501
|
Baloch T, López-Ozuna VM, Wang Q, Matanis E, Kessous R, Kogan L, Yasmeen A, Gotlieb WH. Sequential therapeutic targeting of ovarian Cancer harboring dysfunctional BRCA1. BMC Cancer 2019; 19:44. [PMID: 30630446 PMCID: PMC6327434 DOI: 10.1186/s12885-018-5250-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022] Open
Abstract
Background Poly (ADP-ribose) polymerase inhibitors (PARPi) have become the first targeted therapies available in the treatment of patients with high-grade serous ovarian cancer (HGSOC). We recently described a significant reduction in PARP1 protein levels in vitro and in vivo in patients treated with standard carboplatinum-paclitaxel chemotherapy, raising the question whether the sequence of treatment used today with chemotherapy followed by PARPi is optimal. In this study, we aim to evaluate if the sequence of PARPi followed by chemotherapy could be more beneficial. Methods BRCA1-mutated (UWB1.287, SNU-251), epigenetically-silenced (OVCAR8), and wild-type (SKOV3, A2780PAR & A2780CR) ovarian cancer cell lines were exposed to clinically relevant doses of PARPi followed by different doses of standard chemotherapy and compared to the inverse treatment. The therapeutic efficacy was assessed using colony formation assays. Flow cytometry was used to evaluate cell apoptosis rate and the changes in cell cycle. Finally, apoptotic and cell cycle protein expression was immunodetected using western blot. Results Exposure to PARPi prior to standard chemotherapy sensitized BRCA1-mutated or epigenetically-silenced BRCA1 cell lines to lower doses of chemotherapy. Similar results were observed in BRCA1 wild-type and cell lines in which BRCA1 functionality was restored. Moreover, this treatment increased the apoptotic rate in these cell lines. Conclusion Pre-treatment with PARPi followed by standard chemotherapy in vitro is more efficient in growth inhibition and induction of apoptosis compared to the administration of standard chemotherapy followed by PARPi. Electronic supplementary material The online version of this article (10.1186/s12885-018-5250-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tahira Baloch
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada.,Department of Experimental Surgery, McGill University, Montreal, QC, Canada
| | - Vanessa M López-Ozuna
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada
| | - Qiong Wang
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada
| | - Emad Matanis
- Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada
| | - Roy Kessous
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, 3755 Cote Ste. Catherine Road, Montreal, QC, H3T 1E2, Canada.,Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada
| | - Liron Kogan
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, 3755 Cote Ste. Catherine Road, Montreal, QC, H3T 1E2, Canada.,Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada
| | - Amber Yasmeen
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, 3755 Cote Ste. Catherine Road, Montreal, QC, H3T 1E2, Canada. .,Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada.
| | - Walter H Gotlieb
- Division of Gynecologic Oncology, Jewish General Hospital, McGill University, 3755 Cote Ste. Catherine Road, Montreal, QC, H3T 1E2, Canada.,Segal Cancer Center, Lady Davis Institute of Medical Research, McGill University, Montreal, QC, Canada.,Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
502
|
Shapiro GI, Kristeleit RS, Burris HA, LoRusso P, Patel MR, Drew Y, Giordano H, Maloney L, Watkins S, Goble S, Jaw‐Tsai S, Xiao JJ. Pharmacokinetic Study of Rucaparib in Patients With Advanced Solid Tumors. Clin Pharmacol Drug Dev 2019; 8:107-118. [PMID: 29799676 PMCID: PMC6585632 DOI: 10.1002/cpdd.575] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/15/2018] [Indexed: 01/14/2023]
Abstract
The phase 1-2 study CO-338-010 (Study 10; NCT01482715) is evaluating single-agent rucaparib, a poly(ADP-ribose) polymerase inhibitor, administered orally to patients with an advanced solid tumor. In the dose escalation phase (Part 1), we characterized the single-dose and steady-state pharmacokinetic profiles of rucaparib administered once daily (QD; dose range, 40-500 mg; n = 16) or twice daily (BID; dose range, 240-840 mg; n = 30). Across all dosing schedules examined, the plasma exposure of rucaparib was approximately dose proportional; half-life was approximately 17 hours, and median time to maximum concentration (tmax ) ranged from 1.5 to 6.0 hours after a single dose and 1.5 to 4.0 hours following repeated dosing. The steady-state accumulation ratio ranged from 1.60 to 2.33 following QD dosing and 1.47 to 5.44 following BID dosing. No effect of food on rucaparib pharmacokinetics was observed with a single dose of 40 mg (n = 3) or 300 mg (n = 6). In a phase 2 portion of the study (Part 3), the pharmacokinetic profile of rucaparib was further evaluated at the recommended phase 2 dose of 600 mg BID (n = 26). The mean (coefficient of variation) steady-state maximum concentration (Cmax ) and area under the concentration-time curve from time zero to 12 hours (AUC0-12h ) were 1940 ng/mL (54%) and 16 900 ng ⋅ h/mL (54%), respectively. A high-fat meal moderately increased rucaparib exposure. The fed-to-fasted geometric mean ratios (90% confidence interval [CI]) for AUC0-24h and Cmax were 138% (117%-162%) and 120% (99.1%-146%); the median (90%CI) tmax delay was 2.5 (0.5-4.4) hours.
Collapse
Affiliation(s)
| | | | - Howard A. Burris
- Sarah Cannon Research Institute at Tennessee OncologyNashvilleTNUSA
| | | | - Manish R. Patel
- Sarah Cannon Research Institute at Florida Cancer SpecialistsSarasotaFLUSA
| | - Yvette Drew
- Northern Centre for Cancer CareNewcastle upon TyneUnited Kingdom
| | | | | | | | | | | | | |
Collapse
|
503
|
Sun K, Mikule K, Wang Z, Poon G, Vaidyanathan A, Smith G, Zhang ZY, Hanke J, Ramaswamy S, Wang J. A comparative pharmacokinetic study of PARP inhibitors demonstrates favorable properties for niraparib efficacy in preclinical tumor models. Oncotarget 2018; 9:37080-37096. [PMID: 30647846 PMCID: PMC6324689 DOI: 10.18632/oncotarget.26354] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/28/2018] [Indexed: 01/19/2023] Open
Abstract
Niraparib is an orally bioavailable and selective poly (ADP-ribose) polymerase (PARP)-1/-2 inhibitor approved for maintenance treatment of both BRCA mutant (mut) and BRCA wildtype (wt) adult patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancers who have demonstrated a complete or partial response to platinum-based chemotherapy. In patients without germline BRCA mutations (non-gBRCAmut), niraparib improved progression-free survival (PFS) by 5.4 months, whereas another PARP inhibitor (PARPi) olaparib supplied only 1.9 months of improvement in a similar patient population. Previous studies revealed higher cell membrane permeability and volume of distribution (VD) as unique features of niraparib in comparison to other PARPi including olaparib. Here, we explore the potential correlation of these pharmacokinetic properties to preclinical antitumor effects in BRCAwt tumors. Our results show that at steady state, tumor exposure to niraparib is 3.3 times greater than plasma exposure in tumor xenograft mouse models. In comparison, the tumor exposure to olaparib is less than observed in plasma. In addition, niraparib crosses the blood-brain barrier and shows good sustainability in the brain, whereas sustained brain exposure to olaparib is not observed in the same models. Consistent with its favorable tumor and brain distribution, niraparib achieves more potent tumor growth inhibition than olaparib in BRCAwt models and an intracranial tumor model at maximum tolerated doses (MTD). These findings demonstrate favorable pharmacokinetic profiles and potent antitumor effects of niraparib in BRCAwt tumors, consistent with its broader clinical effect in patients with both BRCAmut and BRCAwt tumors.
Collapse
Affiliation(s)
| | | | | | | | - Aparajitha Vaidyanathan
- Division of Cellular Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital & Medical School, Dundee, UK
| | - Gillian Smith
- Division of Cellular Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital & Medical School, Dundee, UK
| | | | | | | | | |
Collapse
|
504
|
Ding L, Kim HJ, Wang Q, Kearns M, Jiang T, Ohlson CE, Li BB, Xie S, Liu JF, Stover EH, Howitt BE, Bronson RT, Lazo S, Roberts TM, Freeman GJ, Konstantinopoulos PA, Matulonis UA, Zhao JJ. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep 2018; 25:2972-2980.e5. [PMID: 30540933 PMCID: PMC6366450 DOI: 10.1016/j.celrep.2018.11.054] [Citation(s) in RCA: 355] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/02/2018] [Accepted: 11/13/2018] [Indexed: 12/19/2022] Open
Abstract
PARP inhibitors have shown promising clinical activities for patients with BRCA mutations and are changing the landscape of ovarian cancer treatment. However, the therapeutic mechanisms of action for PARP inhibition in the interaction of tumors with the tumor microenvironment and the host immune system remain unclear. We find that PARP inhibition by olaparib triggers robust local and systemic antitumor immunity involving both adaptive and innate immune responses through a STING-dependent antitumor immune response in mice bearing Brca1-deficient ovarian tumors. This effect is further augmented when olaparib is combined with PD-1 blockade. Our findings thus provide a molecular mechanism underlying antitumor activity by PARP inhibition and lay a foundation to improve therapeutic outcome for cancer patients.
Collapse
Affiliation(s)
- Liya Ding
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Hye-Jung Kim
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Qiwei Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Kearns
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Tao Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Carolynn E Ohlson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ben B Li
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Shaozhen Xie
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Joyce F Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Elizabeth H Stover
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Brooke E Howitt
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roderick T Bronson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Suzan Lazo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Thomas M Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Ursula A Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Jean J Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
505
|
Abstract
Introduction: Rucaparib is increasingly being utilized for women with recurrent ovarian cancer both as treatment and maintenance therapy. Poly-ADP ribose polymerase (PARP) inhibitors like rucaparib are daily oral medication that exploit the DNA repair pathway. The most significant clinical benefit is in those tumors exhibiting deficiency in their homologous recombination pathway. Areas covered: This review will discuss the mechanism of action, clinical efficacy data, and safety of rucaparib as presented from phase 1, 2, and 3 clinical trials. Expert opinion: Rucaparib is a promising therapeutic option for women where prolongation of survival with favorable side effects is the goal. The side effect profile of rucaparib is similar or more favorable when evaluating it against other PARP inhibitors. Physicians will likely need to have increasing comfort with unique side effects like transaminitis and serum creatinine increases. Clinicians should be prepared for not only increasing utilization of PARP inhibitors in the recurrent setting but also upfront usage may be on the horizon. As >50% of high grade serous ovarian cancers likely have the predictive biomarker, HRD, for PARP inhibition a substantial group of patients stand to have PARP inhibitors as part of their oncologic care.
Collapse
Affiliation(s)
- Casey M Cosgrove
- a Department of Obstetrics and Gynecology , The Ohio State University Wexner Medical Center , Columbus , Ohio, USA
| | - David M O'Malley
- a Department of Obstetrics and Gynecology , The Ohio State University Wexner Medical Center , Columbus , Ohio, USA
| |
Collapse
|
506
|
Tumiati M, Hietanen S, Kauppi L. Time to go functional! Determining tumors' DNA repair capacity ex vivo. Oncotarget 2018; 9:36826-36827. [PMID: 30627321 PMCID: PMC6305149 DOI: 10.18632/oncotarget.26419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 11/26/2018] [Indexed: 11/25/2022] Open
Affiliation(s)
- Manuela Tumiati
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sakari Hietanen
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Liisa Kauppi
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
507
|
Toma M, Witusik-Perkowska M, Szwed M, Stawski R, Szemraj J, Drzewiecka M, Nieborowska-Skorska M, Radek M, Kolasa P, Matlawska-Wasowska K, Sliwinski T, Skorski T. Eradication of LIG4-deficient glioblastoma cells by the combination of PARP inhibitor and alkylating agent. Oncotarget 2018; 9:36867-36877. [PMID: 30627327 PMCID: PMC6305145 DOI: 10.18632/oncotarget.26409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer cells often accumulate spontaneous and treatment-induced DNA damage i.e. potentially lethal DNA double strand breaks (DSBs). Targeting DSB repair mechanisms with specific inhibitors could potentially sensitize cancer cells to the toxic effect of DSBs. Current treatment for glioblastoma includes tumor resection followed by radiotherapy and/or temozolomide (TMZ) - an alkylating agent inducing DNA damage. We hypothesize that combination of PARP inhibitor (PARPi) with TMZ in glioblastoma cells displaying downregulation of DSB repair genes could trigger synthetic lethality. In our study, we observed that PARP inhibitor (BMN673) was able to specifically sensitize DNA ligase 4 (LIG4)-deprived glioblastoma cells to TMZ while normal astrocytes were not affected. LIG4 downregulation resulting in low effectiveness of DNA-PK-mediated non-homologous end-joining (D-NHEJ), which in combination with BMN673 and TMZ resulted in accumulation of lethal DSBs and specific eradication of glioblastoma cells. Restoration of the LIG4 expression caused loss of sensitivity to BMN673+TMZ. In conclusion, PARP inhibitor combined with DNA damage inducing agents can be utilized in patients with glioblastoma displaying defects in D-NHEJ.
Collapse
Affiliation(s)
- Monika Toma
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | | | - Marzena Szwed
- Department of Medical Biophysics, University of Lodz, Lodz, Poland
| | - Robert Stawski
- Department of Clinical Physiology, Medical University of Lodz, Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Malgorzata Drzewiecka
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Margaret Nieborowska-Skorska
- Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Maciej Radek
- Department of Neurosurgery, Surgery of Spine and Peripheral Nerves, Medical University of Lodz, University Hospital WAM-CSW, Lodz, Poland
| | - Pawel Kolasa
- Department of Neurosurgery, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
- Social Sciences Academy in Lodz, Lodz, Poland
| | - Ksenia Matlawska-Wasowska
- Division of Pediatric Research, Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Tomasz Sliwinski
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Tomasz Skorski
- Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
508
|
Castroviejo-Bermejo M, Cruz C, Llop-Guevara A, Gutiérrez-Enríquez S, Ducy M, Ibrahim YH, Gris-Oliver A, Pellegrino B, Bruna A, Guzmán M, Rodríguez O, Grueso J, Bonache S, Moles-Fernández A, Villacampa G, Viaplana C, Gómez P, Vidal M, Peg V, Serres-Créixams X, Dellaire G, Simard J, Nuciforo P, Rubio IT, Dienstmann R, Barrett JC, Caldas C, Baselga J, Saura C, Cortés J, Déas O, Jonkers J, Masson JY, Cairo S, Judde JG, O'Connor MJ, Díez O, Balmaña J, Serra V. A RAD51 assay feasible in routine tumor samples calls PARP inhibitor response beyond BRCA mutation. EMBO Mol Med 2018; 10:e9172. [PMID: 30377213 PMCID: PMC6284440 DOI: 10.15252/emmm.201809172] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are effective in cancers with defective homologous recombination DNA repair (HRR), including BRCA1/2-related cancers. A test to identify additional HRR-deficient tumors will help to extend their use in new indications. We evaluated the activity of the PARPi olaparib in patient-derived tumor xenografts (PDXs) from breast cancer (BC) patients and investigated mechanisms of sensitivity through exome sequencing, BRCA1 promoter methylation analysis, and immunostaining of HRR proteins, including RAD51 nuclear foci. In an independent BC PDX panel, the predictive capacity of the RAD51 score and the homologous recombination deficiency (HRD) score were compared. To examine the clinical feasibility of the RAD51 assay, we scored archival breast tumor samples, including PALB2-related hereditary cancers. The RAD51 score was highly discriminative of PARPi sensitivity versus PARPi resistance in BC PDXs and outperformed the genomic test. In clinical samples, all PALB2-related tumors were classified as HRR-deficient by the RAD51 score. The functional biomarker RAD51 enables the identification of PARPi-sensitive BC and broadens the population who may benefit from this therapy beyond BRCA1/2-related cancers.
Collapse
Affiliation(s)
| | - Cristina Cruz
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- High Risk and Familial Cancer Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Llop-Guevara
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Mandy Ducy
- Genome Stability Laboratory, CHU de Québec Research Center, Québec City, QC, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University Cancer Research Center, Québec City, QC, Canada
- CHU de Quebec - Université Laval Research Center, Genomics Center CHUL, Québec City, QC, Canada
| | - Yasir Hussein Ibrahim
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Albert Gris-Oliver
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Benedetta Pellegrino
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medical Oncology, University Hospital of Parma, Parma, Italy
| | - Alejandra Bruna
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Marta Guzmán
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Olga Rodríguez
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Judit Grueso
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Sandra Bonache
- Oncogenetics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Guillermo Villacampa
- Oncology Data Science (OdysSey Group), Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Cristina Viaplana
- Oncology Data Science (OdysSey Group), Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Patricia Gómez
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Breast Cancer and Melanoma Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Maria Vidal
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Breast Cancer and Melanoma Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Vicente Peg
- Pathology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Serres-Créixams
- Department of Radiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Graham Dellaire
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Jacques Simard
- CHU de Quebec - Université Laval Research Center, Genomics Center CHUL, Québec City, QC, Canada
| | - Paolo Nuciforo
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Isabel T Rubio
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Breast Surgical Unit, Breast Cancer Center, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (OdysSey Group), Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
- Breast Cancer Programme, Cancer Research UK (CRUK) Cambridge Cancer Centre, Cambridge, UK
| | - José Baselga
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cristina Saura
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Breast Cancer and Melanoma Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Javier Cortés
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Oncology, Ramón y Cajal University Hospital, Madrid, Spain
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Jos Jonkers
- Division of Molecular Pathology and Cancer Genomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU de Québec Research Center, Québec City, QC, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University Cancer Research Center, Québec City, QC, Canada
| | | | | | - Mark J O'Connor
- Oncology Innovative Medicines and Early Clinical Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - Orland Díez
- Oncogenetics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Clinical and Molecular Genetics Area, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Judith Balmaña
- High Risk and Familial Cancer Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
509
|
Hacker KE, Bolland DE, Tan L, Saha AK, Niknafs YS, Markovitz DM, McLean K. The DEK Oncoprotein Functions in Ovarian Cancer Growth and Survival. Neoplasia 2018; 20:1209-1218. [PMID: 30412857 PMCID: PMC6226625 DOI: 10.1016/j.neo.2018.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022]
Abstract
DNA damage repair alterations play a critical role in ovarian cancer tumorigenesis. Mechanistic drivers of the DNA damage response consequently present opportunities for therapeutic targeting. The chromatin-binding DEK oncoprotein functions in DNA double-strand break repair. We therefore sought to determine the role of DEK in epithelial ovarian cancer. DEK is overexpressed in both primary epithelial ovarian cancers and ovarian cancer cell lines. To assess the impact of DEK expression levels on cell growth, small interfering RNA and short hairpin RNA approaches were utilized. Decreasing DEK expression in ovarian cancer cell lines slows cell growth and induces apoptosis and DNA damage. The biologic effects of DEK depletion are enhanced with concurrent chemotherapy treatment. The in vitro effects of DEK knockdown are reproduced in vivo, as DEK depletion in a mouse xenograft model results in slower tumor growth and smaller tumors compared to tumors expressing DEK. These findings provide a compelling rationale to target the DEK oncoprotein and its pathways as a therapeutic strategy for treating epithelial ovarian cancer.
Collapse
Affiliation(s)
- Kari E Hacker
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Danielle E Bolland
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Lijun Tan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Anjan K Saha
- Department of Internal Medicine and Cancer Biology Program, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Yashar S Niknafs
- Michigan Center for Translational Pathology, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - David M Markovitz
- Department of Internal Medicine and Cancer Biology Program, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Karen McLean
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Michigan Medical Center, Ann Arbor, MI 48109.
| |
Collapse
|
510
|
Konstantinopoulos PA, Matulonis UA. Targeting DNA Damage Response and Repair as a Therapeutic Strategy for Ovarian Cancer. Hematol Oncol Clin North Am 2018; 32:997-1010. [DOI: 10.1016/j.hoc.2018.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
511
|
Freimund AE, Beach JA, Christie EL, Bowtell DD. Mechanisms of Drug Resistance in High-Grade Serous Ovarian Cancer. Hematol Oncol Clin North Am 2018; 32:983-996. [DOI: 10.1016/j.hoc.2018.07.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
512
|
Mishra B, Zhang S, Zhao H, Darzynkiewicz Z, Lee EY, Lee MY, Zhang Z. Discovery of a novel DNA polymerase inhibitor and characterization of its antiproliferative properties. Cancer Biol Ther 2018; 20:474-486. [PMID: 30427259 PMCID: PMC6422523 DOI: 10.1080/15384047.2018.1529126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 09/22/2018] [Indexed: 12/31/2022] Open
Abstract
Chromosomal duplication is targeted by various chemotherapeutic agents for the treatment of cancer. However, there is no specific inhibitor of DNA polymerases that is viable for cancer management. Through structure-based in silico screening of the ZINC database, we identified a specific inhibitor of DNA polymerase δ. The discovered inhibitor, Zelpolib, is projected to bind to the active site of Pol δ when it is actively engaged in DNA replication through interactions with DNA template and primer. Zelpolib shows robust inhibition of Pol δ activity in reconstituted DNA replication assays. Under cellular conditions, Zelpolib is taken up readily by cancer cells and inhibits DNA replication in assays to assess global DNA synthesis or single-molecule bases by DNA fiber fluorography. In addition, we show that Zelpolib displays superior antiproliferative properties to methotrexate, 5-flourouracil, and cisplatin in triple-negative breast cancer cell line, pancreatic cancer cell line and platinum-resistant pancreatic cancer cell line. Pol δ is not only involved in DNA replication, it is also a key component in many DNA repair pathways. Pol δ is the key enzyme responsible for D-loop extension during homologous recombination. Indeed, Zelpolib shows robust inhibition of homologous recombination repair of DNA double-strand breaks and induces "BRCAness" in HR-proficient cancer cells and enhances their sensitivity to PARP inhibitors.
Collapse
Affiliation(s)
- Bhanvi Mishra
- Department of Biochemistry and Molecular Biology, Valhalla, NY, USA
| | - Sufang Zhang
- Department of Biochemistry and Molecular Biology, Valhalla, NY, USA
| | - Hong Zhao
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | | | - Ernest Y.C. Lee
- Department of Biochemistry and Molecular Biology, Valhalla, NY, USA
| | | | - Zhongtao Zhang
- Department of Biochemistry and Molecular Biology, Valhalla, NY, USA
| |
Collapse
|
513
|
Kotoula V, Lakis S, Tikas I, Giannoulatou E, Lazaridis G, Papadopoulou K, Manoussou K, Efstratiou I, Papanikolaou A, Fostira F, Vlachos I, Tarlatzis B, Fountzilas G. Pathogenic BRCA1 mutations may be necessary but not sufficient for tissue genomic heterogeneity: Deep sequencing data from ovarian cancer patients. Gynecol Oncol 2018; 152:375-386. [PMID: 30446274 DOI: 10.1016/j.ygyno.2018.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/05/2018] [Accepted: 11/11/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Tissue genomic heterogeneity (t-HET) in patients with epithelial ovarian cancer (OVCA) is related to tissue plasticity, i.e., flexibility to adapt to adverse molecular environments. Here, we interrogated the presence and clinical relevance of OVCA t-HET. METHODS We applied high-depth (>2000×) sequencing on 297 paraffin tissue samples (fallopian tubes, ovaries, intra-abdominal metastases) from 71 treatment-naïve patients who subsequently received first-line platinum-based chemotherapy. Based on tissue mutation patterns, we distinguished tissue genotypes into: no mutation (33/297 samples; 11.1%), stable (173; 58.2%) and unstable (91; 30.7%). We profiled genotypes per patient and assessed t-HET in 69 patients. Predicted pathogenic mutations refer to germline and/or tissues. RESULTS Among all 71 patients, 46 (64.8%) had pathogenic BRCA1 mutations and 15 (21.7%) had BRCA1/2 disruption (i.e., pathogenic mutations with position-LOH). We classified 29 patients with t-HET (42%), all with pathogenic BRCA1; t-HET was observed in 64% with such mutations (p < 0.001). As opposed to non-t-HET, matched tissues in t-HET shared pathogenic BRCA1 (p < 0.001) but not BRCA2 and TP53. Germline BRCA1 mutations in tissues exhibited position-LOH; heterozygous status; or, partial loss of the inherited allele accompanied by additional clonal mutations. Patients with t-HET had worse outcome (log-rank p = 0.048 [progression-free]; p = 0.037 [overall survival]), including 12/15 patients with disrupted BRCA1/2 and 3 BRCA1 carriers with partial germline loss in tissues. CONCLUSIONS Pathogenic BRCA1 mutations appear necessary but may not be sufficient for the establishment of t-HET. t-HET may be associated with worse outcome, including in patients with disrupted BRCA1/2, which is usually considered as a favourable marker. OVCA t-HET may need to be addressed for treatment decisions.
Collapse
Affiliation(s)
- Vassiliki Kotoula
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Thessaloniki, Greece; Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece.
| | - Sotirios Lakis
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Tikas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; University of New South Wales, Kensington, NSW, Australia
| | - Georgios Lazaridis
- Department of Medical Oncology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece
| | - Kyriaki Papadopoulou
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriaki Manoussou
- Section of Biostatistics, Hellenic Cooperative Oncology Group, Athens, Greece.
| | | | - Alexios Papanikolaou
- First Department of Obstetrics and Gynecology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece.
| | - Florentia Fostira
- Molecular Diagnostics Laboratory, INRASTES, National Center for Scientific Research NCSR Demokritos, Athens, Greece
| | - Ioannis Vlachos
- Molecular Diagnostics Laboratory, INRASTES, National Center for Scientific Research NCSR Demokritos, Athens, Greece.
| | - Basil Tarlatzis
- First Department of Obstetrics and Gynecology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Thessaloniki, Greece; Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
514
|
Zeng M, Kwiatkowski NP, Zhang T, Nabet B, Xu M, Liang Y, Quan C, Wang J, Hao M, Palakurthi S, Zhou S, Zeng Q, Kirschmeier PT, Meghani K, Leggett AL, Qi J, Shapiro GI, Liu JF, Matulonis UA, Lin CY, Konstantinopoulos PA, Gray NS. Targeting MYC dependency in ovarian cancer through inhibition of CDK7 and CDK12/13. eLife 2018; 7:39030. [PMID: 30422115 PMCID: PMC6251623 DOI: 10.7554/elife.39030] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022] Open
Abstract
High-grade serous ovarian cancer is characterized by extensive copy number alterations, among which the amplification of MYC oncogene occurs in nearly half of tumors. We demonstrate that ovarian cancer cells highly depend on MYC for maintaining their oncogenic growth, indicating MYC as a therapeutic target for this difficult-to-treat malignancy. However, targeting MYC directly has proven difficult. We screen small molecules targeting transcriptional and epigenetic regulation, and find that THZ1 - a chemical inhibiting CDK7, CDK12, and CDK13 - markedly downregulates MYC. Notably, abolishing MYC expression cannot be achieved by targeting CDK7 alone, but requires the combined inhibition of CDK7, CDK12, and CDK13. In 11 patient-derived xenografts models derived from heavily pre-treated ovarian cancer patients, administration of THZ1 induces significant tumor growth inhibition with concurrent abrogation of MYC expression. Our study indicates that targeting these transcriptional CDKs with agents such as THZ1 may be an effective approach for MYC-dependent ovarian malignancies.
Collapse
Affiliation(s)
- Mei Zeng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Nicholas P Kwiatkowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Behnam Nabet
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Mousheng Xu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Medicine, Harvard Medical School, Boston, United States
| | - Yanke Liang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Chunshan Quan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Mingfeng Hao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Sangeetha Palakurthi
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, United States
| | - Shan Zhou
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, United States
| | - Qing Zeng
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, United States
| | - Paul T Kirschmeier
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, United States
| | - Khyati Meghani
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, United States
| | - Alan L Leggett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Medicine, Harvard Medical School, Boston, United States
| | - Geoffrey I Shapiro
- Early Drug Development Center, Dana-Farber Cancer Institute, Boston, United States
| | - Joyce F Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, United States
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, United States
| | - Charles Y Lin
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | | | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| |
Collapse
|
515
|
Stover EH, Feltmate C, Berkowitz RS, Lindeman NI, Matulonis UA, Konstantinopoulos PA. Targeted Next-Generation Sequencing Reveals Clinically Actionable BRAF and ESR1 Mutations in Low-Grade Serous Ovarian Carcinoma. JCO Precis Oncol 2018; 2018. [PMID: 30828692 PMCID: PMC6394870 DOI: 10.1200/po.18.00135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Elizabeth H Stover
- Elizabeth H. Stover, Ursula A. Matulonis, and Panagiotis A. Konstantinopoulos, Dana-Farber Cancer Institute, Harvard Medical School; and Colleen Feltmate, Ross S. Berkowitz, and Neal I. Lindeman, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Colleen Feltmate
- Elizabeth H. Stover, Ursula A. Matulonis, and Panagiotis A. Konstantinopoulos, Dana-Farber Cancer Institute, Harvard Medical School; and Colleen Feltmate, Ross S. Berkowitz, and Neal I. Lindeman, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ross S Berkowitz
- Elizabeth H. Stover, Ursula A. Matulonis, and Panagiotis A. Konstantinopoulos, Dana-Farber Cancer Institute, Harvard Medical School; and Colleen Feltmate, Ross S. Berkowitz, and Neal I. Lindeman, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Neal I Lindeman
- Elizabeth H. Stover, Ursula A. Matulonis, and Panagiotis A. Konstantinopoulos, Dana-Farber Cancer Institute, Harvard Medical School; and Colleen Feltmate, Ross S. Berkowitz, and Neal I. Lindeman, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ursula A Matulonis
- Elizabeth H. Stover, Ursula A. Matulonis, and Panagiotis A. Konstantinopoulos, Dana-Farber Cancer Institute, Harvard Medical School; and Colleen Feltmate, Ross S. Berkowitz, and Neal I. Lindeman, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Panagiotis A Konstantinopoulos
- Elizabeth H. Stover, Ursula A. Matulonis, and Panagiotis A. Konstantinopoulos, Dana-Farber Cancer Institute, Harvard Medical School; and Colleen Feltmate, Ross S. Berkowitz, and Neal I. Lindeman, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
516
|
Chartron E, Theillet C, Guiu S, Jacot W. Targeting homologous repair deficiency in breast and ovarian cancers: Biological pathways, preclinical and clinical data. Crit Rev Oncol Hematol 2018; 133:58-73. [PMID: 30661659 DOI: 10.1016/j.critrevonc.2018.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/25/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
Mutation or epigenetic silencing of homologous recombination (HR) repair genes is characteristic of a growing proportion of triple-negative breast cancers (TNBCs) and high-grade serous ovarian carcinomas. Defects in HR lead to genome instability, allowing cells to acquire the multiple genetic alterations essential for cancer development. However, this deficiency can also be exploited by using DNA damaging agents or by targeting compensatory repair pathways. A noteworthy example is treatment of TNBC and epithelial ovarian cancer harboring BRCA1/2 germline mutations using platinum salts and/or PARP inhibitors. Dramatic responses to PARP inhibitors may support a wider use in the HR-deficient population beyond those with mutated germline BRCA1 and 2. In this review, we discuss HR deficiency hallmarks as predictive biomarkers for platinum salt and PARP inhibitor sensitivity for selecting patients affected by TNBC or epithelial ovarian cancer who could benefit from these therapeutic options.
Collapse
Affiliation(s)
- Elodie Chartron
- Department of medical oncology, Montpellier Academic Hospital, Montpellier, France
| | - Charles Theillet
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France
| | - Séverine Guiu
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - William Jacot
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France; Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France.
| |
Collapse
|
517
|
Pierce SR, Clark LH. Current First-line Therapy for Ovarian Cancer: A Comprehensive Review. Obstet Gynecol Surv 2018; 73:650-657. [DOI: 10.1097/ogx.0000000000000613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
518
|
Hill SJ, Decker B, Roberts EA, Horowitz NS, Muto MG, Worley MJ, Feltmate CM, Nucci MR, Swisher EM, Nguyen H, Yang C, Morizane R, Kochupurakkal BS, Do KT, Konstantinopoulos PA, Liu JF, Bonventre JV, Matulonis UA, Shapiro GI, Berkowitz RS, Crum CP, D'Andrea AD. Prediction of DNA Repair Inhibitor Response in Short-Term Patient-Derived Ovarian Cancer Organoids. Cancer Discov 2018; 8:1404-1421. [PMID: 30213835 PMCID: PMC6365285 DOI: 10.1158/2159-8290.cd-18-0474] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/15/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022]
Abstract
Based on genomic analysis, 50% of high-grade serous ovarian cancers (HGSC) are predicted to have DNA repair defects. Whether this substantial subset of HGSCs actually have functional repair defects remains unknown. Here, we devise a platform for functional profiling of DNA repair in short-term patient-derived HGSC organoids. We tested 33 organoid cultures derived from 22 patients with HGSC for defects in homologous recombination (HR) and replication fork protection. Regardless of DNA repair gene mutational status, a functional defect in HR in the organoids correlated with PARP inhibitor sensitivity. A functional defect in replication fork protection correlated with carboplatin and CHK1 and ATR inhibitor sensitivity. Our results indicate that a combination of genomic analysis and functional testing of organoids allows for the identification of targetable DNA damage repair defects. Larger numbers of patient-derived organoids must be analyzed to determine whether these assays can reproducibly predict patient response in the clinic.Significance: Patient-derived ovarian tumor organoids grow rapidly and match the tumors from which they are derived, both genetically and functionally. These organoids can be used for DNA repair profiling and therapeutic sensitivity testing and provide a rapid means of assessing targetable defects in the parent tumor, offering more suitable treatment options. Cancer Discov; 8(11); 1404-21. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 1333.
Collapse
Affiliation(s)
- Sarah J Hill
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brennan Decker
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emma A Roberts
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Neil S Horowitz
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael G Muto
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael J Worley
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Colleen M Feltmate
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marisa R Nucci
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth M Swisher
- Division of Gynecologic Oncology, University of Washington, Seattle, Washington
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington
| | - Huy Nguyen
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Chunyu Yang
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ryuji Morizane
- Renal Division, Brigham and Women's Hospital, Boston, Massachusetts; Department of Medicine, Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Bose S Kochupurakkal
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Khanh T Do
- Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Joyce F Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Joseph V Bonventre
- Renal Division, Brigham and Women's Hospital, Boston, Massachusetts; Department of Medicine, Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Geoffrey I Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
- Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ross S Berkowitz
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Christopher P Crum
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
519
|
Romeo M, Pardo JC, Martínez-Cardús A, Martínez-Balibrea E, Quiroga V, Martínez-Román S, Solé F, Margelí M, Mesía R. Translational Research Opportunities Regarding Homologous Recombination in Ovarian Cancer. Int J Mol Sci 2018; 19:ijms19103249. [PMID: 30347758 PMCID: PMC6214122 DOI: 10.3390/ijms19103249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 09/29/2018] [Accepted: 10/16/2018] [Indexed: 02/07/2023] Open
Abstract
Homologous recombination (HR) is a DNA repair pathway that is deficient in 50% of high-grade serous ovarian carcinomas (HGSOC). Deficient HR (DHR) constitutes a therapeutic opportunity for these patients, thanks to poly (ADP-ribose) polymerases (PARP) inhibitors (PARPi; olaparib, niraparib, and rucaparib are already commercialized). Although initially, PARPi were developed for patients with BRCA1/2 mutations, robust clinical data have shown their benefit in a broader population without DHR. This breakthrough in daily practice has raised several questions that necessitate further research: How can populations that will most benefit from PARPi be selected? At which stage of ovarian cancer should PARPi be used? Which strategies are reasonable to overcome PARPi resistance? In this paper, we present a summary of the literature and discuss the present clinical research involving PARPi (after reviewing ClinicalTrials.gov) from a translational perspective. Research into the functional biomarkers of DHR and clinical trials testing PARPi benefits as first-line setting or rechallenge are currently ongoing. Additionally, in the clinical setting, only secondary restoring mutations of BRCA1/2 have been identified as events inducing resistance to PARPi. The clinical frequency of this and other mechanisms that have been described in preclinics is unknown. It is of great importance to study mechanisms of resistance to PARPi to guide the clinical development of drug combinations.
Collapse
Affiliation(s)
- Margarita Romeo
- Medical Oncology Department, B-ARGO Group, Institut Català d'Oncologia Badalona, Carretera del Canyet s/n, 08916 Badalona, Spain.
- Campus de la UAB, Universitat Autónoma de Barcelona, Plaça Cívica, 08193 Bellaterra, Spain.
| | - Juan Carlos Pardo
- Medical Oncology Department, B-ARGO Group, Institut Català d'Oncologia Badalona, Carretera del Canyet s/n, 08916 Badalona, Spain.
| | - Anna Martínez-Cardús
- Health Sciences Research Institute of the Germans Trias i Pujol Foundation (IGTP), B-ARGO Group, Carretera del Canyet s/n, 08916 Badalona, Spain.
| | - Eva Martínez-Balibrea
- Program against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia Badalona, Program for Predictive and Personalized Cancer Medicine (PMPPC), Health Sciences Research Institute Germans Trias i Pujo (IGTP), Carretera de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Spain.
| | - Vanesa Quiroga
- Medical Oncology Department, B-ARGO Group, Institut Català d'Oncologia Badalona, Carretera del Canyet s/n, 08916 Badalona, Spain.
| | - Sergio Martínez-Román
- Gynecology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain.
| | - Francesc Solé
- Institut de Recerca contra la Leucemia Josep Carreras, 08916 Badalona, Spain.
| | - Mireia Margelí
- Medical Oncology Department, B-ARGO Group, Institut Català d'Oncologia Badalona, Carretera del Canyet s/n, 08916 Badalona, Spain.
| | - Ricard Mesía
- Medical Oncology Department, B-ARGO Group, Institut Català d'Oncologia Badalona, Carretera del Canyet s/n, 08916 Badalona, Spain.
| |
Collapse
|
520
|
Lallo A, Frese KK, Morrow CJ, Sloane R, Gulati S, Schenk MW, Trapani F, Simms N, Galvin M, Brown S, Hodgkinson CL, Priest L, Hughes A, Lai Z, Cadogan E, Khandelwal G, Simpson KL, Miller C, Blackhall F, O'Connor MJ, Dive C. The Combination of the PARP Inhibitor Olaparib and the WEE1 Inhibitor AZD1775 as a New Therapeutic Option for Small Cell Lung Cancer. Clin Cancer Res 2018; 24:5153-5164. [PMID: 29941481 DOI: 10.1158/1078-0432.ccr-17-2805] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/05/2018] [Accepted: 06/20/2018] [Indexed: 12/12/2022]
Abstract
Purpose: Introduced in 1987, platinum-based chemotherapy remains standard of care for small cell lung cancer (SCLC), a most aggressive, recalcitrant tumor. Prominent barriers to progress are paucity of tumor tissue to identify drug targets and patient-relevant models to interrogate novel therapies. Following our development of circulating tumor cell patient-derived explants (CDX) as models that faithfully mirror patient disease, here we exploit CDX to examine new therapeutic options for SCLC.Experimental Design: We investigated the efficacy of the PARP inhibitor olaparib alone or in combination with the WEE1 kinase inhibitor AZD1775 in 10 phenotypically distinct SCLC CDX in vivo and/or ex vivo These CDX represent chemosensitive and chemorefractory disease including the first reported paired CDX generated longitudinally before treatment and upon disease progression.Results: There was a heterogeneous depth and duration of response to olaparib/AZD1775 that diminished when tested at disease progression. However, efficacy of this combination consistently exceeded that of cisplatin/etoposide, with cures in one CDX model. Genomic and protein analyses revealed defects in homologous recombination repair genes and oncogenes that induce replication stress (such as MYC family members), predisposed CDX to combined olaparib/AZD1775 sensitivity, although universal predictors of response were not noted.Conclusions: These preclinical data provide a strong rationale to trial this combination in the clinic informed by prevalent, readily accessed circulating tumor cell-based biomarkers. New therapies will be evaluated in SCLC patients after first-line chemotherapy, and our data suggest that the combination of olaparib/AZD1775 should be used as early as possible and before disease relapse. Clin Cancer Res; 24(20); 5153-64. ©2018 AACR.
Collapse
Affiliation(s)
- Alice Lallo
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Kristopher K Frese
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Christopher J Morrow
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Robert Sloane
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Sakshi Gulati
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Maximillian W Schenk
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Francesca Trapani
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Nicole Simms
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Melanie Galvin
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Stewart Brown
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Cassandra L Hodgkinson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Lynsey Priest
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Adina Hughes
- Oncology Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Zhongwu Lai
- Oncology Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Waltham, Massachusetts
| | - Elaine Cadogan
- Oncology Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Garima Khandelwal
- RNA Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Kathryn L Simpson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Crispin Miller
- RNA Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Fiona Blackhall
- Institute of Cancer Sciences, University of Manchester, and Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Mark J O'Connor
- Oncology Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, United Kingdom.
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
521
|
Engqvist H, Parris TZ, Rönnerman EW, Söderberg EMV, Biermann J, Mateoiu C, Sundfeldt K, Kovács A, Karlsson P, Helou K. Transcriptomic and genomic profiling of early-stage ovarian carcinomas associated with histotype and overall survival. Oncotarget 2018; 9:35162-35180. [PMID: 30416686 PMCID: PMC6205557 DOI: 10.18632/oncotarget.26225] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/01/2018] [Indexed: 12/28/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy in the western world. Despite recent efforts to characterize ovarian cancer using molecular profiling, few targeted treatment options are currently available. Here, we examined genetic variants, fusion transcripts, SNP genotyping, and gene expression patterns for early-stage (I and II) ovarian carcinomas (n=96) in relation to clinicopathological characteristics and clinical outcome, thereby identifying novel genetic features of ovarian carcinomas. Furthermore, mutation frequencies of specific genetic variants and/or their gene expression patterns were associated with histotype and overall survival, e.g. SLC28A2 (mucinous ovarian carcinoma histotype), ARCN1 (low expression in 0-2 year survival group), and tumor suppressor MTUS1 (mutation status and overall survival). The long non-coding RNA MALAT1 was identified as a highly promiscuous fusion transcript in ovarian carcinoma. Moreover, gene expression deregulation for 23 genes was associated with tumor aggressiveness. Taken together, the novel biomarkers identified here may improve ovarian carcinoma subclassification and patient stratification according to histotype and overall survival.
Collapse
Affiliation(s)
- Hanna Engqvist
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Werner Rönnerman
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Sahlgrenska University Hospital, Department of Clinical Pathology and Genetics, Gothenburg, Sweden
| | - Elin M V Söderberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jana Biermann
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claudia Mateoiu
- Sahlgrenska University Hospital, Department of Clinical Pathology and Genetics, Gothenburg, Sweden
| | - Karin Sundfeldt
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anikó Kovács
- Sahlgrenska University Hospital, Department of Clinical Pathology and Genetics, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
522
|
Zhai B, Steinø A, Bacha J, Brown D, Daugaard M. Dianhydrogalactitol induces replication-dependent DNA damage in tumor cells preferentially resolved by homologous recombination. Cell Death Dis 2018; 9:1016. [PMID: 30283085 PMCID: PMC6170372 DOI: 10.1038/s41419-018-1069-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/30/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
1,2:5,6-Dianhydrogalactitol (DAG) is a bifunctional DNA-targeting agent causing N7-guanine alkylation and inter-strand DNA crosslinks currently in clinical trial for treatment of glioblastoma. While preclinical studies and clinical trials have demonstrated antitumor activity of DAG in a variety of malignancies, understanding the molecular mechanisms underlying DAG-induced cytotoxicity is essential for proper clinical qualification. Using non-small cell lung cancer (NSCLC) as a model system, we show that DAG-induced cytotoxicity materializes when cells enter S phase with unrepaired N7-guanine DNA crosslinks. In S phase, DAG-mediated DNA crosslink lesions translated into replication-dependent DNA double-strand breaks (DSBs) that subsequently triggered irreversible cell cycle arrest and loss of viability. DAG-treated NSCLC cells attempt to repair the DSBs by homologous recombination (HR) and inhibition of the HR repair pathway sensitized NSCLC cells to DAG-induced DNA damage. Accordingly, our work describes a molecular mechanism behind N7-guanine crosslink-induced cytotoxicity in cancer cells and provides a rationale for using DAG analogs to treat HR-deficient tumors.
Collapse
Affiliation(s)
- Beibei Zhai
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Anne Steinø
- DelMar Pharmaceuticals, Inc., Vancouver, BC, V5Z 1K5, Canada.,DelMar Pharmaceuticals, Inc., Menlo Park, CA, 94025, USA
| | - Jeffrey Bacha
- DelMar Pharmaceuticals, Inc., Vancouver, BC, V5Z 1K5, Canada.,DelMar Pharmaceuticals, Inc., Menlo Park, CA, 94025, USA
| | - Dennis Brown
- DelMar Pharmaceuticals, Inc., Vancouver, BC, V5Z 1K5, Canada.,DelMar Pharmaceuticals, Inc., Menlo Park, CA, 94025, USA
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada. .,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
523
|
Brovkina OI, Shigapova L, Chudakova DA, Gordiev MG, Enikeev RF, Druzhkov MO, Khodyrev DS, Shagimardanova EI, Nikitin AG, Gusev OA. The Ethnic-Specific Spectrum of Germline Nucleotide Variants in DNA Damage Response and Repair Genes in Hereditary Breast and Ovarian Cancer Patients of Tatar Descent. Front Oncol 2018; 8:421. [PMID: 30333958 PMCID: PMC6176317 DOI: 10.3389/fonc.2018.00421] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022] Open
Abstract
The Russian population consists of more than 100 ethnic groups, presenting a unique opportunity for the identification of hereditary pathogenic mutations. To gain insight into the landscape of heredity pathogenic variants, we employed targeted next-generation sequencing to analyze the germline mutation load in the DNA damage response and repair genes of hereditary breast and ovary cancer syndrome (HBOCS) patients of Tatar ethnicity, which represents ~4% of the total Russian population. Several pathogenic mutations were identified in DNA double-strand break repair genes, and the spectrum of these markers in Tatar patients varied from that previously reported for patients of Slavic ancestry. The CDK12 gene encodes cyclin-dependent kinase 12, the key transcriptional regulator of the genes involved in DNA damage response and repair. CDK12 analysis in a cohort of HBOCS patients of Tatar decent identified a c.1047-2A>G nucleotide variant in the CDK12 gene in 8 of the 106 cases (7.6%). The c.1047-2A>G nucleotide variant was identified in 1 of the 93 (1.1%) HBOCS patients with mixed or unknown ethnicity and in 1 of the 238 (0.42%) healthy control patients of mixed ethnicity (Tatars and non-Tatars) (p = 0.0066, OR = 11.18, CI 95% = 1.53-492.95, Tatar and non-Tatar patients vs. healthy controls). In a group of mixed ethnicity patients from Tatarstan, with sporadic breast and/or ovarian cancer, this nucleotide variant was detected in 2 out of 93 (2.2%) cases. In a cohort of participants of Slavic descent from Moscow, comprising of 95 HBOCS patients, 80 patients with sporadic breast and/or ovarian cancer, and 372 healthy controls, this nucleotide variant was absent. Our study demonstrates a strong predisposition for the CDK12 c.1047-2A>G nucleotide variant in HBOCS in patients of Tatar ethnicity and identifies CDK12 as a novel gene involved in HBOCS susceptibility.
Collapse
Affiliation(s)
- Olga I Brovkina
- Federal Research and Clinical Center, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | | | - Daria A Chudakova
- Institute of Natural and Mathematical Sciences, Massey University, Auckland, New Zealand
| | - Marat G Gordiev
- Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Tatarstan, Kazan, Russia
| | - Rafael F Enikeev
- Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Tatarstan, Kazan, Russia
| | - Maxim O Druzhkov
- Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Tatarstan, Kazan, Russia
| | - Dmitriy S Khodyrev
- Federal Research and Clinical Center, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | | | - Alexey G Nikitin
- Federal Research and Clinical Center, Federal Medical-Biological Agency of Russia, Moscow, Russia.,Pulmonology Research Institute, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Oleg A Gusev
- Kazan (Volga Region) Federal University, Kazan, Russia.,RIKEN, Yokohama, Japan
| |
Collapse
|
524
|
Curigliano G. Gyneco-oncological genomics and emerging biomarkers for cancer treatment with immune-checkpoint inhibitors. Semin Cancer Biol 2018; 52:253-258. [DOI: 10.1016/j.semcancer.2018.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
|
525
|
Haynes B, Murai J, Lee JM. Restored replication fork stabilization, a mechanism of PARP inhibitor resistance, can be overcome by cell cycle checkpoint inhibition. Cancer Treat Rev 2018; 71:1-7. [PMID: 30269007 DOI: 10.1016/j.ctrv.2018.09.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/06/2018] [Accepted: 09/09/2018] [Indexed: 02/06/2023]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibition serves as a potent therapeutic option eliciting synthetic lethality in cancers harboring homologous recombination (HR) repair defects, such as BRCA mutations. However, the development of resistance to PARP inhibitors (PARPis) poses a clinical challenge. Restoration of HR competency is one of the many molecular factors contributing to PARPi resistance. Combination therapy with cell cycle checkpoint (ATR, CHK1, and WEE1) inhibitors is being investigated clinically in many cancers, particularly in ovarian cancer, to enhance the efficacy and circumvent resistance to PARPis. Ideally, inhibition of ATR, CHK1 and WEE1 proteins will abrogate G2 arrest and subsequent DNA repair via restored HR in PARPi-treated cells. Replication fork stabilization has recently been identified as a potential compensatory PARPi resistance mechanism, found in the absence of restored HR. ATR, CHK1, and WEE1 each possess different roles in replication fork stabilization, providing different mechanisms to consider when developing combination therapies to avoid continued development of drug resistance. This review examines the impact of ATR, CHK1, and WEE1 on replication fork stabilization. We also address the therapeutic potential for combining PARPis with cell cycle inhibitors and the possible consequence of combination therapies which do not adequately address both restored HR and replication fork stabilization as PARPi resistance mechanisms.
Collapse
Affiliation(s)
- Brittany Haynes
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Junko Murai
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
526
|
Mattoo AR, Joun A, Jessup JM. Repurposing of mTOR Complex Inhibitors Attenuates MCL-1 and Sensitizes to PARP Inhibition. Mol Cancer Res 2018; 17:42-53. [DOI: 10.1158/1541-7786.mcr-18-0650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/09/2018] [Accepted: 08/30/2018] [Indexed: 11/16/2022]
|
527
|
Prati B, Marangoni B, Boccardo E. Human papillomavirus and genome instability: from productive infection to cancer. Clinics (Sao Paulo) 2018; 73:e539s. [PMID: 30208168 PMCID: PMC6113919 DOI: 10.6061/clinics/2018/e539s] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/16/2018] [Indexed: 12/29/2022] Open
Abstract
Infection with high oncogenic risk human papillomavirus types is the etiological factor of cervical cancer and a major cause of other epithelial malignancies, including vulvar, vaginal, anal, penile and head and neck carcinomas. These agents affect epithelial homeostasis through the expression of specific proteins that deregulate important cellular signaling pathways to achieve efficient viral replication. Among the major targets of viral proteins are components of the DNA damage detection and repair machinery. The activation of many of these cellular factors is critical to process viral genome replication intermediates and, consequently, to sustain faithful viral progeny production. In addition to the important role of cellular DNA repair machinery in the infective human papillomavirus cycle, alterations in the expression and activity of many of its components are observed in human papillomavirus-related tumors. Several studies from different laboratories have reported the impact of the expression of human papillomavirus oncogenes, mainly E6 and E7, on proteins in almost all the main cellular DNA repair mechanisms. This has direct consequences on cellular transformation since it causes the accumulation of point mutations, insertions and deletions of short nucleotide stretches, as well as numerical and structural chromosomal alterations characteristic of tumor cells. On the other hand, it is clear that human papillomavirus-transformed cells depend on the preservation of a basal cellular DNA repair activity level to maintain tumor cell viability. In this review, we summarize the data concerning the effect of human papillomavirus infection on DNA repair mechanisms. In addition, we discuss the potential of exploiting human papillomavirus-transformed cell dependency on DNA repair pathways as effective antitumoral therapies.
Collapse
Affiliation(s)
- Bruna Prati
- Departamento de Microbiologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Bruna Marangoni
- Departamento de Microbiologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Enrique Boccardo
- Departamento de Microbiologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
528
|
Beshiri ML, Tice CM, Tran C, Nguyen HM, Sowalsky AG, Agarwal S, Jansson KH, Yang Q, McGowen KM, Yin J, Alilin AN, Karzai FH, Dahut WL, Corey E, Kelly K. A PDX/Organoid Biobank of Advanced Prostate Cancers Captures Genomic and Phenotypic Heterogeneity for Disease Modeling and Therapeutic Screening. Clin Cancer Res 2018; 24:4332-4345. [PMID: 29748182 PMCID: PMC6125202 DOI: 10.1158/1078-0432.ccr-18-0409] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/06/2018] [Accepted: 05/07/2018] [Indexed: 12/22/2022]
Abstract
Purpose: Prostate cancer translational research has been hampered by the lack of comprehensive and tractable models that represent the genomic landscape of clinical disease. Metastatic castrate-resistant prostate cancer (mCRPC) patient-derived xenografts (PDXs) recapitulate the genetic and phenotypic diversity of the disease. We sought to establish a representative, preclinical platform of PDX-derived organoids that is experimentally facile for high-throughput and mechanistic analysis.Experimental Design: Using 20 models from the LuCaP mCRPC PDX cohort, including adenocarcinoma and neuroendocrine lineages, we systematically tested >20 modifications to prostate organoid conditions. Organoids were evaluated for genomic and phenotypic stability and continued reliance on the AR signaling pathway. The utility of the platform as a genotype-dependent model of drug sensitivity was tested with olaparib and carboplatin.Results: All PDX models proliferated as organoids in culture. Greater than 50% could be continuously cultured long-term in modified conditions; however, none of the PDXs could be established long-term as organoids under previously reported conditions. In addition, the modified conditions improved the establishment of patient biopsies over current methods. The genomic heterogeneity of the PDXs was conserved in organoids. Lineage markers and transcriptomes were maintained between PDXs and organoids. Dependence on AR signaling was preserved in adenocarcinoma organoids, replicating a dominant characteristic of CRPC. Finally, we observed maximum cytotoxicity to the PARP inhibitor olaparib in BRCA2-/- organoids, similar to responses observed in patients.Conclusions: The LuCaP PDX/organoid models provide an expansive, genetically characterized platform to investigate the mechanisms of pathogenesis as well as therapeutic responses and their molecular correlates in mCRPC. Clin Cancer Res; 24(17); 4332-45. ©2018 AACR.
Collapse
Affiliation(s)
- Michael L Beshiri
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Caitlin M Tice
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Crystal Tran
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Holly M Nguyen
- Department of Urology, University of Washington, Seattle, Washington
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Supreet Agarwal
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Keith H Jansson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Qi Yang
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Kerry M McGowen
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - JuanJuan Yin
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Aian Neil Alilin
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Fatima H Karzai
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - William L Dahut
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
529
|
Chedgy ECP, Vandekerkhove G, Herberts C, Annala M, Donoghue AJ, Sigouros M, Ritch E, Struss W, Konomura S, Liew J, Parimi S, Vergidis J, Hurtado-Coll A, Sboner A, Fazli L, Beltran H, Chi KN, Wyatt AW. Biallelic tumour suppressor loss and DNA repair defects in de novo
small-cell prostate carcinoma. J Pathol 2018; 246:244-253. [DOI: 10.1002/path.5137] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/18/2018] [Accepted: 07/05/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Edmund CP Chedgy
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Gillian Vandekerkhove
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Matti Annala
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
- Institute of Biosciences and Medical Technology; University of Tampere; Tampere Finland
| | - Adam J Donoghue
- Department of Medicine, Division of Hematology and Medical Oncology; Weill Cornell Medical College; New York NY USA
| | - Michael Sigouros
- Department of Medicine, Division of Hematology and Medical Oncology; Weill Cornell Medical College; New York NY USA
| | - Elie Ritch
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Werner Struss
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Saki Konomura
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Janet Liew
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Sunil Parimi
- Department of Medical Oncology; British Columbia Cancer Agency; British Columbia Canada
| | - Joanna Vergidis
- Department of Medical Oncology; British Columbia Cancer Agency; British Columbia Canada
| | - Antonio Hurtado-Coll
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Andrea Sboner
- Department of Medicine, Division of Hematology and Medical Oncology; Weill Cornell Medical College; New York NY USA
| | - Ladan Fazli
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| | - Himisha Beltran
- Department of Medicine, Division of Hematology and Medical Oncology; Weill Cornell Medical College; New York NY USA
| | - Kim N Chi
- Department of Medical Oncology; British Columbia Cancer Agency; British Columbia Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences; University of British Columbia; British Columbia Canada
| |
Collapse
|
530
|
Dal Molin GZ, Westin SN, Coleman RL. Rucaparib in ovarian cancer: extending the use of PARP inhibitors in the recurrent disease. Future Oncol 2018; 14:3101-3110. [PMID: 30105925 DOI: 10.2217/fon-2018-0215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rucaparib is a potent inhibitor of poly (ADP-ribose) polymerase (PARP) PARP1, PARP2 and PARP3, and to a lesser extent, PARP4, PARP10, PARP12, PARP15 and PARP16. Study 10 and ARIEL2 evaluated the use of rucaparib as treatment in patients with recurrent high-grade ovarian carcinoma and resulting in approval of rucaparib for patients with both germline and somatic BRCA mutation. Data from the Phase III trial ARIEL3 led to approval in platinum-sensitive disease as maintenance. This article reviews the efficacy, safety, pharmacokinetics and pharmacodynamics of rucaparib as well as future and ongoing trials.
Collapse
Affiliation(s)
- Graziela Z Dal Molin
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
531
|
Tessari A, Parbhoo K, Pawlikowski M, Fassan M, Rulli E, Foray C, Fabbri A, Embrione V, Ganzinelli M, Capece M, Campbell MJ, Broggini M, La Perle K, Farina G, Cole S, Marabese M, Hernandez M, Amann JM, Pruneri G, Carbone DP, Garassino MC, Croce CM, Palmieri D, Coppola V. RANBP9 affects cancer cells response to genotoxic stress and its overexpression is associated with worse response to platinum in NSCLC patients. Oncogene 2018; 37:6463-6476. [PMID: 30076413 DOI: 10.1038/s41388-018-0424-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/28/2018] [Accepted: 07/05/2018] [Indexed: 01/11/2023]
Abstract
Although limited by severe side effects and development of resistance, platinum-based therapies still represent the most common first-line treatment for non-small cell lung cancer (NSCLC). However, a crucial need in the clinical management of NSCLC is represented by the identification of cases sensitive to DNA damage response (DDR)-targeting drugs, such as cisplatin or PARP inhibitors. Here, we provide a molecular rationale for the stratification of NSCLC patients potentially benefitting from platinum compounds based on the expression levels of RANBP9, a recently identified player of the cellular DDR. RANBP9 was found overexpressed by immunohistochemistry (IHC) in NSCLC compared to normal adjacent tissues (NATs) (n = 147). Moreover, a retrospective analysis of 132 platinum-treated patients from the multi-centric TAILOR trial showed that RANBP9 overexpression levels are associated with clinical response to platinum compounds [Progression Free Survival Hazard Ratio (RANBP9 high vs low) 1.73, 95% CI 1.15-2.59, p = 0.0084; Overall Survival HR (RANBP9 high vs low) 1.99, 95% CI 1.27-3.11, p = 0.003]. Accordingly, RANBP9 KO cells showed higher sensitivity to cisplatin in comparison with WT controls both in vitro and in vivo models. NSCLC RANBP9 KO cells were also more sensitive than control cells to the PARP inhibitor olaparib alone and in combination with cisplatin, due to defective ATM-dependent and hyper-activated PARP-dependent DDR. The current investigation paves the way to prospective studies to assess the clinical value of RANBP9 protein levels as prognostic and predictive biomarker of response to DDR-targeting drugs, leading to the development of new tools for the management of NSCLC patients.
Collapse
Affiliation(s)
- Anna Tessari
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Kareesma Parbhoo
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Meghan Pawlikowski
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Eliana Rulli
- Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Claudia Foray
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Alessandra Fabbri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valerio Embrione
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Monica Ganzinelli
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Marina Capece
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Moray J Campbell
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, 536 Parks Hall, Columbus, OH, 43210, USA
| | - Massimo Broggini
- Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Krista La Perle
- Department of Veterinary Biosciences and Comparative Pathology and Mouse Phenotyping Shared Resource, College of Veterinary Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabriella Farina
- Department of Oncology, Ospedale Fatebenefratelli and Oftalmico, Milan, Italy
| | - Sara Cole
- Campus Microscopy and Imaging Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Mirko Marabese
- Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Marianna Hernandez
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Joseph M Amann
- Department of Internal Medicine, College of Medicine, James Thoracic Center, Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Giancarlo Pruneri
- Division of Pathology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - David P Carbone
- Department of Internal Medicine, College of Medicine, James Thoracic Center, Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Marina C Garassino
- Thoracic Oncology Unit, Department of Medical Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo M Croce
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Dario Palmieri
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
532
|
Durable response in a woman with recurrent low-grade endometrioid endometrial cancer and a germline BRCA2 mutation treated with a PARP inhibitor. Gynecol Oncol 2018; 150:219-226. [DOI: 10.1016/j.ygyno.2018.05.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
533
|
Kim J, Do KA, Ha MJ, Peterson CB. Bayesian inference of hub nodes across multiple networks. Biometrics 2018; 75:172-182. [PMID: 30051914 DOI: 10.1111/biom.12958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 03/01/2018] [Accepted: 07/01/2018] [Indexed: 11/30/2022]
Abstract
Hub nodes within biological networks play a pivotal role in determining phenotypes and disease outcomes. In the multiple network setting, we are interested in understanding network similarities and differences across different experimental conditions or subtypes of disease. The majority of proposed approaches for joint modeling of multiple networks focus on the sharing of edges across graphs. Rather than assuming the network similarities are driven by individual edges, we instead focus on the presence of common hub nodes, which are more likely to be preserved across settings. Specifically, we formulate a Bayesian approach to the problem of multiple network inference which allows direct inference on shared and differential hub nodes. The proposed method not only allows a more intuitive interpretation of the resulting networks and clearer guidance on potential targets for treatment, but also improves power for identifying the edges of highly connected nodes. Through simulations, we demonstrate the utility of our method and compare its performance to current popular methods that do not borrow information regarding hub nodes across networks. We illustrate the applicability of our method to inference of co-expression networks from The Cancer Genome Atlas ovarian carcinoma dataset.
Collapse
Affiliation(s)
- Junghi Kim
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, U.S.A
| | - Kim-Anh Do
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, U.S.A
| | - Min Jin Ha
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, U.S.A
| | - Christine B Peterson
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, U.S.A
| |
Collapse
|
534
|
Dal Molin GZ, Omatsu K, Sood AK, Coleman RL. Rucaparib in ovarian cancer: an update on safety, efficacy and place in therapy. Ther Adv Med Oncol 2018; 10:1758835918778483. [PMID: 29977351 PMCID: PMC6024342 DOI: 10.1177/1758835918778483] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/25/2018] [Indexed: 12/21/2022] Open
Abstract
Rucaparib is a poly (ADP-ribose) polymerase (PARP) inhibitor and potent inhibitor of PARP1, PARP2 and PARP3 enzymes. Phase II and III trials have documented that rucaparib has single-agent antitumor activity in patients with high-grade ovarian carcinoma, with both BRCA-mutated (germline and somatic) and with homologous recombination deficiency (HRD). Rucaparib as a maintenance treatment showed increased progression-free survival in patients with ovarian carcinoma who achieved a response to platinum-based chemotherapy, with an acceptable safety profile. The approval of this drug, along with the companion diagnostic FoundationFocus CDxBRCA test represents an important new therapeutic option in the treatment of ovarian cancer. This article reviews the mechanisms of action, safety, pharmacokinetics and pharmacodynamics and indications for use of rucaparib as well as future trials.
Collapse
Affiliation(s)
- Graziela Z Dal Molin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kohei Omatsu
- Department of Gynecology, The Cancer Institute Hospital, Tokyo, Japan
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas, MD Anderson Cancer Center, 1155 Herman Pressler Drive, CPB 6.3590, Houston, TX 77030, USA
| |
Collapse
|
535
|
de Jonge MM, Ruano D, van Eijk R, van der Stoep N, Nielsen M, Wijnen JT, Ter Haar NT, Baalbergen A, Bos MEMM, Kagie MJ, Vreeswijk MPG, Gaarenstroom KN, Kroep JR, Smit VTHBM, Bosse T, van Wezel T, van Asperen CJ. Validation and Implementation of BRCA1/2 Variant Screening in Ovarian Tumor Tissue. J Mol Diagn 2018; 20:600-611. [PMID: 29936257 DOI: 10.1016/j.jmoldx.2018.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/01/2018] [Indexed: 12/19/2022] Open
Abstract
BRCA1/2 variant analysis in tumor tissue could streamline the referral of patients with epithelial ovarian, fallopian tube, or primary peritoneal cancer to genetic counselors and select patients who benefit most from targeted treatment. We investigated the sensitivity of BRCA1/2 variant analysis in formalin-fixed, paraffin-embedded tumor tissue using a combination of next-generation sequencing and copy number variant multiplex ligation-dependent probe amplification. After optimization using a training cohort of known BRCA1/2 mutation carriers, validation was performed in a prospective cohort in which screening of BRCA1/2 tumor DNA and leukocyte germline DNA was performed in parallel. BRCA1 promoter hypermethylation and pedigree analysis were also performed. In the training cohort, 45 of 46 germline BRCA1/2 variants were detected (sensitivity, 98%). In the prospective cohort (n = 62), all six germline variants were identified (sensitivity, 100%), together with five somatic BRCA1/2 variants and eight cases with BRCA1 promoter hypermethylation. In four BRCA1/2 variant-negative patients, surveillance or prophylactic management options were offered on the basis of positive family histories. We conclude that BRCA1/2 formalin-fixed, paraffin-embedded tumor tissue analysis reliably detects BRCA1/2 variants. When taking family history of BRCA1/2 variant-negative patients into account, tumor BRCA1/2 variant screening allows more efficient selection of epithelial ovarian cancer patients for genetic counseling and simultaneously selects patients who benefit most from targeted treatment.
Collapse
Affiliation(s)
- Marthe M de Jonge
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dina Ruano
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ronald van Eijk
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nienke van der Stoep
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Maartje Nielsen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Juul T Wijnen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Natalja T Ter Haar
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Astrid Baalbergen
- Department of Gynaecology, Reinier de Graaf Hospital, Delft, the Netherlands
| | - Monique E M M Bos
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marjolein J Kagie
- Department of Gynecology, Haaglanden Medisch Centrum, The Hague, the Netherlands
| | - Maaike P G Vreeswijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Katja N Gaarenstroom
- Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Christi J van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
536
|
Rauch A, Carlstedt A, Emmerich C, Mustafa AHM, Göder A, Knauer SK, Linnebacher M, Heinzel T, Krämer OH. Survivin antagonizes chemotherapy-induced cell death of colorectal cancer cells. Oncotarget 2018; 9:27835-27850. [PMID: 29963241 PMCID: PMC6021236 DOI: 10.18632/oncotarget.25600] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/08/2018] [Indexed: 02/06/2023] Open
Abstract
Irinotecan (CPT-11) and oxaliplatin (L-OHP) are among the most frequently used drugs against colorectal tumors. Therefore, it is important to define the molecular mechanisms that these agents modulate in colon cancer cells. Here we demonstrate that CPT-11 stalls such cells in the G2/M phase of the cell cycle, induces an accumulation of the tumor suppressor p53, the replicative stress/DNA damage marker γH2AX, phosphorylation of the checkpoint kinases ATM and ATR, and an ATR-dependent accumulation of the pro-survival molecule survivin. L-OHP reduces the number of cells in S-phase, stalls cell cycle progression, transiently triggers an accumulation of low levels of γH2AX and phosphorylated checkpoint kinases, and L-OHP suppresses survivin expression at the mRNA and protein levels. Compared to CPT-11, L-OHP is a stronger inducer of caspases and p53-dependent apoptosis. Overexpression and RNAi against survivin reveal that this factor critically antagonizes caspase-dependent apoptosis in cells treated with CPT-11 and L-OHP. We additionally show that L-OHP suppresses survivin through p53 and its downstream target p21, which stalls cell cycle progression as a cyclin-dependent kinase inhibitor (CDKi). These data shed new light on the regulation of survivin by two clinically significant drugs and its biological and predictive relevance in drug-exposed cancer cells.
Collapse
Affiliation(s)
- Anke Rauch
- Center for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University Jena, 07745 Jena, Germany
| | - Annemarie Carlstedt
- Center for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University Jena, 07745 Jena, Germany.,Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
| | - Claudia Emmerich
- Center for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University Jena, 07745 Jena, Germany
| | - Al-Hassan M Mustafa
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Anja Göder
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Shirley K Knauer
- Department of Molecular Biology, Centre for Medical Biotechnology (ZMB), University of Duisburg-Essen, 45141 Essen, Germany
| | - Michael Linnebacher
- Department of General Surgery, Molecular Oncology and Immunotherapy, University of Rostock, 18057 Rostock, Germany
| | - Thorsten Heinzel
- Center for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University Jena, 07745 Jena, Germany
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
537
|
Affiliation(s)
- Kent W. Mouw
- Kent W. Mouw, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA; and Alan D. D’Andrea, Dana-Farber Cancer Institute, Boston, MA
| | - Alan D. D’Andrea
- Kent W. Mouw, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA; and Alan D. D’Andrea, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
538
|
Duma N, Gast KC, Choong GM, Leon-Ferre RA, O'Sullivan CC. Where Do We Stand on the Integration of PARP Inhibitors for the Treatment of Breast Cancer? Curr Oncol Rep 2018; 20:63. [PMID: 29884921 DOI: 10.1007/s11912-018-0709-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW To provide an overview of the clinical development of poly(ADP-ribose) polymerase inhibitors (PARPi) in breast cancer to date and to review existing challenges and future research directions. RECENT FINDINGS We summarize the clinical development of PARPi in breast cancer from bench to bedside, and discuss the results of recent phase 3 trials in patients with metastatic breast cancer (MBC) and germline mutations in BRCA1/2 (gBRCAm). We will also provide an update regarding mechanisms of action and resistance to PARPi, and review clinical trials of PARPi as monotherapy or in combination regimens. PARPi are a novel treatment approach in persons with gBRCA1/2m-associated MBC. Going forward, the clinical applicability of these compounds outside the gBRCAm setting will be studied in greater detail. The identification of accurate predictive biomarkers of response is a research priority.
Collapse
Affiliation(s)
- Narjust Duma
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kelly C Gast
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Grace M Choong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | | | | |
Collapse
|
539
|
Konstantinopoulos PA, Matulonis UA. PARP Inhibitors in Ovarian Cancer: A Trailblazing and Transformative Journey. Clin Cancer Res 2018; 24:4062-4065. [PMID: 29871906 DOI: 10.1158/1078-0432.ccr-18-1314] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 05/24/2018] [Accepted: 06/01/2018] [Indexed: 11/16/2022]
Abstract
PARP inhibitors have transformed treatment for ovarian cancer, a cancer notable for homologous recombination (HR) deficiencies and aberrant DNA repair, especially in the high-grade serous subtype. PARP inhibitors are now approved for recurrent ovarian cancer as maintenance following response to platinum chemotherapy and BRCA-mutated (BRCAm) cancer treatment. Clin Cancer Res; 24(17); 4062-5. ©2018 AACRSee related article by Ison et al., p. 4066.
Collapse
Affiliation(s)
| | - Ursula A Matulonis
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston Massachusetts.
| |
Collapse
|
540
|
Tumiati M, Hietanen S, Hynninen J, Pietilä E, Färkkilä A, Kaipio K, Roering P, Huhtinen K, Alkodsi A, Li Y, Lehtonen R, Erkan EP, Tuominen MM, Lehti K, Hautaniemi SK, Vähärautio A, Grénman S, Carpén O, Kauppi L. A Functional Homologous Recombination Assay Predicts Primary Chemotherapy Response and Long-Term Survival in Ovarian Cancer Patients. Clin Cancer Res 2018; 24:4482-4493. [PMID: 29858219 DOI: 10.1158/1078-0432.ccr-17-3770] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/25/2018] [Accepted: 05/25/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Homologous recombination deficiency (HRD) correlates with platinum sensitivity in patients with ovarian cancer, which clinically is the most useful predictor of sensitivity to PARPi. To date, there are no reliable diagnostic tools to anticipate response to platinum-based chemotherapy, thus we aimed to develop an ex vivo functional HRD detection test that could predict both platinum-sensitivity and patient eligibility to targeted drug treatments.Experimental Design: We obtained a functional HR score by quantifying homologous recombination (HR) repair after ionizing radiation-induced DNA damage in primary ovarian cancer samples (n = 32). Samples clustered in 3 categories: HR-deficient, HR-low, and HR-proficient. We analyzed the HR score association with platinum sensitivity and treatment response, platinum-free interval (PFI) and overall survival (OS), and compared it with other clinical parameters. In parallel, we performed DNA-sequencing of HR genes to assess if functional HRD can be predicted by currently offered genetic screening.Results: Low HR scores predicted primary platinum sensitivity with high statistical significance (P = 0.0103), associated with longer PFI (HR-deficient vs. HR-proficient: 531 vs. 53 days), and significantly correlated with improved OS (HR score <35 vs. ≥35, hazard ratio = 0.08, P = 0.0116). At the genomic level, we identified a few unclear mutations in HR genes and the mutational signature associated with HRD, but, overall, genetic screening failed to predict functional HRD.Conclusions: We developed an ex vivo assay that detects tumor functional HRD and an HR score able to predict platinum sensitivity, which holds the clinically relevant potential to become the routine companion diagnostic in the management of patients with ovarian cancer. Clin Cancer Res; 24(18); 4482-93. ©2018 AACR.
Collapse
Affiliation(s)
- Manuela Tumiati
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland.
| | - Sakari Hietanen
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku, Finland
| | - Johanna Hynninen
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku, Finland
| | - Elina Pietilä
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Anniina Färkkilä
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Katja Kaipio
- Department of Pathology, University of Turku and Turku University Hospital, Finland
| | - Pia Roering
- Department of Pathology, University of Turku and Turku University Hospital, Finland
| | - Kaisa Huhtinen
- Department of Pathology, University of Turku and Turku University Hospital, Finland
| | - Amjad Alkodsi
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Yilin Li
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Rainer Lehtonen
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Erdogan Pekcan Erkan
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Minna M Tuominen
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Kaisa Lehti
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sampsa K Hautaniemi
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Anna Vähärautio
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Seija Grénman
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku, Finland
| | - Olli Carpén
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Pathology, University of Turku and Turku University Hospital, Finland
| | - Liisa Kauppi
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
541
|
Ricci F, Affatato R, Carrassa L, Damia G. Recent Insights into Mucinous Ovarian Carcinoma. Int J Mol Sci 2018; 19:ijms19061569. [PMID: 29795040 PMCID: PMC6032258 DOI: 10.3390/ijms19061569] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/11/2018] [Accepted: 05/18/2018] [Indexed: 12/31/2022] Open
Abstract
Ovarian mucinous tumors represent a group of rare neoplasms with a still undefined cell of origin but with an apparent progression from benign to borderline to carcinoma. Even though these tumors are different from the other histological subtypes of epithelial ovarian neoplasms, they are still treated with a similar chemotherapeutic approach. Here, we review its pathogenesis, molecular alterations, (differential) diagnosis, clinical presentation and current treatment, and how recent molecular and biological information on this tumor might lead to better and more specific clinical management of patients with mucinous ovarian carcinoma.
Collapse
Affiliation(s)
- Francesca Ricci
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Laboratory of Molecular Pharmacology, Via Giuseppe La Masa 19, 20156 Milan, Italy.
| | - Roberta Affatato
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Laboratory of Molecular Pharmacology, Via Giuseppe La Masa 19, 20156 Milan, Italy.
| | - Laura Carrassa
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Laboratory of Molecular Pharmacology, Via Giuseppe La Masa 19, 20156 Milan, Italy.
| | - Giovanna Damia
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Laboratory of Molecular Pharmacology, Via Giuseppe La Masa 19, 20156 Milan, Italy.
| |
Collapse
|
542
|
Clifford C, Vitkin N, Nersesian S, Reid-Schachter G, Francis JA, Koti M. Multi-omics in high-grade serous ovarian cancer: Biomarkers from genome to the immunome. Am J Reprod Immunol 2018; 80:e12975. [DOI: 10.1111/aji.12975] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/16/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Cole Clifford
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
| | - Natasha Vitkin
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
- Cancer Biology and Genetics; Queen's Cancer Research Institute; Queen's University; Kingston ON Canada
| | - Sarah Nersesian
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
- Cancer Biology and Genetics; Queen's Cancer Research Institute; Queen's University; Kingston ON Canada
| | | | - Julie-Ann Francis
- Department of Obstetrics and Gynecology; Kingston Health Sciences Center; Queen's University; Kingston ON Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
- Cancer Biology and Genetics; Queen's Cancer Research Institute; Queen's University; Kingston ON Canada
- Department of Obstetrics and Gynecology; Kingston Health Sciences Center; Queen's University; Kingston ON Canada
| |
Collapse
|
543
|
Longoria TC, Tewari KS. Pharmacokinetic drug evaluation of niraparib for the treatment of ovarian cancer. Expert Opin Drug Metab Toxicol 2018; 14:543-550. [DOI: 10.1080/17425255.2018.1461838] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
544
|
Guffanti F, Fratelli M, Ganzinelli M, Bolis M, Ricci F, Bizzaro F, Chilà R, Sina FP, Fruscio R, Lupia M, Cavallaro U, Cappelletti MR, Generali D, Giavazzi R, Damia G. Platinum sensitivity and DNA repair in a recently established panel of patient-derived ovarian carcinoma xenografts. Oncotarget 2018; 9:24707-24717. [PMID: 29872499 PMCID: PMC5973859 DOI: 10.18632/oncotarget.25185] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/05/2018] [Indexed: 01/22/2023] Open
Abstract
A xenobank of patient-derived (PDX) ovarian tumor samples has been established consisting of tumors with different sensitivity to cisplatin (DDP), from very responsive to resistant. As the DNA repair pathway is an important driver in tumor response to DDP, we analyzed the mRNA expression of 20 genes involved in the nucleotide excision repair, fanconi anemia, homologous recombination, base excision repair, mismatch repair and translesion repair pathways and the methylation patterns of some of these genes. We also investigated the correlation with the response to platinum-based therapy. The mRNA levels of the selected genes were evaluated by Real Time-PCR (RT-PCR) with ad hoc validated primers and gene promoter methylation by pyrosequencing. All the DNA repair genes were variably expressed in all 42 PDX samples analyzed, with no particular histotype-specific pattern of expression. In high-grade serous/endometrioid PDXs, the CDK12 mRNA expression levels positively correlated with the expression of TP53BP1, PALB2, XPF and POLB. High-grade serous/endometrioid PDXs with TP53 mutations had significantly higher levels of POLQ, FANCD2, RAD51 and POLB than high-grade TP53 wild type PDXs. The mRNA levels of CDK12, PALB2 and XPF inversely associated with the in vivo DDP antitumor activity; higher CDK12 mRNA levels were associated with a higher recurrence rate in ovarian patients with low residual tumor. These data support the important role of CDK12 in the response to a platinum based therapy in ovarian patients.
Collapse
Affiliation(s)
- Federica Guffanti
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Maddalena Fratelli
- Department of Biochemistry, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Monica Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Bolis
- Department of Biochemistry, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Francesca Ricci
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Francesca Bizzaro
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Rosaria Chilà
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Federica Paola Sina
- Clinic of Obstetrics and Gynecology, San Gerardo Hospital, University of Milan-Bicocca, Department of Medicine and Surgery, Milan, Italy
| | - Robert Fruscio
- Clinic of Obstetrics and Gynecology, San Gerardo Hospital, University of Milan-Bicocca, Department of Medicine and Surgery, Milan, Italy
| | - Michela Lupia
- Unit of Gynecological Oncology Research, European Institute of Oncology, Milan, Italy
| | - Ugo Cavallaro
- Unit of Gynecological Oncology Research, European Institute of Oncology, Milan, Italy
| | | | - Daniele Generali
- Breast Cancer Unit and Translational Research Unit, ASST Cremona, Cremona, Italy.,Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Raffaella Giavazzi
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Giovanna Damia
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
545
|
Fleury H, Carmona E, Morin VG, Meunier L, Masson JY, Tonin PN, Provencher D, Mes-Masson AM. Cumulative defects in DNA repair pathways drive the PARP inhibitor response in high-grade serous epithelial ovarian cancer cell lines. Oncotarget 2018; 8:40152-40168. [PMID: 27374179 PMCID: PMC5522225 DOI: 10.18632/oncotarget.10308] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/09/2016] [Indexed: 01/17/2023] Open
Abstract
PARP inhibitors (PARPi), such as Olaparib, have shown promising results in high-grade serous (HGS) epithelial ovarian cancer (EOC) treatment. PARPi sensitivity has been mainly associated with homologous recombination (HR) deficiency, but clinical trials have shown that predicting actual patient response is complex. Here, we investigated gene expression microarray, HR functionality and Olaparib sensitivity of 18 different HGS EOC cell lines and demonstrate that PARPi sensitivity is not only associated with HR defects. Gene target validation show that down regulation of genes in the nucleotide excision repair (NER) and mismatch repair (MMR) pathways (ERCC8 and MLH1, respectively) increases PARPi response. The highest sensitivity was observed when genes in both the HR and either NER or MMR pathways were concomitantly down regulated. Using clinical samples, patients with these concurrent down regulations could be identified. Based on these results, a novel model to predict PARPi sensitivity is herein proposed. This model implies that the extreme responders identified in clinical trials have deficiencies in HR and either NER or MMR.
Collapse
Affiliation(s)
- Hubert Fleury
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Euridice Carmona
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Vincent G Morin
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Liliane Meunier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU Research Center, Québec City, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University Cancer Research Center, Québec City, Canada
| | - Patricia N Tonin
- Cancer Research Program (CRP), The Research Institute of the McGill University Health Centre, Montreal, Canada.,Department of Human Genetics, McGill University, Montreal, Canada.,Department of Medicine, McGill University, Montreal, Canada
| | - Diane Provencher
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Division of Gynecologic Oncology, Université de Montréal, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Department of Medicine, Université de Montréal, Montreal, Canada
| |
Collapse
|
546
|
Chao A, Lai CH, Wang TH, Jung SM, Lee YS, Chang WY, Yang LY, Ku FC, Huang HJ, Chao AS, Wang CJ, Chang TC, Wu RC. Genomic scar signatures associated with homologous recombination deficiency predict adverse clinical outcomes in patients with ovarian clear cell carcinoma. J Mol Med (Berl) 2018; 96:527-536. [DOI: 10.1007/s00109-018-1643-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/31/2018] [Accepted: 04/23/2018] [Indexed: 01/19/2023]
|
547
|
Cruz C, Castroviejo-Bermejo M, Gutiérrez-Enríquez S, Llop-Guevara A, Ibrahim YH, Gris-Oliver A, Bonache S, Morancho B, Bruna A, Rueda OM, Lai Z, Polanska UM, Jones GN, Kristel P, de Bustos L, Guzman M, Rodríguez O, Grueso J, Montalban G, Caratú G, Mancuso F, Fasani R, Jiménez J, Howat WJ, Dougherty B, Vivancos A, Nuciforo P, Serres-Créixams X, Rubio IT, Oaknin A, Cadogan E, Barrett JC, Caldas C, Baselga J, Saura C, Cortés J, Arribas J, Jonkers J, Díez O, O'Connor MJ, Balmaña J, Serra V. RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer. Ann Oncol 2018; 29:1203-1210. [PMID: 29635390 PMCID: PMC5961353 DOI: 10.1093/annonc/mdy099] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background BRCA1 and BRCA2 (BRCA1/2)-deficient tumors display impaired homologous recombination repair (HRR) and enhanced sensitivity to DNA damaging agents or to poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi). Their efficacy in germline BRCA1/2 (gBRCA1/2)-mutated metastatic breast cancers has been recently confirmed in clinical trials. Numerous mechanisms of PARPi resistance have been described, whose clinical relevance in gBRCA-mutated breast cancer is unknown. This highlights the need to identify functional biomarkers to better predict PARPi sensitivity. Patients and methods We investigated the in vivo mechanisms of PARPi resistance in gBRCA1 patient-derived tumor xenografts (PDXs) exhibiting differential response to PARPi. Analysis included exome sequencing and immunostaining of DNA damage response proteins to functionally evaluate HRR. Findings were validated in a retrospective sample set from gBRCA1/2-cancer patients treated with PARPi. Results RAD51 nuclear foci, a surrogate marker of HRR functionality, were the only common feature in PDX and patient samples with primary or acquired PARPi resistance. Consistently, low RAD51 was associated with objective response to PARPi. Evaluation of the RAD51 biomarker in untreated tumors was feasible due to endogenous DNA damage. In PARPi-resistant gBRCA1 PDXs, genetic analysis found no in-frame secondary mutations, but BRCA1 hypomorphic proteins in 60% of the models, TP53BP1-loss in 20% and RAD51-amplification in one sample, none mutually exclusive. Conversely, one of three PARPi-resistant gBRCA2 tumors displayed BRCA2 restoration by exome sequencing. In PDXs, PARPi resistance could be reverted upon combination of a PARPi with an ataxia-telangiectasia mutated (ATM) inhibitor. Conclusion Detection of RAD51 foci in gBRCA tumors correlates with PARPi resistance regardless of the underlying mechanism restoring HRR function. This is a promising biomarker to be used in the clinic to better select patients for PARPi therapy. Our study also supports the clinical development of PARPi combinations such as those with ATM inhibitors.
Collapse
Affiliation(s)
- C Cruz
- Experimental Therapeutics Group; High Risk and Familial Cancer, Vall d'Hebron Institute of Oncology, Barcelona; Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona
| | | | | | | | | | | | | | - B Morancho
- Growth Factors Laboratory, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - A Bruna
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge
| | - O M Rueda
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge
| | - Z Lai
- AstraZeneca, Gatehouse Park, Waltham, USA
| | - U M Polanska
- DNA Damage Response Biology Area, Oncology iMed, AstraZeneca, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - G N Jones
- DNA Damage Response Biology Area, Oncology iMed, AstraZeneca, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - P Kristel
- Division of Molecular Pathology and Cancer Genomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | - R Fasani
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology, Barcelona
| | - J Jiménez
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology, Barcelona
| | - W J Howat
- DNA Damage Response Biology Area, Oncology iMed, AstraZeneca, Cancer Research UK Cambridge Institute, Cambridge, UK
| | | | | | - P Nuciforo
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology, Barcelona
| | | | - I T Rubio
- Breast Surgical Unit, Breast Cancer Center, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona
| | - A Oaknin
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona; Gynecological Malignancies Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - E Cadogan
- DNA Damage Response Biology Area, Oncology iMed, AstraZeneca, Cancer Research UK Cambridge Institute, Cambridge, UK
| | | | - C Caldas
- Department of Oncology and Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cambridge Breast Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre at Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - J Baselga
- Human Oncology and Pathogenesis Program (HOPP); Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - C Saura
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona; Breast Cancer and Melanoma Group, Vall d'Hebron Institute of Oncology, Barcelona
| | - J Cortés
- Ramón y Cajal University Hospital, Madrid; Vall d'Hebron Institute of Oncology, Barcelona
| | - J Arribas
- Growth Factors Laboratory, Vall d'Hebron Institute of Oncology, Barcelona, Spain; Department of Biochemistry and Molecular Biology, Building M, Campus UAB, Bellaterra (Cerdanyola del Vallès); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona; CIBERONC, Barcelona
| | | | - O Díez
- Oncogenetics Group; Clinical and Molecular Genetics Area, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M J O'Connor
- DNA Damage Response Biology Area, Oncology Innovative Medicine and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - J Balmaña
- High Risk and Familial Cancer, Vall d'Hebron Institute of Oncology, Barcelona; Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona
| | - V Serra
- Experimental Therapeutics Group; CIBERONC, Barcelona.
| |
Collapse
|
548
|
Makvandi M, Pantel A, Schwartz L, Schubert E, Xu K, Hsieh CJ, Hou C, Kim H, Weng CC, Winters H, Doot R, Farwell MD, Pryma DA, Greenberg RA, Mankoff DA, Simpkins F, Mach RH, Lin LL. A PET imaging agent for evaluating PARP-1 expression in ovarian cancer. J Clin Invest 2018; 128:2116-2126. [PMID: 29509546 PMCID: PMC5919879 DOI: 10.1172/jci97992] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/28/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Poly(ADP-ribose) polymerase (PARP) inhibitors are effective in a broad population of patients with ovarian cancer; however, resistance caused by low enzyme expression of the drug target PARP-1 remains to be clinically evaluated in this context. We hypothesize that PARP-1 expression is variable in ovarian cancer and can be quantified in primary and metastatic disease using a novel PET imaging agent. METHODS We used a translational approach to describe the significance of PET imaging of PARP-1 in ovarian cancer. First, we produced PARP1-KO ovarian cancer cell lines using CRISPR/Cas9 gene editing to test the loss of PARP-1 as a resistance mechanism to all clinically used PARP inhibitors. Next, we performed preclinical microPET imaging studies using ovarian cancer patient-derived xenografts in mouse models. Finally, in a phase I PET imaging clinical trial we explored PET imaging as a regional marker of PARP-1 expression in primary and metastatic disease through correlative tissue histology. RESULTS We found that deletion of PARP1 causes resistance to all PARP inhibitors in vitro, and microPET imaging provides proof of concept as an approach to quantify PARP-1 in vivo. Clinically, we observed a spectrum of standard uptake values (SUVs) ranging from 2-12 for PARP-1 in tumors. In addition, we found a positive correlation between PET SUVs and fluorescent immunohistochemistry for PARP-1 (r2 = 0.60). CONCLUSION This work confirms the translational potential of a PARP-1 PET imaging agent and supports future clinical trials to test PARP-1 expression as a method to stratify patients for PARP inhibitor therapy. TRIAL REGISTRATION Clinicaltrials.gov NCT02637934. FUNDING Research reported in this publication was supported by the Department of Defense OC160269, a Basser Center team science grant, NIH National Cancer Institute R01CA174904, a Department of Energy training grant DE-SC0012476, Abramson Cancer Center Radiation Oncology pilot grants, the Marsha Rivkin Foundation, Kaleidoscope of Hope Foundation, and Paul Calabresi K12 Career Development Award 5K12CA076931.
Collapse
Affiliation(s)
- Mehran Makvandi
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Austin Pantel
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lauren Schwartz
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erin Schubert
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kuiying Xu
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Chia-Ju Hsieh
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Catherine Hou
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hyoung Kim
- Department of OBGYN, Division of Gynecology and Oncology
| | - Chi-Chang Weng
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Robert Doot
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael D. Farwell
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Daniel A. Pryma
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - David A. Mankoff
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Fiona Simpkins
- Department of OBGYN, Division of Gynecology and Oncology
| | - Robert H. Mach
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lilie L. Lin
- Department of Radiation Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
549
|
Hou D, Liu Z, Xu X, Liu Q, Zhang X, Kong B, Wei JJ, Gong Y, Shao C. Increased oxidative stress mediates the antitumor effect of PARP inhibition in ovarian cancer. Redox Biol 2018; 17:99-111. [PMID: 29684820 PMCID: PMC6006521 DOI: 10.1016/j.redox.2018.03.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 01/09/2023] Open
Abstract
PARP inhibitors have been widely tested in clinical trials, especially for the treatment of breast cancer and ovarian cancer, and were shown to be highly successful. Because PARP primarily functions in sensing and repairing DNA strand breaks, the therapeutic effect of PARP inhibition is generally believed to be attributed to impaired DNA repair. We here report that oxidative stress is also increased by PARP inhibition and mediates the antitumor effect. We showed that PARP1 is highly expressed in specimens of high grade serous ovarian carcinoma and its activity is required for unperturbed proliferation of ovarian cancer cells. Inhibition or depletion of PARP leads to not only an increase in DNA damage, but also an elevation in the levels of reactive oxygen species (ROS). Importantly, antioxidant N-acetylcysteine (NAC) significantly attenuated the induction of DNA damage and the perturbation of proliferation by PARP inhibition or depletion. We further showed that NADPH oxidases 1 and 4 were significantly upregulated by PARP inhibition and were partially responsible for the induction of oxidative stress. Depletion of NOX1 and NOX4 partially rescued the growth inhibition of PARP1-deficient tumor xenografts. Our findings suggest that in addition to compromising the repair of DNA damage, PARP inhibition or depletion may exert extra antitumor effect by elevating oxidative stress in ovarian cancer cells. PARP1 is overexpressed in ovarian cancer. PARP inhibition increases oxidative stress and oxidative DNA damage. PARP inhibition increases ROS by upregulating NOX1 and NOX4. Oxidative stress mediates the antitumor effect of PARP inhibition.
Collapse
Affiliation(s)
- Dong Hou
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Zhaojian Liu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xiuhua Xu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Qiao Liu
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xiyu Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jian-Jun Wei
- Department of Pathology, Northwestern University School of Medicine, Chicago, IL, USA
| | - Yaoqin Gong
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Changshun Shao
- Key Laboratory of Experimental Teratology, Ministry of Education/Department of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China; The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Ren Ai Road, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
550
|
From checkpoint to checkpoint: DNA damage ATR/Chk1 checkpoint signalling elicits PD-L1 immune checkpoint activation. Br J Cancer 2018. [PMID: 29531322 PMCID: PMC5931110 DOI: 10.1038/s41416-018-0017-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Multiple clinical studies have revealed a link between genomic instability and response to anti-PD-1/PD-L1 therapy in cancer management. A recent study has revealed an important role for the ATR/Chk1 DNA damage checkpoint in regulating PD-L1 expression, raising important clinical and translational questions for therapy selection and study design.
Collapse
|