activity against FLT3.
Collapse
Affiliation(s)
- Elshaymaa I. Elmongy
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh P.O. Box 84428, Saudi Arabia; (N.G.M.A.); (N.A.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo P.O. Box 11795, Egypt
- Correspondence: or
| | - Nashwah G. M. Attallah
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh P.O. Box 84428, Saudi Arabia; (N.G.M.A.); (N.A.)
- Egyptian Drug Authority (EDA) (Previously NODCAR), Giza 8655, Egypt
| | - Najla Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh P.O. Box 84428, Saudi Arabia; (N.G.M.A.); (N.A.)
| | - Manal Mubarak AlKahtani
- Research Department, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh P.O. Box 84428, Saudi Arabia; (M.M.A.); (H.A.H.)
| | - Hanan Ali Henidi
- Research Department, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh P.O. Box 84428, Saudi Arabia; (M.M.A.); (H.A.H.)
| |
Collapse
|
11
|
Perez-Fidalgo JA, Cortés A, Guerra E, García Y, Iglesias M, Bohn Sarmiento U, Calvo García E, Manso Sánchez L, Santaballa A, Oaknin A, Redondo A, Rubio MJ, González-Martín A. Olaparib in combination with pegylated liposomal doxorubicin for platinum-resistant ovarian cancer regardless of BRCA status: a GEICO phase II trial (ROLANDO study). ESMO Open 2021; 6:100212. [PMID: 34329939 PMCID: PMC8446804 DOI: 10.1016/j.esmoop.2021.100212] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND There is limited evidence for the benefit of olaparib in platinum-resistant ovarian cancer (PROC) patients with BRCA wild-type tumors. This study investigated whether this combination of a DNA-damaging chemotherapy plus olaparib is effective in PROC regardless BRCA status. PATIENTS AND METHODS Patients with high-grade serous or endometrioid ovarian carcinoma and one previous PROC recurrence were enrolled regardless of BRCA status. Patients with ≤4 previous lines (up to 5 in BRCA-mut) with at least one previous platinum-sensitive relapse were included; primary PROC was allowed only in case of BRCA-mut. Patients initially received six cycles of olaparib 300 mg b.i.d. (biduum) + intravenous pegylated liposomal doxorubicin (PLD) 40 mg/m2 (PLD40) every 28 days, followed by maintenance with olaparib 300 mg b.i.d. until progression or toxicity. The PLD dose was reduced to 30 mg/m2 (PLD30) due to toxicity. The primary endpoint was progression-free survival (PFS) at 6 months (6m-PFS) by RECIST version 1.1. A proportion of 40% 6m-PFS or more was considered of clinical interest. RESULTS From 2017 to 2020, 31 PROC patients were included. BRCA mutations were present in 16%. The median of previous lines was 2 (range 1-5). The overall disease control rate was 77% (partial response rate of 29% and stable disease rate of 48%). After a median follow-up of 10 months, the 6m-PFS and median PFS were 47% and 5.8 months, respectively. Grade ≥3 treatment-related adverse events occurred in 74% of patients, with neutropenia/anemia being the most frequent. With PLD30 serious AEs were less frequent than with PLD40 (21% versus 47%, respectively); moreover, PLD30 was associated with less PLD delays (32% versus 38%) and reductions (16% versus 22%). CONCLUSIONS The PLD-olaparib combination has shown significant activity in PROC regardless of BRCA status. PLD at 30 mg/m2 is better tolerated in the combination.
Collapse
Affiliation(s)
- J A Perez-Fidalgo
- Department of Medical Oncology, Hospital Clínico Universitario de Valencia, Valencia, Spain.
| | - A Cortés
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - E Guerra
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Y García
- Department of Medical Oncology, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Iglesias
- Department of Medical Oncology, Hospital Son Llatzer, Palma De Mallorca, Spain
| | - U Bohn Sarmiento
- Department of Medical Oncology, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas De Gran Canaria, Spain
| | - E Calvo García
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - L Manso Sánchez
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - A Santaballa
- Department of Medical Oncology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - A Oaknin
- Department of Medical Oncology, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - A Redondo
- Department of Medical Oncology, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
| | - M J Rubio
- Department of Medical Oncology, Hospital Universitario Reina Sofia, Córdoba, Spain
| | - A González-Martín
- Department of Medical Oncology Department, Clínica Universidad de Navarra, Madrid, Spain
| |
Collapse
|
12
|
Role of PARP1-mediated autophagy in EGFR-TKI resistance in non-small cell lung cancer. Sci Rep 2020; 10:20924. [PMID: 33262410 PMCID: PMC7708842 DOI: 10.1038/s41598-020-77908-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/06/2020] [Indexed: 12/29/2022] Open
Abstract
Resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) has become the main clinical challenge of advanced lung cancer. This research aimed to explore the role of PARP1-mediated autophagy in the progression of TKI therapy. PARP1-mediated autophagy was evaluated in vitro by CCK-8 assay, clonogenic assay, immunofluorescence, and western blot in the HCC-827, H1975, and H1299 cells treated with icotinib (Ico), rapamycin, and AZD2281 (olaparib) alone or in combination. Our results and GEO dataset analysis confirmed that PARP1 is expressed at lower levels in TKI-sensitive cells than in TKI-resistant cells. Low PARP1 expression and high p62 expression were associated with good outcomes among patients with NSCLC after TKI therapy. AZD2281 and a lysosomal inhibitor reversed resistance to Ico by decreasing PARP1 and LC3 in cells, but an mTOR inhibitor did not decrease Ico resistance. The combination of AZD2281 and Ico exerted a markedly enhanced antitumor effect by reducing PARP1 expression and autophagy in vivo. Knockdown of PARP1 expression reversed the resistance to TKI by the mTOR/Akt/autophagy pathway in HCC-827IR, H1975, and H1299 cells. PARP1-mediated autophagy is a key pathway for TKI resistance in NSCLC cells that participates in the resistance to TKIs. Olaparib may serve as a novel method to overcome the resistance to TKIs.
Collapse
|
13
|
Abstract
PARP (poly(ADP-ribose) polymerase) inhibitors represent a novel class of anti-cancer therapy; they take advantage of synthetic lethality and induce cell death by exploiting a defect in DNA repair. This class of medication was initially evaluated in patients with BRCA-associated tumors, but efficacy was also demonstrated in other populations. Since 2014, four PARP inhibitors have been approved in various indications: olaparib, niraparib, and rucaparib in high-grade serous ovarian cancer, and olaparib and talazoparib in metastatic breast cancer. The exact indications and study populations vary slightly between the different approvals in both disease states but there is significant overlap. PARP inhibitors continue to be investigated in ongoing clinical trials. In line with other targeted therapies, benefit appears to be strongest in a distinct population of patients with BRCA mutations or other defects in homologous recombination repair. Combination therapies, which include anti-angiogenesis agents and immunotherapy, show promise as a strategy to broaden efficacy for unselected patients. Initial studies of PARP inhibitors in combination with chemotherapy were limited by toxicity, but further studies are underway. To date, head-to-head trials comparing various PARP inhibitors have not been conducted, so questions remain in terms of choosing a PARP inhibitor to administer when indications overlap, as well as how to sequence these medications. Here we review both completed and ongoing clinical trials involving PARP inhibitors and mechanisms of resistance to this class of drugs.
Collapse
|
14
|
Cook SA, Tinker AV. PARP Inhibitors and the Evolving Landscape of Ovarian Cancer Management: A Review. BioDrugs 2019; 33:255-273. [PMID: 30895466 DOI: 10.1007/s40259-019-00347-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
As a drug class, inhibitors of poly-(ADP-ribose) polymerase (PARP) have had their greatest impact on the treatment of women with epithelial ovarian cancers (EOC), in particular, those with the most common histological subtype, high-grade serous cancer, as it has high rates of homologous recombination (HR) deficiency. PARP inhibition exploits this cancer vulnerability by further disrupting DNA repair, thus leading to genomic catastrophe. Early clinical data demonstrated the effectiveness of PARP inhibition in women with recurrent EOC harbouring BRCA1/2 mutations and those with platinum-sensitive recurrences. Three PARP inhibitors (olaparib, niraparib, and rucaparib) are now approved for use in women with recurrent EOC. Based upon randomised controlled trials, PARP inhibitors are in use as "maintenance" therapy for those with platinum-sensitive and platinum-responsive recurrences (irrespective of BRCA1/2 mutation status). Among women with BRCA1/2 mutations (either germline or somatic), maintenance PARP inhibitor therapy for those with recurrence has led to a nearly fourfold prolongation of progression-free survival compared to placebo control. Those without BRCA1/2 mutations experience an approximately twofold increase in progression-free survival. The latest clinical data demonstrate that women with BRCA1/2 mutations who respond to first-line chemotherapy and go on to have maintenance olaparib experience a doubling of the rate of freedom from death at 3 years when compared to placebo (60% vs 27%). PARP inhibitors are also approved as active therapy for women with germline or tumour BRCA1/2 mutations and recurrent EOC treated with three or more prior lines of therapy. Apart from the presence of a BRCA1/2 mutation (germline or somatic) and clinical factors such as platinum sensitivity and responsiveness, other predictive biomarkers are not in routine clinical use. Assays to identify genomic aberrations caused by HR deficiency, or mutations in genes involved in HR, have not been sufficiently sensitive to identify all patients who benefit from treatment. The mechanisms of PARP-inhibitor resistance include restoration of HR through reversion mutations in HR genes, capable of re-establishing the DNA open-reading frame and leading to resumed HR function. Other mechanisms that sustain sufficient DNA repair may also be important. This review focuses on the rationale for the use of PARP inhibitors in EOC. The data that have shaped clinical research are presented, and the trials that have changed management standards are reviewed and discussed. Highlighted are the past and ongoing efforts to further improve and explore the use of PARP inhibitors in EOC.
Collapse
Affiliation(s)
- Sarah A Cook
- BC Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Anna V Tinker
- BC Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada.
| |
Collapse
|
15
|
van der Noll R, Jager A, Ang JE, Marchetti S, Mergui-Roelvink MWJ, Lolkema MP, de Jonge MJA, van der Biessen DA, Brunetto AT, Arkenau HT, Tchakov I, Beijnen JH, de Bono JS, Schellens JHM. Phase I study of continuous olaparib capsule dosing in combination with carboplatin and/or paclitaxel (Part 1). Invest New Drugs 2019; 38:1117-1128. [PMID: 31667659 DOI: 10.1007/s10637-019-00856-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 09/12/2019] [Indexed: 11/26/2022]
Abstract
Background The PARP inhibitor olaparib has shown acceptable toxicity at doses of up to 400 mg twice daily (bid; capsule formulation) with encouraging signs of antitumor activity. Based on its mode of action, olaparib may sensitize tumor cells to DNA-damaging agents. This Phase I trial (NCT00516724) evaluated the safety, pharmacokinetics (PK) and preliminary efficacy of olaparib combined with carboplatin and/or paclitaxel. Methods Patients with advanced solid tumors received olaparib (capsule bid) plus carboplatin (Part A), carboplatin and paclitaxel (Part B), or paclitaxel (Part C). In each part of the study, different drug doses were given to define the most appropriate dose/drug combination to use in further studies. Safety assessments included evaluation of dose-limiting toxicities (DLTs; cycle 1 only), adverse events (AEs) and physical examinations. PK assessments of olaparib, carboplatin and paclitaxel were performed. Tumor responses (RECIST) were assessed every two cycles. Results Fifty-seven patients received treatment. DLTs were reported in two patients (both receiving olaparib 100 mg bid and carboplatin AUC 4; Part A, cohort 2): grade 1 thrombocytopenia with grade 2 neutropenia lasting for 16 days, and grade 2 neutropenia lasting for 7 days. Non-hematologic AEs were predominantly grade 1-2 and included fatigue (70%) and nausea (40%). Bone marrow suppression, mainly neutropenia (51%) and thrombocytopenia (25%), frequently led to dose modifications. Conclusions Olaparib in combination with carboplatin and/or paclitaxel resulted in increased hematologic toxicities, making it challenging to establish a dosing regimen that could be tolerated for multiple cycles without dose modifications.
Collapse
Affiliation(s)
- Ruud van der Noll
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
| | - Agnes Jager
- Department of Medical Oncology, Erasmus University MC Cancer Institute, PO Box 5201, 3008, AE, Rotterdam, The Netherlands
| | - Joo Ern Ang
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Serena Marchetti
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - Marja W J Mergui-Roelvink
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus University MC Cancer Institute, PO Box 5201, 3008, AE, Rotterdam, The Netherlands
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus University MC Cancer Institute, PO Box 5201, 3008, AE, Rotterdam, The Netherlands
| | - Diane A van der Biessen
- Department of Medical Oncology, Erasmus University MC Cancer Institute, PO Box 5201, 3008, AE, Rotterdam, The Netherlands
| | - Andre T Brunetto
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Hendrik-Tobias Arkenau
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Ilian Tchakov
- AstraZeneca, Alderley Park, Park Estate, Macclesfield, SK10 4TF, UK
- Eisai, Mosquito Way, Hatfield, AL10 9SN, UK
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
- Utrecht Institute of Pharmaceutical Science s (UIPS), Utrecht University, Domplein 29, 3512, JE, Utrecht, The Netherlands
| | - Johann S de Bono
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
- Utrecht Institute of Pharmaceutical Science s (UIPS), Utrecht University, Domplein 29, 3512, JE, Utrecht, The Netherlands
| |
Collapse
|
16
|
Houghton PJ, Kurmasheva RT. Challenges and Opportunities for Childhood Cancer Drug Development. Pharmacol Rev 2019; 71:671-697. [PMID: 31558580 PMCID: PMC6768308 DOI: 10.1124/pr.118.016972] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer in children is rare with approximately 15,700 new cases diagnosed in the United States annually. Through use of multimodality therapy (surgery, radiation therapy, and aggressive chemotherapy), 70% of patients will be "cured" of their disease, and 5-year event-free survival exceeds 80%. However, for patients surviving their malignancy, therapy-related long-term adverse effects are severe, with an estimated 50% having chronic life-threatening toxicities related to therapy in their fourth or fifth decade of life. While overall intensive therapy with cytotoxic agents continues to reduce cancer-related mortality, new understanding of the molecular etiology of many childhood cancers offers an opportunity to redirect efforts to develop effective, less genotoxic therapeutic options, including agents that target oncogenic drivers directly, and the potential for use of agents that target the tumor microenvironment and immune-directed therapies. However, for many high-risk cancers, significant challenges remain.
Collapse
Affiliation(s)
- Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, University of Texas Health, San Antonio, Texas
| |
Collapse
|
17
|
Zhang D, Baldwin P, Leal AS, Carapellucci S, Sridhar S, Liby KT. A nano-liposome formulation of the PARP inhibitor Talazoparib enhances treatment efficacy and modulates immune cell populations in mammary tumors of BRCA-deficient mice. Am J Cancer Res 2019; 9:6224-6238. [PMID: 31534547 PMCID: PMC6735511 DOI: 10.7150/thno.36281] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
Two recently approved PARP inhibitors provide an important new therapeutic option for patients with BRCA-mutated metastatic breast cancer. PARP inhibitors significantly prolong progression-free survival in patients, but conventional oral delivery of PARP inhibitors is hindered by limited bioavailability and off-target toxicities, thus compromising the therapeutic benefits and quality of life for patients. Here, we developed a new delivery system, in which the PARP inhibitor Talazoparib is encapsulated in the bilayer of a nano-liposome, to overcome these limitations. Methods: Nano-Talazoparib (NanoTLZ) was characterized both in vitro and in vivo. The therapeutic efficacy and toxicity of Nano-Talazoparib (NanoTLZ) were evaluated in BRCA-deficient mice. The regulation of NanoTLZ on gene transcription and immunomodulation were further investigated in spontaneous BRCA-deficient tumors. Results: NanoTLZ significantly (p<0.05) prolonged the overall survival of BRCA-deficient mice compared to all of the other experimental groups, including saline control, empty nanoparticles, and free Talazoparib groups (oral and i.v.). Moreover, NanoTLZ was better tolerated than treatment with free Talazoparib, with no significant weight lost or alopecia as was observed with the free drug. After 5 doses, NanoTLZ altered the expression of over 140 genes and induced DNA damage, cell cycle arrest and inhibition of cell proliferation in the tumor. In addition, NanoTLZ favorably modulated immune cell populations in vivo and significantly (p<0.05) decreased the percentage of myeloid derived suppressor cells in both the tumor and spleen compared to control groups. Conclusions: Our results demonstrate that delivering nanoformulated Talazoparib not only enhances treatment efficacy but also reduces off-target toxicities in BRCA-deficient mice; the same potential is predicted for patients with BRCA-deficient breast cancer.
Collapse
|
18
|
Toma M, Skorski T, Sliwinski T. DNA Double Strand Break Repair - Related Synthetic Lethality. Curr Med Chem 2019; 26:1446-1482. [PMID: 29421999 DOI: 10.2174/0929867325666180201114306] [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] [Received: 09/05/2017] [Revised: 11/10/2017] [Accepted: 11/16/2017] [Indexed: 12/25/2022]
Abstract
Cancer is a heterogeneous disease with a high degree of diversity between and within tumors. Our limited knowledge of their biology results in ineffective treatment. However, personalized approach may represent a milestone in the field of anticancer therapy. It can increase specificity of treatment against tumor initiating cancer stem cells (CSCs) and cancer progenitor cells (CPCs) with minimal effect on normal cells and tissues. Cancerous cells carry multiple genetic and epigenetic aberrations which may disrupt pathways essential for cell survival. Discovery of synthetic lethality has led a new hope of creating effective and personalized antitumor treatment. Synthetic lethality occurs when simultaneous inactivation of two genes or their products causes cell death whereas individual inactivation of either gene is not lethal. The effectiveness of numerous anti-tumor therapies depends on induction of DNA damage therefore tumor cells expressing abnormalities in genes whose products are crucial for DNA repair pathways are promising targets for synthetic lethality. Here, we discuss mechanistic aspects of synthetic lethality in the context of deficiencies in DNA double strand break repair pathways. In addition, we review clinical trials utilizing synthetic lethality interactions and discuss the mechanisms of resistance.
Collapse
Affiliation(s)
- Monika Toma
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Tomasz Skorski
- Department of Microbiology and Immunology, 3400 North Broad Street, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, United States
| | - Tomasz Sliwinski
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
19
|
PARP Inhibition Increases the Response to Chemotherapy in Uveal Melanoma. Cancers (Basel) 2019; 11:cancers11060751. [PMID: 31146482 PMCID: PMC6628115 DOI: 10.3390/cancers11060751] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/16/2019] [Accepted: 05/27/2019] [Indexed: 12/18/2022] Open
Abstract
Uveal melanoma (UM) remains without effective therapy at the metastatic stage, which is associated with BAP-1 (BRCA1 associated protein) mutations. However, no data on DNA repair capacities in UM are available. Here, we use UM patient-derived xenografts (PDXs) to study the therapeutic activity of the PARP inhibitor olaparib, alone or in combination. First, we show that the expression and the activity of PARP proteins is similar between the PDXs and the corresponding patient’s tumors. In vivo experiments in the PDX models showed that olaparib was not efficient alone, but significantly increased the efficacy of dacarbazine. Finally, using reverse phase protein arrays and immunohistochemistry, we identified proteins involved in DNA repair and apoptosis as potential biomarkers predicting response to the combination of olaparib and dacarbazine. We also observed a high increase of phosphorylated YAP and TAZ proteins after dacarbazine + olaparib treatment. Our results suggest that PARP inhibition in combination with the alkylating agent dacarbazine could be of clinical interest for UM treatment. We also observe an interesting effect of dacarbazine on the Hippo pathway, confirming the importance of this pathway in UM.
Collapse
|
20
|
Buechel M, Herzog TJ, Westin SN, Coleman RL, Monk BJ, Moore KN. Treatment of patients with recurrent epithelial ovarian cancer for whom platinum is still an option. Ann Oncol 2019; 30:721-732. [PMID: 30887020 PMCID: PMC8887593 DOI: 10.1093/annonc/mdz104] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Ovarian cancer remains the most deadly gynecologic cancer with the majority of patients relapsing within 3 years of diagnosis. Traditional treatment paradigms linked to platinum sensitivity or resistance are currently being questioned in the setting of new diagnostic methods and treatment options. DESIGN Authors carried out review of the literature on key topics in treatment of recurrent epithelial ovarian cancer (EOC) when platinum is still an option; including secondary surgical cytoreduction, chemotherapy, novel treatment options, and maintenance therapy. A treatment algorithm is proposed. RESULTS Molecular characterization of EOC is critical to help guide treatment decisions. The role of secondary cytoreductive surgery is currently being evaluated with results from Gynecologic Oncology Group (GOG) 213 and anticipated results from DESKTOP III clinical trials. Chemotherapy backbone has remained relatively unchanged but utilizing non-platinum-based regimens is under investigation. In addition, maintenance therapy with anti-angiogenic therapy and Poly (ADP-ribose) Polymerase (PARP) inhibitors has emerged as the standard of care. Novel combinations, including immunotherapy and anti-angiogenesis agents, may further change the current landscape. CONCLUSIONS The treatment of recurrent EOC is rapidly changing. Clinical trial design will need to continue to evolve as many novel therapies move to the upfront setting. Ultimately, the treatment of patients with recurrent EOC must incorporate individual patient and tumor factors.
Collapse
Affiliation(s)
- M Buechel
- Section of Gynecologic Oncology, Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City.
| | - T J Herzog
- Division of Gynecologic Oncology, University of Cincinnati Cancer Institute, University of Cincinnati, Cincinnati
| | - S N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - R L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - B J Monk
- Division of Gynecologic Oncology, Arizona Oncology, Phoenix, USA
| | - K N Moore
- Section of Gynecologic Oncology, Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City
| |
Collapse
|
21
|
Yonemori K, Shimomura A, Yasojima H, Masuda N, Aogi K, Takahashi M, Naito Y, Shimizu S, Nakamura R, Hashimoto J, Yamamoto H, Hirakawa A, Michimae H, Hamada A, Yoshida T, Sukigara T, Tamura K, Fujiwara Y. A phase I/II trial of olaparib tablet in combination with eribulin in Japanese patients with advanced or metastatic triple-negative breast cancer previously treated with anthracyclines and taxanes. Eur J Cancer 2019; 109:84-91. [DOI: 10.1016/j.ejca.2018.11.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/09/2018] [Accepted: 11/10/2018] [Indexed: 01/17/2023]
|
22
|
LaFargue CJ, Dal Molin GZ, Sood AK, Coleman RL. Exploring and comparing adverse events between PARP inhibitors. Lancet Oncol 2019; 20:e15-e28. [PMID: 30614472 PMCID: PMC7292736 DOI: 10.1016/s1470-2045(18)30786-1] [Citation(s) in RCA: 336] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022]
Abstract
Ovarian cancer remains one of the most challenging malignancies to treat. Targeted therapies such as poly (ADP-ribose) polymerase (PARP) inhibitors have emerged as one of the most exciting new treatments for ovarian cancer, particularly in women with BRCA1 or BRCA2 mutations or those without a functional homologous recombination repair pathway. Perhaps the most advantageous characteristic of PARP inhibitors is their mechanism of action, which targets cancer cells on the basis of their inherent deficiencies while seemingly avoiding normally functioning cells. Although health-care providers might assume a low toxicity profile because of their specific mechanism of action, PARP inhibitors are not completely benign and overall show a class effect adverse-event profile. Further complicating this situation, three different PARP inhibitors have been approved by the US Food and Drug Administration since 2014, each with their own specific indications and individual toxicity profiles. The diversity of adverse events seen both within and across this class of drug underscores the importance of having a comprehensive reference to help guide clinical decision making when treating patients. This Review characterises and compares all toxicities associated with each PARP inhibitor, both in monotherapy and in novel combinations with other drugs, with a particular focus on potential management strategies to help mitigate toxic effects. Although the excitement surrounding PARP inhibitors might certainly be warranted, a thorough understanding of all associated toxicities is imperative to ensure that patients can achieve maximal clinical benefit.
Collapse
Affiliation(s)
- Christopher J LaFargue
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Graziela Z Dal Molin
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Cseh AM, Fábián Z, Sümegi B, Scorrano L. Poly(adenosine diphosphate-ribose) polymerase as therapeutic target: lessons learned from its inhibitors. Oncotarget 2018; 8:50221-50239. [PMID: 28430591 PMCID: PMC5564845 DOI: 10.18632/oncotarget.16859] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 01/27/2023] Open
Abstract
Poly(ADP-ribose) polymerases are a family of DNA-dependent nuclear enzymes catalyzing the transfer of ADP-ribose moieties from cellular nicotinamide-adenine-dinucleotide to a variety of target proteins. Although they have been considered as resident nuclear elements of the DNA repair machinery, recent works revealed a more intricate physiologic role of poly(ADP-ribose) polymerases with numerous extranuclear activities. Indeed, poly(ADP-ribose) polymerases participate in fundamental cellular processes like chromatin remodelling, transcription or regulation of the cell-cycle. These new insight into the physiologic roles of poly(ADP-ribose) polymerases widens the range of human pathologies in which pharmacologic inhibition of these enzymes might have a therapeutic potential. Here, we overview our current knowledge on extranuclear functions of poly(ADP-ribose) polymerases with a particular focus on the mitochondrial ones and discuss potential fields of future clinical applications.
Collapse
Affiliation(s)
- Anna Mária Cseh
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary.,Department of Biology, University of Padova, Padova, Italy
| | - Zsolt Fábián
- Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Balázs Sümegi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Luca Scorrano
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
24
|
Song L, McNeil EM, Ritchie AM, Astell KR, Gourley C, Melton DW. Melanoma cells replicate through chemotherapy by reducing levels of key homologous recombination protein RAD51 and increasing expression of translesion synthesis DNA polymerase ζ. BMC Cancer 2017; 17:864. [PMID: 29254481 PMCID: PMC5735618 DOI: 10.1186/s12885-017-3864-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 11/30/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The global incidence of melanoma has been increasing faster than any other form of cancer. New therapies offer exciting prospects for improved survival, but the development of resistance is a major problem and there remains a need for additional effective melanoma therapy. Platinum compounds, such as cisplatin, are the most effective chemotherapeutics for a number of major cancers, but are ineffective on metastatic melanoma. They cause monofunctional adducts and intrastrand crosslinks that are repaired by nucleotide excision repair, as well as the more toxic interstrand crosslinks that are repaired by a combination of nuclease activity and homologous recombination. METHODS We investigated the mechanism of melanoma resistance to cisplatin using a panel of melanoma and control cell lines. Cisplatin-induced changes in levels of the key homologous recombination protein RAD51 and compensatory changes in translesion synthesis DNA polymerases were identified by western blotting and qRT-PCR. Flow cytometry, immunofluorescence and western blotting were used to compare the cell cycle and DNA damage response and the induction of apoptosis in cisplatin-treated melanoma and control cells. Ectopic expression of a tagged form of RAD51 and siRNA knockdown of translesion synthesis DNA polymerase zeta were used to investigate the mechanism that allowed cisplatin-treated melanoma cells to continue to replicate. RESULTS We have identified and characterised a novel DNA damage response mechanism in melanoma. Instead of increasing levels of RAD51 on encountering cisplatin-induced interstrand crosslinks during replication, melanoma cells shut down RAD51 synthesis and instead boost levels of translesion synthesis DNA polymerase zeta to allow replication to proceed. This response also resulted in synthetic lethality to the PARP inhibitor olaparib. CONCLUSIONS This unusual DNA damage response may be a more appropriate strategy for an aggressive and rapidly growing tumour like melanoma that enables it to better survive chemotherapy, but also results in increased sensitivity of cultured melanoma cells to the PARP inhibitor olaparib.
Collapse
Affiliation(s)
- Liang Song
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Ewan M McNeil
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Ann-Marie Ritchie
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Katy R Astell
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK.,Present Address: Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Charlie Gourley
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - David W Melton
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
25
|
Nishio S, Takekuma M, Takeuchi S, Kawano K, Tsuda N, Tasaki K, Takahashi N, Abe M, Tanaka A, Nagasawa T, Shoji T, Xiong H, Nuthalapati S, Leahy T, Hashiba H, Kiriyama T, Komarnitsky P, Hirashima Y, Ushijima K. Phase 1 study of veliparib with carboplatin and weekly paclitaxel in Japanese patients with newly diagnosed ovarian cancer. Cancer Sci 2017; 108:2213-2220. [PMID: 28837250 PMCID: PMC5665762 DOI: 10.1111/cas.13381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/31/2017] [Accepted: 08/09/2017] [Indexed: 01/06/2023] Open
Abstract
This phase 1, open-label, dose-escalation study was conducted to determine the safety, tolerability, pharmacokinetics and preliminary efficacy of veliparib with carboplatin and weekly paclitaxel in Japanese women with newly diagnosed, advanced ovarian cancer. Patients received veliparib at 100 or 150 mg b.i.d. on days 1-21 with carboplatin (area under the concentration-time curve 6 mg/mL•min) on day 1 and paclitaxel 80 mg/m2 on days 1, 8 and 15 every 3 weeks for up to 6 21-day cycles. Dose escalation followed a 3 + 3 design to determine dose-limiting toxicities, maximum tolerated dose and the recommended phase 2 dose. Nine patients (median age 62 [range 27-72] years) received a median of 5 (range 3-6) cycles of treatment (3 at 100 mg, 6 at 150 mg). There were no dose-limiting toxicities. The most common adverse events of any grade were neutropenia (100%), alopecia (89%), peripheral sensory neuropathy (78%), and anemia, nausea and malaise (67% each). Grade 3 or 4 adverse events were associated with myelosuppression. Pharmacokinetics of carboplatin/paclitaxel were similar at both veliparib doses. Response, assessed in five patients, was partial in four and complete in one (objective response rate 100%). The response could not be assessed in four patients who had no measurable disease at baseline. The recommended phase 2 dose of veliparib, when combined with carboplatin/paclitaxel, is 150 mg b.i.d. Findings from this phase 1 trial demonstrate the tolerability and safety of veliparib with carboplatin/paclitaxel, a regimen with potential clinical benefit in Japanese women with ovarian cancer.
Collapse
Affiliation(s)
- Shin Nishio
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Munetaka Takekuma
- Division of Gynecology, Shizuoka Cancer Center, Nagaizumi, Shizuoka, Japan
| | - Satoshi Takeuchi
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Iwate, Japan
| | - Kouichirou Kawano
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Naotake Tsuda
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kazuto Tasaki
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Nobutaka Takahashi
- Division of Gynecology, Shizuoka Cancer Center, Nagaizumi, Shizuoka, Japan
| | - Masakazu Abe
- Division of Gynecology, Shizuoka Cancer Center, Nagaizumi, Shizuoka, Japan
| | - Aki Tanaka
- Division of Gynecology, Shizuoka Cancer Center, Nagaizumi, Shizuoka, Japan
| | - Takayuki Nagasawa
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Iwate, Japan
| | - Tadahiro Shoji
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Iwate, Japan
| | - Hao Xiong
- AbbVie, North Chicago, Illinois, USA
| | | | | | | | | | | | - Yasuyuki Hirashima
- Division of Gynecology, Shizuoka Cancer Center, Nagaizumi, Shizuoka, Japan
| | - Kimio Ushijima
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
26
|
Pignochino Y, Capozzi F, D'Ambrosio L, Dell'Aglio C, Basiricò M, Canta M, Lorenzato A, Vignolo Lutati F, Aliberti S, Palesandro E, Boccone P, Galizia D, Miano S, Chiabotto G, Napione L, Gammaitoni L, Sangiolo D, Benassi MS, Pasini B, Chiorino G, Aglietta M, Grignani G. PARP1 expression drives the synergistic antitumor activity of trabectedin and PARP1 inhibitors in sarcoma preclinical models. Mol Cancer 2017; 16:86. [PMID: 28454547 PMCID: PMC5410089 DOI: 10.1186/s12943-017-0652-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 04/17/2017] [Indexed: 01/05/2023] Open
Abstract
Background Enhancing the antitumor activity of the DNA-damaging drugs is an attractive strategy to improve current treatment options. Trabectedin is an isoquinoline alkylating agent with a peculiar mechanism of action. It binds to minor groove of DNA inducing single- and double-strand-breaks. These kinds of damage lead to the activation of PARP1, a first-line enzyme in DNA-damage response pathways. We hypothesized that PARP1 targeting could perpetuate trabectedin-induced DNA damage in tumor cells leading finally to cell death. Methods We investigated trabectedin and PARP1 inhibitor synergism in several tumor histotypes both in vitro and in vivo (subcutaneous and orthotopic tumor xenografts in mice). We searched for key determinants of drug synergism by comparative genomic hybridization (aCGH) and gene expression profiling (GEP) and validated their functional role. Results Trabectedin activated PARP1 enzyme and the combination with PARP1 inhibitors potentiated DNA damage, cell cycle arrest at G2/M checkpoint and apoptosis, if compared to single agents. Olaparib was the most active PARP1 inhibitor to combine with trabectedin and we confirmed the antitumor and antimetastatic activity of trabectedin/olaparib combination in mice models. However, we observed different degree of trabectedin/olaparib synergism among different cell lines. Namely, in DMR leiomyosarcoma models the combination was significantly more active than single agents, while in SJSA-1 osteosarcoma models no further advantage was obtained if compared to trabectedin alone. aCGH and GEP revealed that key components of DNA-repair pathways were involved in trabectedin/olaparib synergism. In particular, PARP1 expression dictated the degree of the synergism. Indeed, trabectedin/olaparib synergism was increased after PARP1 overexpression and reduced after PARP1 silencing. Conclusions PARP1 inhibition potentiated trabectedin activity in a PARP1-dependent manner and PARP1 expression in tumor cells might be a useful predictive biomarker that deserves clinical evaluation. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0652-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ymera Pignochino
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy. .,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy.
| | - Federica Capozzi
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | - Lorenzo D'Ambrosio
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | - Carmine Dell'Aglio
- Pathology Unit, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy
| | - Marco Basiricò
- Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy
| | - Marta Canta
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | - Annalisa Lorenzato
- Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | | | - Sandra Aliberti
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy
| | - Erica Palesandro
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | - Paola Boccone
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | - Danilo Galizia
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | - Sara Miano
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy
| | - Giulia Chiabotto
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy
| | - Lucia Napione
- Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy.,Laboratory of Vascular Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.,Current address: Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Loretta Gammaitoni
- Laboratory of Vascular Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy.,Laboratory of Vascular Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy
| | - Maria Serena Benassi
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Barbara Pasini
- Department of Genetics, Biology and Biochemistry, University of Torino, Torino, Italy
| | | | - Massimo Aglietta
- Department of Oncology, University of Torino Medical School, Candiolo, Torino, Italy.,Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy
| | - Giovanni Grignani
- Sarcoma Unit, Medical Oncology, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Torino, Italy.
| |
Collapse
|
27
|
Valdez BC, Li Y, Murray D, Liu Y, Nieto Y, Champlin RE, Andersson BS. The PARP inhibitor olaparib enhances the cytotoxicity of combined gemcitabine, busulfan and melphalan in lymphoma cells. Leuk Lymphoma 2017; 58:2705-2716. [PMID: 28394191 DOI: 10.1080/10428194.2017.1306647] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The combination of gemcitabine (Gem), busulfan (Bu), and melphalan (Mel) is a promising regimen for autologous stem-cell transplantation (SCT) for lymphomas. To further improve the efficacy of [Gem + Bu + Mel], we added poly(ADP-ribose) polymerase (PARP) inhibitor olaparib (Ola). We hypothesized that Ola would inhibit the repair of damaged DNA caused by [Gem + Bu + Mel]. Exposure of J45.01 and Toledo cell lines to IC10-20 of individual drug inhibited proliferation by 6-16%; [Gem + Bu + Mel] by 20-27%; and [Gem + Bu + Mel + Ola] by 61-67%. The synergistic cytotoxicity of the four-drug combination may be attributed to activation of the DNA-damage response, inhibition of PARP activity and DNA repair, decreased mitochondrial membrane potential, increased production of reactive oxygen species, and activation of the SAPK/JNK stress signaling pathway, all of which may enhance apoptosis. Similar observations were obtained using mononuclear cells isolated from patients with T-cell lymphocytic leukemia. Our results provide a rationale for undertaking clinical trials of this drug combination for lymphoma patients undergoing SCT.
Collapse
Affiliation(s)
- Benigno C Valdez
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Yang Li
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - David Murray
- b Department of Experimental Oncology , Cross Cancer Institute , Edmonton , Canada
| | - Yan Liu
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Yago Nieto
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Richard E Champlin
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Borje S Andersson
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
28
|
Du Y, Yamaguchi H, Hsu JL, Hung MC. PARP inhibitors as precision medicine for cancer treatment. Natl Sci Rev 2017. [DOI: 10.1093/nsr/nwx027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AbstractPersonalized or precision medicine is an emerging treatment approach tailored to individuals or certain groups of patients based on their unique characteristics. These types of therapies guided by biomarkers tend to be more effective than traditional approaches, especially in cancer. The inhibitor against poly (ADP-ribose) polymerase (PARP), olaparib (Lynparza, AstraZeneca), which was approved by the US Food and Drug Administration (FDA) in 2014, demonstrated efficacy specifically for ovarian cancer patients harboring mutations in BRCA genes, which encode proteins in DNA double-strand break repairs. However, the response to PARP inhibitors has been less encouraging in other cancer types that also carry defects in the BRCA genes. Thus, furthering our understanding of the underlying mechanism of PARP inhibitors and resistance is critical to improve their efficacy. In this review, we summarize the results of preclinical studies and the clinical application of PARP inhibitors, and discuss the future direction of PARP inhibitors as a potential marker-guided personalized medicine for cancer treatment.
Collapse
Affiliation(s)
- Yi Du
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
| | - Hirohito Yamaguchi
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
| | - Jennifer L. Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 40402
- Department of Biotechnology, Asia University, Taichung 41354
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 40402
- Department of Biotechnology, Asia University, Taichung 41354
| |
Collapse
|
29
|
Wilson RH, Evans TRJ, Middleton MR, Molife LR, Spicer J, Dieras V, Roxburgh P, Giordano H, Jaw-Tsai S, Goble S, Plummer R. A phase I study of intravenous and oral rucaparib in combination with chemotherapy in patients with advanced solid tumours. Br J Cancer 2017; 116:884-892. [PMID: 28222073 PMCID: PMC5379148 DOI: 10.1038/bjc.2017.36] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/14/2016] [Accepted: 01/20/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND This study evaluated safety, pharmacokinetics, and clinical activity of intravenous and oral rucaparib, a poly(ADP-ribose) polymerase inhibitor, combined with chemotherapy in patients with advanced solid tumours. METHODS Initially, patients received escalating doses of intravenous rucaparib combined with carboplatin, carboplatin/paclitaxel, cisplatin/pemetrexed, or epirubicin/cyclophosphamide. Subsequently, the study was amended to focus on oral rucaparib (once daily, days 1-14) combined with carboplatin (day 1) in 21-day cycles. Dose-limiting toxicities (DLTs) were assessed in cycle 1 and safety in all cycles. RESULTS Eighty-five patients were enrolled (22 breast, 15 ovarian/peritoneal, and 48 other primary cancers), with a median of three prior therapies (range, 1-7). Neutropenia (27.1%) and thrombocytopenia (18.8%) were the most common grade ⩾3 toxicities across combinations and were DLTs with the oral rucaparib/carboplatin combination. Maximum tolerated dose for the combination was 240 mg per day oral rucaparib and carboplatin area under the curve 5 mg ml-1 min-1. Oral rucaparib demonstrated dose-proportional kinetics, a long half-life (≈17 h), and good bioavailability (36%). Pharmacokinetics were unchanged by carboplatin coadministration. The rucaparib/carboplatin combination had radiologic antitumour activity, primarily in BRCA1- or BRCA2-mutated breast and ovarian/peritoneal cancers. CONCLUSIONS Oral rucaparib can be safely combined with a clinically relevant dose of carboplatin in patients with advanced solid tumours (Trial registration ID: NCT01009190).
Collapse
Affiliation(s)
- Richard H Wilson
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
- Northern Ireland Cancer Center, Belfast City Hospital, 51 Lisburn Road, Belfast BT9 7AB, UK
| | - TR Jeffry Evans
- Beatson West of Scotland Cancer Centre, and Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Mark R Middleton
- National Institute for Health Research Biomedical Research Centre, Churchill Hospital, and Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - L Rhoda Molife
- Drug Development Unit, Royal Marsden Hospital/Institute of Cancer Research, Downs Road, Sutton, Surrey SM2 5PT, UK
| | - James Spicer
- Division of Cancer Studies, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Veronique Dieras
- Department of Medical Oncology, Institut Curie, 26, rue d'Ulm, Paris 75005 France
| | - Patricia Roxburgh
- Beatson West of Scotland Cancer Centre, and Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Heidi Giordano
- Clovis Oncology, Inc., Boulder, 5500 Flatiron Parkway, Boulder, CO 80301, USA
| | - Sarah Jaw-Tsai
- Clovis Oncology, Inc., Boulder, 5500 Flatiron Parkway, Boulder, CO 80301, USA
| | - Sandra Goble
- Clovis Oncology, Inc., Boulder, 5500 Flatiron Parkway, Boulder, CO 80301, USA
| | - Ruth Plummer
- Northern Centre for Cancer Care, Freeman Hospital, Freeman Road, High Heaton, Newcastle Upon Tyne NE7 7DN, UK
- Northern Institute for Cancer Research, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| |
Collapse
|
30
|
Bao Z, Cao C, Geng X, Tian B, Wu Y, Zhang C, Chen Z, Li W, Shen H, Ying S. Effectiveness and safety of poly (ADP-ribose) polymerase inhibitors in cancer therapy: A systematic review and meta-analysis. Oncotarget 2016; 7:7629-39. [PMID: 26399274 PMCID: PMC4884943 DOI: 10.18632/oncotarget.5367] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/11/2015] [Indexed: 12/30/2022] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors are a class of small-molecule drugs suppressing PARP enzymes activity, inducing the death of cells deficient in homologous recombination repair (HRR). HRR deficiency is common in tumor cells with BRCA gene mutation. Since their first clinical trial in 2003, PARP inhibitors have shown benefit in the treatment of HRR-deficient tumors. Recently, several randomized clinical trials (RCTs) have been conducted to investigate the potential benefit of administration of PARP inhibitors in cancer patients. However, the results remain controversial. To evaluate the efficiency and safety of PARP inhibitors in patients with cancer, we performed a comprehensive meta-analysis of RCTs. According to our study, PARP inhibitors could clearly improve progression-free survival (PFS), especially in patients with BRCA mutation. However, our study showed no significant difference in overall survival (OS) between the PARP inhibitors and controls, even in the BRCA mutation group. Little toxicity was reported in the rate of treatment correlated adverse events (AEs) in PARP inhibitor group compared with controls. In conclusion, PARP inhibitors do well in improving PFS with little toxicity, especially in patients with BRCA deficiency.
Collapse
Affiliation(s)
- Zhengqiang Bao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinwei Geng
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Baoping Tian
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanping Wu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Zhang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Chen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,State Key Laboratory for Respiratory Diseases, Guangzhou, China
| | - Songmin Ying
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
31
|
A comprehensive look of poly(ADP-ribose) polymerase inhibition strategies and future directions for cancer therapy. Future Med Chem 2016; 9:37-60. [PMID: 27995810 DOI: 10.4155/fmc-2016-0113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The finding of promising drugs represents a huge challenge in cancer therapeutics, therefore it is important to seek out novel approaches and elucidate essential cellular processes in order to identify potential drug targets. Studies on DNA repair pathway suggested that an enzyme, PARP, which plays a significant role in DNA repair responses, could be targeted in cancer therapy. Hence, the efficacy of PARP inhibitors in cancer therapy has been investigated and has progressed from the laboratory to clinics, with olaparib having already been approved by the US FDA for ovarian cancer treatment. Here, we have discussed the development of PARP inhibitors, strategies to improve their selectivity and efficacy, including innovative combinational and synthetic lethality approaches to identify effective PARP inhibitors in cancer treatment.
Collapse
|
32
|
Piao J, Takai S, Kamiya T, Inukai T, Sugita K, Ohyashiki K, Delia D, Masutani M, Mizutani S, Takagi M. Poly (ADP-ribose) polymerase inhibitors selectively induce cytotoxicity in TCF3-HLF-positive leukemic cells. Cancer Lett 2016; 386:131-140. [PMID: 27894958 DOI: 10.1016/j.canlet.2016.11.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 10/20/2022]
Abstract
Poly (ADP-ribose) polymerase (PARP) is an indispensable component of the DNA repair machinery. PARP inhibitors are used as cutting-edge treatments for patients with homologous recombination repair (HRR)-defective breast cancers harboring mutations in BRCA1 or BRCA2. Other tumors defective in HRR, including some hematological malignancies, are predicted to be good candidates for treatment with PARP inhibitors. Screening of leukemia-derived cell lines revealed that lymphoid lineage-derived leukemia cell lines, except for those derived from mature B cells and KMT2A (MLL)-rearranged B-cell precursors, were relatively sensitive to PARP inhibitors. By contrast, acute myelogenous leukemia cell lines, except for RUNX1-RUNXT1 (AML1-ETO)-positive lines, were relatively resistant. Intriguingly, TCF3 (E2A)-HLF-positive leukemia was sensitive to PARP inhibitors. TCF3-HLF expression suppressed HRR activity, suggesting that PARP inhibitor treatment induced synthetic lethality. Furthermore, TCF3-HLF expression decreased levels of MCPH1, which regulates the expression of BRCA1, resulting in attenuation of HRR activity. The PARP inhibitor olaparib was also effective in an in vivo xenograft model. Our results suggest a novel therapeutic approach for treating refractory leukemia, particularly the TCF3-HLF-positive subtype.
Collapse
Affiliation(s)
- Jinhua Piao
- Department of Pediatrics and Developmental Biology, Graduate School of Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Shiori Takai
- Department of Pediatrics and Developmental Biology, Graduate School of Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Takahiro Kamiya
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, 119228, Singapore
| | - Takeshi Inukai
- Department of Pediatrics, Graduate School of Medicine, Yamanashi University, Yamanashi Chuo, 1110 Shimokato, Yamanashi, 409-3898, Japan
| | - Kanji Sugita
- Department of Pediatrics, Graduate School of Medicine, Yamanashi University, Yamanashi Chuo, 1110 Shimokato, Yamanashi, 409-3898, Japan
| | - Kazuma Ohyashiki
- Department of Hematology, Tokyo Medical University, Nishi-Shinjuku 6-7-1, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Domenico Delia
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Experimental Oncology, Via G. Venezian 1, Milan, 20133, Italy
| | - Mitsuko Masutani
- Department of Frontier Life Science, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Shuki Mizutani
- Department of Pediatrics and Developmental Biology, Graduate School of Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8519, Japan.
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Graduate School of Medicine, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
33
|
Wang YQ, Wang PY, Wang YT, Yang GF, Zhang A, Miao ZH. An Update on Poly(ADP-ribose)polymerase-1 (PARP-1) Inhibitors: Opportunities and Challenges in Cancer Therapy. J Med Chem 2016; 59:9575-9598. [PMID: 27416328 DOI: 10.1021/acs.jmedchem.6b00055] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Poly(ADP-ribose)polymerase-1 (PARP-1) is a critical DNA repair enzyme in the base excision repair pathway. Inhibitors of this enzyme comprise a new type of anticancer drug that selectively kills cancer cells by targeting homologous recombination repair defects. Since 2010, important advances have been achieved in PARP-1 inhibitors. Specifically, the approval of olaparib in 2014 for the treatment of ovarian cancer with BRCA mutations validated PARP-1 as an anticancer target and established its clinical importance in cancer therapy. Here, we provide an update on PARP-1 inhibitors, focusing on breakthroughs in their clinical applications and investigations into relevant mechanisms of action, biomarkers, and drug resistance. We also provide an update on the design strategies and the structural types of PARP-1 inhibitors. Opportunities and challenges in PARP-1 inhibitors for cancer therapy will be discussed based on the above advances.
Collapse
Affiliation(s)
- Ying-Qing Wang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203, China
| | - Ping-Yuan Wang
- CAS Key Laboratory of Receptor Research, and Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China.,Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University , Wuhan 430079, China
| | - Yu-Ting Wang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203, China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University , Wuhan 430079, China
| | - Ao Zhang
- CAS Key Laboratory of Receptor Research, and Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China
| | - Ze-Hong Miao
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203, China
| |
Collapse
|
34
|
Crafton SM, Bixel K, Hays JL. PARP inhibition and gynecologic malignancies: A review of current literature and on-going trials. Gynecol Oncol 2016; 142:588-96. [PMID: 27168003 DOI: 10.1016/j.ygyno.2016.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/02/2016] [Accepted: 05/04/2016] [Indexed: 01/05/2023]
Abstract
The poly (ADP-ribose) polymerase (PARP) family of enzymes is important in several DNA repair pathways. Drugs that inhibit these enzymes have been investigated in many types of cancer, but their application in the treatment of gynecologic malignancies has rapidly evolved - as manifested by the 2014 FDA approval for olaparib in the treatment of recurrent ovarian cancer associated with a germline BRCA mutation (gBRCA). In efforts to broaden their efficacy, current clinical trials have demonstrated benefit of olaparib, and other PARP inhibitors (PARPi), as single agents and in combination with cytotoxic chemotherapy and biologic agents, in wide ranging populations. Although the majority of data for PARPi in gynecologic malignancies has been specifically regarding ovarian cancer, their role in the treatment of uterine and cervical cancer is currently being investigated. This review will serve as a synopsis of seminal trials to date, summarize the breadth of clinical application in on-going studies, query how these results may change future practice, and reflect on questions yet to be answered.
Collapse
Affiliation(s)
- S M Crafton
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, United States.
| | - K Bixel
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, United States
| | - J L Hays
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, United States; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
35
|
LoRusso PM, Li J, Burger A, Heilbrun LK, Sausville EA, Boerner SA, Smith D, Pilat MJ, Zhang J, Tolaney SM, Cleary JM, Chen AP, Rubinstein L, Boerner JL, Bowditch A, Cai D, Bell T, Wolanski A, Marrero AM, Zhang Y, Ji J, Ferry-Galow K, Kinders RJ, Parchment RE, Shapiro GI. Phase I Safety, Pharmacokinetic, and Pharmacodynamic Study of the Poly(ADP-ribose) Polymerase (PARP) Inhibitor Veliparib (ABT-888) in Combination with Irinotecan in Patients with Advanced Solid Tumors. Clin Cancer Res 2016; 22:3227-37. [PMID: 26842236 PMCID: PMC4930710 DOI: 10.1158/1078-0432.ccr-15-0652] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 01/26/2016] [Indexed: 01/05/2023]
Abstract
PURPOSE PARP is essential for recognition and repair of DNA damage. In preclinical models, PARP inhibitors modulate topoisomerase I inhibitor-mediated DNA damage. This phase I study determined the MTD, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics (PD) of veliparib, an orally bioavailable PARP1/2 inhibitor, in combination with irinotecan. EXPERIMENTAL DESIGN Patients with advanced solid tumors were treated with 100 mg/m(2) irinotecan on days 1 and 8 of a 21-day cycle. Twice-daily oral dosing of veliparib (10-50 mg) occurred on days 3 to 14 (cycle 1) and days -1 to 14 (subsequent cycles) followed by a 6-day rest. PK studies were conducted with both agents alone and in combination. Paired tumor biopsies were obtained after irinotecan alone and veliparib/irinotecan to evaluate PARP1/2 inhibition and explore DNA damage signals (nuclear γ-H2AX and pNBS1). RESULTS Thirty-five patients were treated. DLTs included fatigue, diarrhea, febrile neutropenia, and neutropenia. The MTD was 100 mg/m(2) irinotecan (days 1 and 8) combined with veliparib 40 mg twice daily (days -1-14) on a 21-day cycle. Of 31 response-evaluable patients, there were six (19%) partial responses. Veliparib exhibited linear PK, and there were no apparent PK interactions between veliparib and irinotecan. At all dose levels, veliparib reduced tumor poly(ADP-ribose) (PAR) content in the presence of irinotecan. Several samples showed increases in γ-H2AX and pNBS1 after veliparib/irinotecan compared with irinotecan alone. CONCLUSIONS Veliparib can be safely combined with irinotecan at doses that inhibit PARP catalytic activity. Preliminary antitumor activity justifies further evaluation of the combination. Clin Cancer Res; 22(13); 3227-37. ©2016 AACR.
Collapse
Affiliation(s)
- Patricia M LoRusso
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Jing Li
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Angelika Burger
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Lance K Heilbrun
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Scott A Boerner
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Daryn Smith
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Mary Jo Pilat
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan. Wayne State University, Detroit, Michigan
| | - Jie Zhang
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Sara M Tolaney
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - James M Cleary
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Lawrence Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Julie L Boerner
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Adam Bowditch
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Dongpo Cai
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Tracy Bell
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Andrew Wolanski
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Allison M Marrero
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Yiping Zhang
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jiuping Ji
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Katherine Ferry-Galow
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robert J Kinders
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ralph E Parchment
- Clinical Pharmacodynamics Biomarker Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Geoffrey I Shapiro
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
36
|
Chung C, Lee R. An update on current and emerging therapies for epithelial ovarian cancer: Focus on poly(adenosine diphosphate-ribose) polymerase inhibition and antiangiogenesis. J Oncol Pharm Pract 2016; 23:454-469. [DOI: 10.1177/1078155216657165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epithelial ovarian cancer is the leading cause of death from gynecologic tumors in western countries. Newly diagnosed epithelial ovarian cancer patients usually have good initial response to combination of platinum-based and taxane-based chemotherapy. However, most patients eventually experience relapses, and responses to subsequent therapies are generally short-lived. Intraperitoneal chemotherapy has been shown to improve survival outcomes, but toxicities and logistics limit its acceptance. Dose-dense schedule of paclitaxel combined with carboplatin remains controversial, and more studies are needed to validate this approach. About 15% of epithelial ovarian cancer patients carry gene mutations in BRCA1 and/or BRCA2. The development of poly(adenosine diphosphate-ribose) polymerase inhibitors represents a novel therapeutic strategy, in which poly(adenosine diphosphate-ribose) inhibition leads to the formation of double-stranded DNA breaks that cannot be accurately repaired in BRCA1- or BRCA2-mutated tumors, thus leading to tumor cell death. This principle of synthetic lethality can be demonstrated by olaparib, an oral agent that inhibits the repair of single strand DNA breaks during DNA replication, causing defective homologous recombination and hence tumor cell death. Currently, many poly(adenosine diphosphate-ribose) inhibitors are in different phases of development. Furthermore, mechanisms of defective homologous recombination pathway may include other genetic and epigenetic abnormalities in addition to either germline or somatic BRCA1 and/or BRCA2 mutations, making these pathways as potential therapeutic targets. The clinical pharmacology, clinical efficacy, safety, administration issues of olaparib and current clinical development of poly(adenosine diphosphate-ribose) inhibitors are described in this article, along with an overview on the treatment options (including intraperitoneal chemotherapy and dose-dense chemotherapy) for epithelial ovarian cancer. On the other hand, overexpression of the vascular endothelial growth factor and increased angiogenesis are associated with the development and progression of epithelial ovarian cancer. Although there are some expected toxicities associated with antiangiogenesis, combination of bevacizumab and systemic chemotherapy improves the progression-free survival and response rate compared to chemotherapy alone. The clinical efficacy of adding bevacizumab and its safety for advanced epithelial ovarian cancer is also reviewed, with emerging data on antiangiogenesis therapy.
Collapse
Affiliation(s)
- Clement Chung
- Hematology/Oncology Clinical Pharmacist Specialist, Lyndon B Johnson General Hospital, Houston, USA
| | - Rosetta Lee
- Hematology/Oncology Clinical Pharmacist Specialist, Smith Clinic, Houston, USA
| |
Collapse
|
37
|
Elmongy EI, Khedr MA, Taleb NA, Awad HM, Abbas SES. Design, Synthesis, and Biological Evaluation of Some Cyclohepta[b]Thiophene and Substituted Pentahydrocycloheptathieno[2,3-d]Pyrimidine Derivatives. J Heterocycl Chem 2016. [DOI: 10.1002/jhet.2678] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Elshaymaa I. Elmongy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy; Helwan University; Ain Helwan, P.O. Box 11795 Cairo Egypt
| | - Mohammed A. Khedr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy; Helwan University; Ain Helwan, P.O. Box 11795 Cairo Egypt
| | - Nageh A. Taleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy; Helwan University; Ain Helwan, P.O. Box 11795 Cairo Egypt
| | - Hanem M. Awad
- Department of Tanning Materials and Leather Technology; National Research Centre; P.O. Box 12622 Dokki, Cairo Egypt
| | - Safinaz E.-S. Abbas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy; Cairo University; Kasr-El-Aini Street, P.O. Box 11562, Cairo Egypt
| |
Collapse
|
38
|
Chen EX, Jonker DJ, Siu LL, McKeever K, Keller D, Wells J, Hagerman L, Seymour L. A Phase I study of olaparib and irinotecan in patients with colorectal cancer: Canadian Cancer Trials Group IND 187. Invest New Drugs 2016; 34:450-7. [PMID: 27075016 DOI: 10.1007/s10637-016-0351-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 04/10/2016] [Indexed: 12/13/2022]
Abstract
Background Olaparib is an orally available inhibitor of PARP-1. In pre-clinical studies, olaparib was shown to potentiate anti-tumor effects of irinotecan in colon cancer cell lines. This phase I study was conducted to evaluate the safety and tolerability of olaparib in combination with irinotecan. Patients and Methods Patients with advanced colorectal cancer whose disease progressed after at least one systemic therapy regimen were enrolled. Dose escalation and de-escalation were based on toxicity assessment. Pharmacokinetic samples were collected in Cycle 1 for olaparib, irinotecan and SN-38. Results Twenty-five patients were enrolled, 11 patients on a schedule of continuous olaparib and irinotecan every 3 weeks (Part A) and 14 patients on a schedule of intermittent olaparib and irinotecan every 2 weeks (Part B). Continuous olaparib administration was associated with higher than expected toxicities and was not considered to be tolerable. Intermittent olaparib administration was better tolerated, and the recommended phase 2 doses were olaparib 50 mg p.o twice daily days 1-5 and irinotecan 125 mg/m(2) i.v. every 2 weeks. Common toxicities included fatigue, anorexia, diarrhea, nausea, vomiting, neutropenia, thrombocytopenia and abdominal pain. Nine patients had stable disease as the best response, 2 from Part A (3 and 9 months respectively), and 7 from Part B (median duration: 7.4 months; range: 4 to 13 months). There was no pharmacokinetic interaction between olaparib and irinotecan. Conclusions Olaparib can be combined with irinotecan if administered intermittently. Both olaparib and irinotecan required significant dose reductions. The lack of anti-tumor efficacy observed in this trial makes this combination of little interest for further clinical development. Trial Registration ID NCT00535353.
Collapse
Affiliation(s)
- Eric X Chen
- Princess Margaret Cancer Centre, University Health Network, Room 5-719, 610 University Ave, Toronto, ON, Canada, M5G 2 M9.
| | | | - Lillian L Siu
- Princess Margaret Cancer Centre, University Health Network, Room 5-719, 610 University Ave, Toronto, ON, Canada, M5G 2 M9
| | - Karyn McKeever
- Princess Margaret Cancer Centre, University Health Network, Room 5-719, 610 University Ave, Toronto, ON, Canada, M5G 2 M9
| | | | - Julie Wells
- Ottawa Health Research Institute, Ottawa, ON, Canada
| | | | | |
Collapse
|
39
|
Xie Z, Chen Y, Xu P, Zhou Y, Zhao Q, Jiao H, Li Z. Design, synthesis and bioevaluation of 1H-indole-4-carboxamide derivatives as potent poly(ADP-ribose) polymerase-1 inhibitors. RSC Adv 2016. [DOI: 10.1039/c6ra12591c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
LX15 is more potent than AG014699 in PARP-1 inhibitory activity and BRCA-1 deficient cell inhibitory activity. It is more effective than AG014699 in potentiating the antitumor activity of TMZin vitro and in vivo.
Collapse
Affiliation(s)
- Zhouling Xie
- Jiangsu Key Laboratory of Drug Design and Optimization
- China Pharmaceutical University
- Nanjing 21009
- China
| | - Yu Chen
- Jiangsu Key Laboratory of Drug Design and Optimization
- China Pharmaceutical University
- Nanjing 21009
- China
| | - Pengfei Xu
- Jiangsu Key Laboratory of Drug Design and Optimization
- China Pharmaceutical University
- Nanjing 21009
- China
| | - Youli Zhou
- Jiangsu Key Laboratory of Drug Design and Optimization
- China Pharmaceutical University
- Nanjing 21009
- China
| | - Qian Zhao
- Jiangsu Key Laboratory of Drug Design and Optimization
- China Pharmaceutical University
- Nanjing 21009
- China
| | - He Jiao
- Jiangsu Key Laboratory of Drug Design and Optimization
- China Pharmaceutical University
- Nanjing 21009
- China
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization
- China Pharmaceutical University
- Nanjing 21009
- China
| |
Collapse
|
40
|
Bang YJ, Im SA, Lee KW, Cho JY, Song EK, Lee KH, Kim YH, Park JO, Chun HG, Zang DY, Fielding A, Rowbottom J, Hodgson D, O'Connor MJ, Yin X, Kim WH. Randomized, Double-Blind Phase II Trial With Prospective Classification by ATM Protein Level to Evaluate the Efficacy and Tolerability of Olaparib Plus Paclitaxel in Patients With Recurrent or Metastatic Gastric Cancer. J Clin Oncol 2015; 33:3858-3865. [PMID: 26282658 DOI: 10.1200/jco.2014.60.0320] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Gastric cancer cell lines, particularly those with low levels of ataxia telangiectasia mutated (ATM), a key activator of DNA damage response, are sensitive to the poly (ADP-ribose) polymerase inhibitor olaparib. We compared the efficacy of olaparib plus paclitaxel (olaparib/paclitaxel) with paclitaxel alone in patients with recurrent or metastatic gastric cancer and assessed whether low ATM expression is predictive of improved clinical outcome for olaparib/paclitaxel. PATIENTS AND METHODS In this phase II, double-blind study (Study 39; NCT01063517), patients were randomly assigned to oral olaparib 100 mg twice per day (tablets) plus paclitaxel (80 mg/m(2) per day intravenously on days 1, 8, and 15 of every 28-day cycle) or placebo plus paclitaxel (placebo/paclitaxel), followed by maintenance monotherapy with olaparib (200 mg twice per day) or placebo. The study population was enriched to 50% for patients with low or undetectable ATM levels (ATMlow). Primary end point was progression-free survival (PFS). RESULTS One hundred twenty-three of 124 randomly assigned patients received treatment (olaparib/paclitaxel, n = 61; placebo/paclitaxel, n = 62). The screening prevalence of ATMlow patients was 14%. Olaparib/paclitaxel did not lead to a significant improvement in PFS versus placebo/paclitaxel (overall population: hazard ratio [HR], 0.80; median PFS, 3.91 v 3.55 months, respectively; ATMlow population: HR, 0.74; median PFS, 5.29 v 3.68 months, respectively). However, olaparib/paclitaxel significantly improved overall survival (OS) versus placebo/paclitaxel in both the overall population (HR, 0.56; 80% CI, 0.41 to 0.75; P = .005; median OS, 13.1 v 8.3 months, respectively) and the ATMlow population (HR, 0.35; 80% CI, 0.22 to 0.56; P = .002; median OS, not reached v 8.2 months, respectively). Olaparib/paclitaxel was generally well tolerated, with no unexpected safety findings. CONCLUSION Olaparib/paclitaxel is active in the treatment of patients with metastatic gastric cancer, with a greater OS benefit in ATMlow patients. A phase III trial in this setting is under way.
Collapse
Affiliation(s)
- Yung-Jue Bang
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China.
| | - Seock-Ah Im
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Keun-Wook Lee
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Jae Yong Cho
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Eun-Kee Song
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Kyung Hee Lee
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Yeul Hong Kim
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Joon Oh Park
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Hoo Geun Chun
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Dae Young Zang
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Anitra Fielding
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Jacqui Rowbottom
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Darren Hodgson
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Mark J O'Connor
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Xiaolu Yin
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| | - Woo Ho Kim
- Yung-Jue Bang, Seock-Ah Im, and Woo Ho Kim, Seoul National University College of Medicine; Jae Yong Cho, Yonsei University College of Medicine, Gangnam Severance Hospital; Yeul Hong Kim, Anam Hospital, Korea University College of Medicine; Joon Oh Park, Samsung Medical Center, Sungkyunkwan University School of Medicine; Hoo Geun Chun, Seoul St Mary's Hospital, Catholic University of Korea, Seoul; Keun-Wook Lee, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam; Eun-Kee Song, Chonbuk National University Medical School, Jeonju; Kyung Hee Lee, Yeungnam University Hospital, Daegu; Dae Young Zang, Hallym University Sacred Heart Hospital, Anyang, Korea; Anitra Fielding, Jacqui Rowbottom, Darren Hodgson, and Mark J. O'Connor, AstraZeneca, Macclesfield, United Kingdom; and Xiaolu Yin, Innovation Centre, AstraZeneca, Shanghai, China
| |
Collapse
|
41
|
Zmuda F, Malviya G, Blair A, Boyd M, Chalmers AJ, Sutherland A, Pimlott SL. Synthesis and Evaluation of a Radioiodinated Tracer with Specificity for Poly(ADP-ribose) Polymerase-1 (PARP-1) in Vivo. J Med Chem 2015; 58:8683-93. [PMID: 26469301 DOI: 10.1021/acs.jmedchem.5b01324] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interest in nuclear imaging of poly(ADP-ribose) polymerase-1 (PARP-1) has grown in recent years due to the ability of PARP-1 to act as a biomarker for glioblastoma and increased clinical use of PARP-1 inhibitors. This study reports the identification of a lead iodinated analog 5 of the clinical PARP-1 inhibitor olaparib as a potential single-photon emission computed tomography (SPECT) imaging agent. Compound 5 was shown to be a potent PARP-1 inhibitor in cell-free and cellular assays, and it exhibited mouse plasma stability but approximately 3-fold greater intrinsic clearance when compared to olaparib. An (123)I-labeled version of 5 was generated using solid state halogen exchange methodology. Ex vivo biodistribution studies of [(123)I]5 in mice bearing subcutaneous glioblastoma xenografts revealed that the tracer had the ability to be retained in tumor tissue and bind to PARP-1 with specificity. These findings support further investigations of [(123)I]5 as a noninvasive PARP-1 SPECT imaging agent.
Collapse
Affiliation(s)
- Filip Zmuda
- WestCHEM, School of Chemistry, University of Glasgow , The Joseph Black Building, Glasgow G12 8QQ, U.K.,Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow , Glasgow G61 1QH, U.K
| | - Gaurav Malviya
- Nuclear Imaging, Cancer Research UK Beatson Institute , Glasgow G61 1BD, U.K
| | - Adele Blair
- WestCHEM, School of Chemistry, University of Glasgow , The Joseph Black Building, Glasgow G12 8QQ, U.K
| | - Marie Boyd
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , John Arbuthnott Building, Glasgow, G4 0RE, U.K
| | - Anthony J Chalmers
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow , Glasgow G61 1QH, U.K
| | - Andrew Sutherland
- WestCHEM, School of Chemistry, University of Glasgow , The Joseph Black Building, Glasgow G12 8QQ, U.K
| | - Sally L Pimlott
- West of Scotland PET Centre, Greater Glasgow and Clyde NHS Trust, Glasgow G12 0YN, U.K
| |
Collapse
|
42
|
Fujii N, Evison BJ, Actis ML, Inoue A. A novel assay revealed that ribonucleotide reductase is functionally important for interstrand DNA crosslink repair. Bioorg Med Chem 2015; 23:6912-21. [PMID: 26462050 DOI: 10.1016/j.bmc.2015.09.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/22/2015] [Accepted: 09/29/2015] [Indexed: 11/17/2022]
Abstract
Cells have evolved complex biochemical pathways for DNA interstrand crosslink (ICL) removal. Despite the chemotherapeutic importance of ICL repair, there have been few attempts to identify which mechanistic DNA repair inhibitor actually inhibits ICL repair. To identify such compounds, a new and robust ICL repair assay was developed using a novel plasmid that contains synthetic ICLs between a CMV promoter region that drives transcription and a luciferase reporter gene, and an SV40 origin of replication and the large T antigen (LgT) gene that enables self-replication in mammalian cells. In a screen against compounds that are classified as inhibitors of DNA repair or synthesis, the reporter generation was exquisitely sensitive to ribonucleotide reductase (RNR) inhibitors such as gemcitabine and clofarabine, but not to inhibitors of PARP, ATR, ATM, Chk1, and others. The effect was observed also by siRNA downregulation of RNR. Moreover, the reporter generation was also particularly sensitive to 3-AP, a non-nucleoside RNR inhibitor, but not significantly sensitive to DNA replication stressors, suggesting that the involvement of RNR in ICL repair is independent of incorporation of a nucleotide RNR inhibitor into DNA to induce replication stress. The reporter generation from a modified version of the plasmid that lacks the LgT-SV40ori motif was also adversely affected by RNR inhibitors, further indicating a role for RNR in ICL repair that is independent of DNA replication. Intriguingly, unhooking of cisplatin-ICL from nuclear DNA was significantly inhibited by low doses of gemcitabine, suggesting an unidentified functional role for RNR in the process of ICL unhooking. The assay approach could identify other molecules essential for ICLR in quantitative and flexible manner.
Collapse
Affiliation(s)
- Naoaki Fujii
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | - Benjamin J Evison
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Marcelo L Actis
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Akira Inoue
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
43
|
Livraghi L, Garber JE. PARP inhibitors in the management of breast cancer: current data and future prospects. BMC Med 2015; 13:188. [PMID: 26268938 PMCID: PMC4535298 DOI: 10.1186/s12916-015-0425-1] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/17/2015] [Indexed: 12/18/2022] Open
Abstract
Poly(ADP-ribose) polymerases (PARP) are enzymes involved in DNA-damage repair. Inhibition of PARPs is a promising strategy for targeting cancers with defective DNA-damage repair, including BRCA1 and BRCA2 mutation-associated breast and ovarian cancers. Several PARP inhibitors are currently in trials in the adjuvant, neoadjuvant, and metastatic settings for the treatment of ovarian, BRCA-mutated breast, and other cancers. We herein review the development of PARP inhibitors and the basis for the excitement surrounding these agents, their use as single agents and in combinations, as well as their toxicities, mechanisms of acquired resistance, and companion diagnostics.
Collapse
Affiliation(s)
- Luca Livraghi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
44
|
Bixel K, Hays JL. Olaparib in the management of ovarian cancer. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2015; 8:127-35. [PMID: 26309417 PMCID: PMC4538690 DOI: 10.2147/pgpm.s62809] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Alterations in the homologous repair pathway are thought to occur in 30%–50% of epithelial ovarian cancers. Cells deficient in homologous recombination rely on alternative pathways for DNA repair in order to survive, thereby providing a potential target for therapy. Olaparib, a poly(ADP-ribose) polymerase (PARP) inhibitor, capitalizes on this concept and is the first drug in its class approved for patients with ovarian cancer. This review article will provide an overview of the BRCA genes and homologous recombination, the role of PARP in DNA repair and the biological rationale for the use of PARP inhibitors as cancer therapy, and ultimately will focus on the use of olaparib in the management of ovarian cancer.
Collapse
Affiliation(s)
- Kristin Bixel
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Ohio State University, Columbus, OH, USA
| | - John L Hays
- Department of Hematology Oncology, Ohio State University, Columbus, OH, USA
| |
Collapse
|
45
|
van der Noll R, Marchetti S, Steeghs N, Beijnen JH, Mergui-Roelvink MWJ, Harms E, Rehorst H, Sonke GS, Schellens JHM. Long-term safety and anti-tumour activity of olaparib monotherapy after combination with carboplatin and paclitaxel in patients with advanced breast, ovarian or fallopian tube cancer. Br J Cancer 2015; 113:396-402. [PMID: 26180927 PMCID: PMC4522644 DOI: 10.1038/bjc.2015.256] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/26/2015] [Accepted: 06/14/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Olaparib (AZD2281), a PARP-1/2 inhibitor, has been extensively investigated in clinical trials. However, limited clinical data are available about its long-term safety and anti-tumour activity. METHODS Patients had first participated in a phase I study of olaparib combined with carboplatin and/or paclitaxel. They continued with olaparib monotherapy in their best interest if they failed to tolerate the combination due to the treatment-related adverse events (TRAEs). Safety data were collected by physical examination and regular laboratory evaluations. Disease evaluations were performed by CT scan. RESULTS At data cutoff, 21 patients were included; 10 with breast, 9 with ovarian and 2 with fallopian tube cancer of whom 16 patients had a BRCA mutation (13 BRCA1; 3 BRCA2). TRAEs were mostly haematological and most prominent shortly after switching from combination to monotherapy, probably due to carry-over effects of chemotherapy. Over time, both severity and frequency of TRAEs decreased. Responses to olaparib were durable with a median treatment duration of 52 (range 7-183) weeks. In total, nine (43%) patients were still on study at data cutoff. CONCLUSION Continued long-term daily olaparib was found to be safe and tolerable. Encouragingly, patients who showed a favourable response on earlier combination therapy maintained this response on olaparib monotherapy.
Collapse
Affiliation(s)
- Ruud van der Noll
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Serena Marchetti
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Neeltje Steeghs
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Jos H Beijnen
- 1] Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands [2] Utrecht Institute of Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
| | - Marja W J Mergui-Roelvink
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Emmy Harms
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Harriet Rehorst
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Jan H M Schellens
- 1] Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands [2] Utrecht Institute of Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
| |
Collapse
|
46
|
Tewari KS, Eskander RN, Monk BJ. Development of Olaparib for BRCA-Deficient Recurrent Epithelial Ovarian Cancer. Clin Cancer Res 2015; 21:3829-35. [PMID: 26169965 DOI: 10.1158/1078-0432.ccr-15-0088] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/22/2015] [Indexed: 11/16/2022]
Abstract
The FDA approval of the PARP inhibitor olaparib for fourth-line therapy of germline BRCA1/2-mutated ovarian cancer represents the first registered indication for this class of drugs in any disease. PARP is a family of proteins involved in the repair of single-strand DNA breaks. High-grade serous ovarian carcinomas with BRCA deficiencies may be particularly vulnerable to both direct and indirect effects of PARP inhibition. This phenotype frequently arises as a consequence of defects in the repair of damaged DNA, rendering cancer cells susceptible to DNA-damaging platinum compounds and targeted therapies affecting homologous recombination repair (HRR). When cells already deficient in HRR are exposed to PARP inhibitors, apoptosis occurs by way of synthetic lethality. In this review, we trace the clinical development of olaparib for women with recurrent epithelial ovarian carcinoma harboring germline BRCA mutations, a biomarker for HRR deficiency present in 15% to 20% of cases. Clinical trials highlighted include not only those pivotal studies that have led to regulatory approval in the United States and in Europe, but also those in which olaparib was studied in novel combinations, including chemotherapy and antiangiogenesis agents.
Collapse
Affiliation(s)
- Krishnansu S Tewari
- The Division of Gynecologic Oncology, University of California, Irvine Medical Center, Orange, California
| | - Ramez N Eskander
- The Division of Gynecologic Oncology, University of California, Irvine Medical Center, Orange, California
| | - Bradley J Monk
- The Division of Gynecologic Oncology, University of Arizona Cancer Center at Dignity Health St. Joseph's Hospital and Medical Center, Phoenix, Arizona.
| |
Collapse
|
47
|
Sistigu A, Manic G, Obrist F, Vitale I. Trial watch - inhibiting PARP enzymes for anticancer therapy. Mol Cell Oncol 2015; 3:e1053594. [PMID: 27308587 DOI: 10.1080/23723556.2015.1053594] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/16/2015] [Accepted: 05/18/2015] [Indexed: 12/25/2022]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are a members of family of enzymes that catalyze poly(ADP-ribosyl)ation (PARylation) and/or mono(ADP-ribosyl)ation (MARylation), two post-translational protein modifications involved in crucial cellular processes including (but not limited to) the DNA damage response (DDR). PARP1, the most abundant family member, is a nuclear protein that is activated upon sensing distinct types of DNA damage and contributes to their resolution by PARylating multiple DDR players. Recent evidence suggests that, along with DDR, activated PARP1 mediates a series of prosurvival and proapoptotic processes aimed at preserving genomic stability. Despite this potential oncosuppressive role, upregulation and/or overactivation of PARP1 or other PARP enzymes has been reported in a variety of human neoplasms. Over the last few decades, several pharmacologic inhibitors of PARP1 and PARP2 have been assessed in preclinical and clinical studies showing potent antineoplastic activity, particularly against homologous recombination (HR)-deficient ovarian and breast cancers. In this Trial Watch, we describe the impact of PARP enzymes and PARylation in cancer, discuss the mechanism of cancer cell killing by PARP1 inactivation, and summarize the results of recent clinical studies aimed at evaluating the safety and therapeutic profile of PARP inhibitors in cancer patients.
Collapse
Affiliation(s)
| | - Gwenola Manic
- Regina Elena National Cancer Institute , Rome, Italy
| | - Florine Obrist
- Université Paris-Sud/Paris XI, Le Kremlin-Bicêtre, France; INSERM, UMRS1138, Paris, France; Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, Rome, Italy; Department of Biology, University of Rome "TorVergata", Rome, Italy
| |
Collapse
|
48
|
Lheureux S, Bowering V, Karakasis K, Oza AM. Safety evaluation of olaparib for treating ovarian cancer. Expert Opin Drug Saf 2015; 14:1305-16. [DOI: 10.1517/14740338.2015.1045875] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
49
|
Gómez-Martín A, Altakroni B, Lozano-Paniagua D, Margison GP, de Vocht F, Povey AC, Hernández AF. Increased N7-methyldeoxyguanosine DNA adducts after occupational exposure to pesticides and influence of genetic polymorphisms of paraoxonase-1 and glutathione S-transferase M1 and T1. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:437-445. [PMID: 25427726 DOI: 10.1002/em.21929] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 10/17/2014] [Accepted: 11/07/2014] [Indexed: 06/04/2023]
Abstract
There are concerns about genetic risks associated with long-term exposure to pesticides as these compounds may damage DNA, resulting in mutations that eventually lead to cancer, neurological, and reproductive adverse health effects. This study assessed DNA damage in intensive agricultural workers exposed to pesticides by determining the levels of N7-methyldeoxyguanosine (N7-MedG), an adduct known to be a robust biomarker of recent exposure to chemical methylating agents. A cohort of 39 plastic greenhouse workers was assessed for changes in lymphocyte DNA N7-MedG levels between low level and high level exposures during the course of a spraying season. The contributions of genetic polymorphisms of the pesticide-metabolizing enzymes paraoxonase-1 (PON1) and the glutathione S-transferases, GSTM1 and GSTT1, on N7-MedG levels and other potential confounders were also assessed. N7-MedG increased in the period of high pesticide exposure as compared to the low exposure period (0.23 and 0.18 µmol N7-MedG/mol dG for the unadjusted and adjusted linear mixed models, P = 0.02 and 0.08, respectively). Significant decreased levels of erythrocyte acetylcholinesterase and plasma cholinesterase were observed in the high versus low exposure period in both the unadjusted (2.85 U/g hemoglobin and 213.13 U/L, respectively) and adjusted linear mixed models (2.99 U/g hemoglobin and 230.77 U/L, respectively), indicating pesticide intake. In intensive agriculture workers, higher pesticide exposure increased DNA alkylation levels, further demonstrating the genotoxicity of pesticides in man. In addition, pesticide-exposed individuals with inherited susceptible metabolic genotypes (particularly, null genotype for GSTM1 and the PON1 192R allele) appear to have an increased risk of genotoxic DNA damage. Environ. Mol. Mutagen. 56:437-445, 2015. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Antonio Gómez-Martín
- Department of Legal Medicine and Toxicology, University of Granada School of Medicine, Granada, Spain
| | - Bashar Altakroni
- Centre for Occupational and Environmental Health (COEH), Centre for Epidemiology, Institute of Population Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - David Lozano-Paniagua
- Department of Legal Medicine and Toxicology, University of Granada School of Medicine, Granada, Spain
| | - Geoffrey P Margison
- Centre for Occupational and Environmental Health (COEH), Centre for Epidemiology, Institute of Population Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Frank de Vocht
- School of Social and Community Medicine, University of Bristol, Bristol, BS8 2PS, United Kingdom
| | - Andrew C Povey
- Centre for Occupational and Environmental Health (COEH), Centre for Epidemiology, Institute of Population Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Antonio F Hernández
- Department of Legal Medicine and Toxicology, University of Granada School of Medicine, Granada, Spain
| |
Collapse
|
50
|
Extent of radiosensitization by the PARP inhibitor olaparib depends on its dose, the radiation dose and the integrity of the homologous recombination pathway of tumor cells. Radiother Oncol 2015; 116:358-65. [PMID: 25981132 DOI: 10.1016/j.radonc.2015.03.028] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/19/2015] [Accepted: 03/26/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND PURPOSE The PARP inhibitor olaparib is currently tested in clinical phase 1 trials to define safe dose levels in combination with RT. However, certain clinically relevant insights are still lacking. Here we test, while comparing to single agent activity, the olaparib dose and genetic background dependence of olaparib-mediated radiosensitization. MATERIALS AND METHODS Long-term growth inhibition and clonogenic assays were used to assess radiosensitization in BRCA2-deficient and BRCA2-complemented cells and in a panel of human head and neck squamous cell carcinoma cell lines. RESULTS The extent of radiosensitization greatly depended on the olaparib dose, the radiation dose and the homologous recombination status of cells. Olaparib concentrations that resulted in radiosensitization prevented PAR induction by irradiation. Seven hours olaparib exposures were sufficient for radiosensitization. Importantly, the radiosensitizing effects can be observed at much lower olaparib doses than the single agent effects. CONCLUSION Extrapolation of these data to the clinic suggests that low olaparib doses are sufficient to cause radiosensitization, underlining the potential of the treatment. Here we show that drug doses achieving radiosensitization can greatly differ from those achieving single agent activities, an important consideration when developing combined radiotherapy strategies with novel targeted agents.
Collapse
|