1
|
Gaillard S, Verma N, Berg M, Harrison J, Huang P, Leatherman JM, Doucet M, Sen R, Suru A, Cai H, Durham J, Jelovac D, Cimino-Mathews A, Cherry C, Ganguly S, Emens LA. A clinical study of tremelimumab, alone or in combination with olaparib, for recurrent epithelial ovarian cancer. Gynecol Oncol 2025; 194:41-47. [PMID: 39951918 PMCID: PMC11993342 DOI: 10.1016/j.ygyno.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE PARP inhibitors may work synergistically to improve the efficacy of immunotherapy in patients with epithelial ovarian cancer (EOC). We performed a parallel-arm study of tremelimumab, alone or with olaparib, in patients with recurrent EOC. METHODS Eligibility criteria included measurable disease and progression <12 months from last platinum. Participants were randomized to Arm A (tremelimumab monotherapy, 10 mg/kg/dose intravenously [IV]) or Arm B (dose level 1 [DL1] olaparib orally 150 mg twice daily with tremelimumab IV 3 mg/kg/dose and DL2 olaparib orally 150 mg twice daily with tremelimumab IV 10 mg/kg/dose). Primary objectives were safety, change in peripheral ICOS+ T cells, and identification of optimal dose combination. RESULTS Among 24 total patients (12 on Arm A, 6 on Arm B-DL1, 6 on Arm B-DL2), the most common grade 3 toxicities were rash (13 %), immune-mediated hepatitis (8 %), and colitis (8 %). No grade ≥ 4 toxicities were identified. No dose-limiting toxicities were identified. One patient (Arm B-DL2) experienced a partial response; no complete responses were observed. Ten patients (7 on Arm A, 2 on Arm B-DL2, and 1 on Arm B-DL1) had a best response of stable disease. There was a significant increase in CD4+ICOS+ and CD8+ICOS+ T cells at both C1D15 and C1D22 in groups treated with tremelimumab IV 10 mg/kg/dose, but not in those treated with tremelimumab 3 mg/kg/dose. CONCLUSIONS Tremelimumab IV 10 mg/kg/dose with olaparib 150 mg orally twice daily was safe and feasible. Tremelimumab 10 mg/kg/dose (as opposed to 3 mg/kg/dose) was required for immune activation, although this did not translate into clinical responses.
Collapse
Affiliation(s)
- Stéphanie Gaillard
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America; Department of Gynecology and Obstetrics Division of Gynecologic Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America.
| | - Neha Verma
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Maureen Berg
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Jeanne Harrison
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Peng Huang
- Department of Oncology Division of Biostatistics and Bioinformatics, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - James M Leatherman
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Michele Doucet
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Rupashree Sen
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Aditya Suru
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Hongyan Cai
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Jennifer Durham
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Danijela Jelovac
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Ashley Cimino-Mathews
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Christopher Cherry
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Sudipto Ganguly
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Leisha A Emens
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
2
|
Piffoux M, Leary A, Follana P, Abdeddaim C, Joly F, Bin S, Bonjour M, Boulai A, Callens C, Villeneuve L, Alexandre M, Schwiertz V, Freyer G, Rodrigues M, You B. Olaparib combined to metronomic cyclophosphamide and metformin in women with recurrent advanced/metastatic endometrial cancer: the ENDOLA phase I/II trial. Nat Commun 2025; 16:1821. [PMID: 39979249 PMCID: PMC11842746 DOI: 10.1038/s41467-025-56914-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Endometrial cancers are characterized by frequent alterations in the PI3K-AKT-mTor, IGF1 and DNA repair signaling pathways. Concomitant inhibition of these pathways was warranted. ENDOLA phase I/II trial (NCT02755844) was designed to assess the safety/efficacy of the triplet combination of the PARP inhibitor olaparib, metronomic cyclophosphamide (50 mg daily), and PI3K-AKT-mTor inhibitor metformin (1500 mg daily) in women with recurrent endometrial carcinomas. Olaparib dose-escalation (100-300 mg twice-a-day (bid)) was used to determine the recommended-phase II-trial-dose (RP2D, primary endpoint), followed by an expansion cohort to determine the non-progression rate at 10 weeks (NPR-10w, secondary endpoint). 31 patients were treated. Olaparib RP2D was defined as 300 mg bid. The tolerability was acceptable, and grade 3-4 adverse events (51% patients) were mainly hematological. The NPR-10w was 61.5%, and the median progression-free survival (mPFS) was 5.2 months. In a post-hoc analysis, when explored by molecular subtypes/alterations, longer PFS were observed in patients with tumors characterized by a non-specific-molecular-profile (NSMP, n = 4; mPFS, 9.1 months), and by both TP53 altered & high number of large genomic alterations (LGA ≥ 8)(n = 10, mPFS, 8.6 months)). The analyses about kinetics of circulating biomarkers and pharmacodynamic effects are not reported here. In total, the benefit/toxicity ratio of the all-oral olaparib/cyclophosphamide/metformin regimen was favorable in heavily pretreated patients with recurrent endometrial cancer.
Collapse
Affiliation(s)
- Max Piffoux
- Medical Oncology, Hospices Civils de Lyon, EPSILYON, Lyon, France; GINECO, Paris, France
| | - Alexandra Leary
- Medical Oncology, Institut Gustave Roussy, Villejuif, France; GINECO, Paris, France
| | | | | | - Florence Joly
- Centre Francois Baclesse, Caen, France; GINECO, Paris, France
| | - Sylvie Bin
- Pôle Santé Publique, Hospices Civils de Lyon, Lyon, France
| | - Maxime Bonjour
- Pôle Santé Publique, Hospices Civils de Lyon, Lyon, France
| | - Anais Boulai
- Genetics Department, Institut Curie and Paris Sciences Lettres University, Paris, France
| | - Celine Callens
- Genetics Department, Institut Curie and Paris Sciences Lettres University, Paris, France
| | | | | | | | - Gilles Freyer
- Medical Oncology, Hospices Civils de Lyon, EPSILYON, Lyon, France; GINECO, Paris, France
| | - Manuel Rodrigues
- Medical Oncology, Institut Curie, Paris, France
- INSERM U830, Institut Curie, Paris, France; GINECO, Paris, France
| | - Benoit You
- Medical Oncology, Hospices Civils de Lyon, EPSILYON, Lyon, France; GINECO, Paris, France.
| |
Collapse
|
3
|
Zhao T, Dong X, Zhao T, Han Z. Next-generation sequencing uncovers crucial mutated genes and potential therapeutic targets in ovarian cancer patients. Am J Transl Res 2024; 16:5990-6007. [PMID: 39544793 PMCID: PMC11558429 DOI: 10.62347/xngv7396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/23/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVES Ovarian cancer is a highly lethal gynecological malignancy, often diagnosed late, resulting in high mortality. While BRCA1 and BRCA2 mutations are known risk factors, the broader genetic landscape needs comprehensive profiling to identify additional diagnostic markers or therapeutic targets. The current study aims to explore the genetic landscape of various cancer-susceptible genes in ovarian cancer patients. METHODS The genetic landscape of ovarian cancer was investigated by analyzing 27 genes via next-generation sequencing (NGS) in 50 ovarian cancer patients. RESULTS Mutations were detected in four genes: Breast Cancer 1 (BRCA1) (62%), Cyclin-Dependent Kinase 4 (CDK4) (58%), MutS Homolog 2 (MSH2) (48%), and Phosphatase and Tensin Homolog (PTEN) (22%). Pathogenic mutations were identified in BRCA1 (p.Tyr1853Ter and p.Gln1848Ter), CDK4 (p.Arg24His), and PTEN (p.Tyr29Ter), occurring in 11 patients. Interestingly, these pathogenic mutations were absent in The Cancer Genome Atlas (TCGA) dataset and the gnomAD for the Asian population, suggesting their unique presence in the Pakistani cohort. Functional assays revealed that these mutations significantly reduced the mRNA and protein expression levels of BRCA1, CDK4, and PTEN, as demonstrated by Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR) and Immunohistochemistry (IHC) analyses. Receiver Operating Characteristic (ROC) curve analysis confirmed the potential of these genes as biomarkers, with downregulated expression accurately distinguishing between normal and cancerous tissues. Structural validation of mutated proteins using Ramachandran plots and Protein Structure Analysis (ProSA-web) analysis confirmed the stability of the mutations. Drug prediction and molecular docking identified Resveratrol as a potential therapeutic agent, indicating strong binding affinities with BRCA1, CDK4, and PTEN proteins. CONCLUSION These findings provide novel insights into the genetic underpinnings of ovarian cancer in the Pakistani population and suggest potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Tianjiao Zhao
- Clinical School, Sanquan College of Xinxiang Medical UniversityXinxiang 453003, Henan, China
| | - Xinghe Dong
- Xunxian Weixi HospitalHebi 458000, Henan, China
| | - Tianshi Zhao
- Department of Basic Medicine, Zhengzhou Health Vocational CollegeZhengzhou 450000, Henan, China
| | - Zhenghua Han
- Clinical School, Sanquan College of Xinxiang Medical UniversityXinxiang 453003, Henan, China
| |
Collapse
|
4
|
Orlikova-Boyer B, Lorant A, Gajulapalli SR, Cerella C, Schnekenburger M, Lee JY, Paik JY, Lee Y, Siegel D, Ross D, Han BW, Nguyen TKY, Christov C, Kang HJ, Dicato M, Diederich M. Antileukemic potential of methylated indolequinone MAC681 through immunogenic necroptosis and PARP1 degradation. Biomark Res 2024; 12:47. [PMID: 38704604 PMCID: PMC11069214 DOI: 10.1186/s40364-024-00594-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Despite advancements in chronic myeloid leukemia (CML) therapy with tyrosine kinase inhibitors (TKIs), resistance and intolerance remain significant challenges. Leukemia stem cells (LSCs) and TKI-resistant cells rely on altered mitochondrial metabolism and oxidative phosphorylation. Targeting rewired energy metabolism and inducing non-apoptotic cell death, along with the release of damage-associated molecular patterns (DAMPs), can enhance therapeutic strategies and immunogenic therapies against CML and prevent the emergence of TKI-resistant cells and LSC persistence. METHODS Transcriptomic analysis was conducted using datasets of CML patients' stem cells and healthy cells. DNA damage was evaluated by fluorescent microscopy and flow cytometry. Cell death was assessed by trypan blue exclusion test, fluorescent microscopy, flow cytometry, colony formation assay, and in vivo Zebrafish xenografts. Energy metabolism was determined by measuring NAD+ and NADH levels, ATP production rate by Seahorse analyzer, and intracellular ATP content. Mitochondrial fitness was estimated by measurements of mitochondrial membrane potential, ROS, and calcium accumulation by flow cytometry, and morphology was visualized by TEM. Bioinformatic analysis, real-time qPCR, western blotting, chemical reaction prediction, and molecular docking were utilized to identify the drug target. The immunogenic potential was assessed by high mobility group box (HMGB)1 ELISA assay, luciferase-based extracellular ATP assay, ectopic calreticulin expression by flow cytometry, and validated by phagocytosis assay, and in vivo vaccination assay using syngeneic C57BL/6 mice. RESULTS Transcriptomic analysis identified metabolic alterations and DNA repair deficiency signatures in CML patients. CML patients exhibited enrichment in immune system, DNA repair, and metabolic pathways. The gene signature associated with BRCA mutated tumors was enriched in CML datasets, suggesting a deficiency in double-strand break repair pathways. Additionally, poly(ADP-ribose) polymerase (PARP)1 was significantly upregulated in CML patients' stem cells compared to healthy counterparts. Consistent with the CML patient DNA repair signature, treatment with the methylated indolequinone MAC681 induced DNA damage, mitochondrial dysfunction, calcium homeostasis disruption, metabolic catastrophe, and necroptotic-like cell death. In parallel, MAC681 led to PARP1 degradation that was prevented by 3-aminobenzamide. MAC681-treated myeloid leukemia cells released DAMPs and demonstrated the potential to generate an immunogenic vaccine in C57BL/6 mice. MAC681 and asciminib exhibited synergistic effects in killing both imatinib-sensitive and -resistant CML, opening new therapeutic opportunities. CONCLUSIONS Overall, increasing the tumor mutational burden by PARP1 degradation and mitochondrial deregulation makes CML suitable for immunotherapy.
Collapse
Affiliation(s)
- Barbora Orlikova-Boyer
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Anne Lorant
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Sruthi Reddy Gajulapalli
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Jin-Young Lee
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
- Present address: Department of Biological Sciences, Keimyung University, Daegu, 42601, Republic of Korea
| | - Ji Yeon Paik
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Yejin Lee
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - David Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - David Ross
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Byung Woo Han
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Thi Kim Yen Nguyen
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | | | - Hyoung Jin Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Cancer Research Institute, Seoul National University Children's Hospital, Seoul, 03080, Republic of Korea
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Marc Diederich
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
5
|
Wei Y, He L, Liu T, Guo T, Xie C, Jia J, Lin Y, Liu J, Fan J. Efficacy and safety of PARP inhibitors combined with antiangiogenic agents in the maintenance treatment of ovarian cancer: a systematic review and meta-analysis with trial sequential analysis of randomized controlled trials. Front Pharmacol 2024; 15:1372077. [PMID: 38584601 PMCID: PMC10995238 DOI: 10.3389/fphar.2024.1372077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/29/2024] [Indexed: 04/09/2024] Open
Abstract
Background: Poly (ADP-ribose) polymerase (PARP) inhibitor and antiangiogenic agent monotherapy have shown to be effective as maintenance treatment in patients with ovarian cancer (OC). However, there is currently a lack of evidence-based study to directly compare the effects of combination therapy with these two drugs. Therefore, this study aimed to compare the efficacy and safety of combination therapy with PARP inhibitors and antiangiogenic agents in women with OC using a meta-analysis. Methods: An exhaustive search of literature was undertaken using multiple databases, including PubMed, Web of Science, Embase, and the Cochrane Library to identify pertinent randomized controlled trials (RCTs) published up until 17 December 2023. The data on progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were pooled. We computed the pooled hazard ratios (HRs) and their 95% confidence intervals (CIs) for PFS and OS, along with the relative risks (RRs) and 95% CIs for AEs. Trial sequential analysis, heterogeneity test, sensitivity analysis, and publication bias assessment were performed. Stata 12.0 and Software R 4.3.1 were utilized for all analyses. Results: This meta-analysis included 7 RCTs with a total of 3,388 participants. The overall analysis revealed that combination therapy of PARP inhibitors and antiangiogenic agents significantly improved PFS (HR = 0.615, 95% CI = 0.517-0.731; 95% PI = 0.379-0.999), but also increased the risk of AEs, including urinary tract infection (RR = 1.500, 95% CI = 1.114-2.021; 95% PI = 0.218-10.346), fatigue (RR = 1.264, 95% CI = 1.141-1.400; 95% PI = 1.012-1.552), headache (RR = 1.868, 95% CI = 1.036-3.369; 95% PI = 0.154-22.642), anorexia (RR = 1.718, 95% CI = 1.320-2.235; 95% PI = 0.050-65.480), and hypertension (RR = 5.009, 95% CI = 1.103-22.744; 95% PI = 0.016-1580.021) compared with PARP inhibitor or antiangiogenic agent monotherapy. Our study has not yet confirmed the benefit of combination therapy on OS in OC patients (HR = 0.885, 95% CI = 0.737-1.063). Additionally, subgroup analyses further showed that combination therapy resulted in an increased risk of AEs, encompassing thrombocytopenia, vomiting, abdominal pain, proteinuria, fatigue, headache, anorexia, and hypertension (all p < 0.05). Conclusion: Our study demonstrated the PFS benefit of combination therapy with PARP inhibitors and antiangiogenic agents in patients with OC. The OS result need to be updated after the original trial data is mature. Clinicians should be vigilant of AEs when administering the combination therapy in clinical practice. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023494482.
Collapse
Affiliation(s)
- Yan Wei
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li He
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Liu
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Guo
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Cong Xie
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jigang Jia
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yonghong Lin
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiang Liu
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiayin Fan
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Mittra A, Coyne GHOS, Zlott J, Kummar S, Meehan R, Rubinstein L, Juwara L, Wilsker D, Ji J, Miller B, Navas T, Ferry-Galow KV, Voth AR, Chang TC, Jiwani S, Parchment RE, Doroshow JH, Chen AP. Pharmacodynamic effects of the PARP inhibitor talazoparib (MDV3800, BMN 673) in patients with BRCA-mutated advanced solid tumors. Cancer Chemother Pharmacol 2024; 93:177-189. [PMID: 38010394 PMCID: PMC10902014 DOI: 10.1007/s00280-023-04600-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/02/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE Talazoparib is an inhibitor of the poly (ADP-ribose) polymerase (PARP) family of enzymes and is FDA-approved for patients with (suspected) deleterious germline BRCA1/2-mutated, HER2‑negative, locally advanced or metastatic breast cancer. Because knowledge of the pharmacodynamic (PD) effects of talazoparib in patients has been limited to studies of PARP enzymatic activity (PARylation) in peripheral blood mononuclear cells, we developed a study to assess tumoral PD response to talazoparib treatment (NCT01989546). METHODS We administered single-agent talazoparib (1 mg/day) orally in 28-day cycles to adult patients with advanced solid tumors harboring (suspected) deleterious BRCA1 or BRCA2 mutations. The primary objective was to examine the PD effects of talazoparib; the secondary objective was to determine overall response rate (ORR). Tumor biopsies were mandatory at baseline and post-treatment on day 8 (optional at disease progression). Biopsies were analyzed for PARylation, DNA damage response (γH2AX), and epithelial‒mesenchymal transition. RESULTS Nine patients enrolled in this trial. Four of six patients (67%) evaluable for the primary PD endpoint exhibited a nuclear γH2AX response on day 8 of treatment, and five of six (83%) also exhibited strong suppression of PARylation. A transition towards a more mesenchymal phenotype was seen in 4 of 6 carcinoma patients, but this biological change did not affect γH2AX or PAR responses. The ORR was 55% with the five partial responses lasting a median of six cycles. CONCLUSION Intra-tumoral DNA damage response and inhibition of PARP enzymatic activity were confirmed in patients with advanced solid tumors harboring BRCA1/2 mutations after 8 days of talazoparib treatment.
Collapse
Affiliation(s)
- Arjun Mittra
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD, 20892, USA
- Division of Medical Oncology, The Ohio State University, Columbus, OH, 43210, USA
| | - Geraldine H O' Sullivan Coyne
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD, 20892, USA
| | - Jennifer Zlott
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD, 20892, USA
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD, 20892, USA
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Robert Meehan
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD, 20892, USA
| | - Lawrence Rubinstein
- Biometric Research Program, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Lamin Juwara
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Deborah Wilsker
- Clinical Pharmacodynamics Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Jiuping Ji
- Clinical Pharmacodynamics Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Brandon Miller
- Clinical Pharmacodynamics Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Tony Navas
- Clinical Pharmacodynamics Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
- Regeneron Pharmaceuticals, Tarrytown, NY, 10591, USA
| | - Katherine V Ferry-Galow
- Clinical Pharmacodynamics Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Andrea Regier Voth
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Ting-Chia Chang
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Shahanawaz Jiwani
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Ralph E Parchment
- Clinical Pharmacodynamics Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD, 20892, USA
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
7
|
Caracciolo D, Juli G, Riillo C, Coricello A, Vasile F, Pollastri S, Rocca R, Scionti F, Polerà N, Grillone K, Arbitrio M, Staropoli N, Caparello B, Britti D, Loprete G, Costa G, Di Martino MT, Alcaro S, Tagliaferri P, Tassone P. Exploiting DNA Ligase III addiction of multiple myeloma by flavonoid Rhamnetin. Lab Invest 2022; 20:482. [PMID: 36273153 PMCID: PMC9588242 DOI: 10.1186/s12967-022-03705-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
Abstract
Background DNA ligases are crucial for DNA repair and cell replication since they catalyze the final steps in which DNA breaks are joined. DNA Ligase III (LIG3) exerts a pivotal role in Alternative-Non-Homologous End Joining Repair (Alt-NHEJ), an error-prone DNA repair pathway often up-regulated in genomically unstable cancer, such as Multiple Myeloma (MM). Based on the three-dimensional (3D) LIG3 structure, we performed a computational screening to identify LIG3-targeting natural compounds as potential candidates to counteract Alt-NHEJ activity in MM. Methods Virtual screening was conducted by interrogating the Phenol Explorer database. Validation of binding to LIG3 recombinant protein was performed by Saturation Transfer Difference (STD)—nuclear magnetic resonance (NMR) experiments. Cell viability was analyzed by Cell Titer-Glo assay; apoptosis was evaluated by flow cytometric analysis following Annexin V-7AAD staining. Alt-NHEJ repair modulation was evaluated using plasmid re-joining assay and Cytoscan HD. DNA Damage Response protein levels were analyzed by Western blot of whole and fractionated protein extracts and immunofluorescence analysis. The mitochondrial DNA (mtDNA) copy number was determined by qPCR. In vivo activity was evaluated in NOD-SCID mice subcutaneously engrafted with MM cells. Results Here, we provide evidence that a natural flavonoid Rhamnetin (RHM), selected by a computational approach, counteracts LIG3 activity and killed Alt-NHEJ-dependent MM cells. Indeed, Nuclear Magnetic Resonance (NMR) showed binding of RHM to LIG3 protein and functional experiments revealed that RHM interferes with LIG3-driven nuclear and mitochondrial DNA repair, leading to significant anti-MM activity in vitro and in vivo. Conclusion Taken together, our findings provide proof of concept that RHM targets LIG3 addiction in MM and may represent therefore a novel promising anti-tumor natural agent to be investigated in an early clinical setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03705-z.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Adriana Coricello
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | | | - Sara Pollastri
- Department of Chemistry, University of Milan, Milan, Italy
| | - Roberta Rocca
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Francesca Scionti
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), Messina, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), Catanzaro, Italy
| | | | - Basilio Caparello
- Presidio Ospedaliero Giovanni Paolo II Lamezia Terme, Catanzaro, Italy
| | - Domenico Britti
- Department of Health Science, Magna Græcia University, Catanzaro, Italy
| | - Giovanni Loprete
- Department of Health Science, Magna Græcia University, Catanzaro, Italy
| | - Giosuè Costa
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy. .,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Chan VKY, Yang R, Wong ICK, Li X. Cost-Effectiveness of Poly ADP-Ribose Polymerase Inhibitors in Cancer Treatment: A Systematic Review. Front Pharmacol 2022; 13:891149. [PMID: 35899114 PMCID: PMC9313592 DOI: 10.3389/fphar.2022.891149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background: PARP inhibitors have shown significant improvement in progression-free survival, but their costs cast a considerable financial burden. In line with value-based oncology, it is important to evaluate whether drug prices justify the outcomes. Objectives: The aim of the study was to systematically evaluate PARP inhibitors on 1) cost-effectiveness against the standard care, 2) impact on cost-effectiveness upon stratification for genetic characteristics, and 3) identify factors determining their cost-effectiveness, in four cancer types. Methods: We systematically searched PubMed, EMBASE, Web of Science, and Cochrane Library using designated search terms, updated to 31 August 2021. Trial-based or modeling cost-effectiveness analyses of four FDA-approved PARP inhibitors were eligible. Other studies known to authors were included. Reference lists of selected articles were screened. Eligible studies were assessed for methodological and reporting quality before review. Results: A total of 20 original articles proceeded to final review. PARP inhibitors were not cost-effective as recurrence maintenance in advanced ovarian cancer despite improved performance upon genetic stratification. Cost-effectiveness was achieved when moved to upfront maintenance in a new diagnosis setting. Limited evidence indicated non–cost-effectiveness in metastatic breast cancer, mixed conclusions in metastatic pancreatic cancer, and cost-effectiveness in metastatic prostate cancer. Stratification by genetic testing displayed an effect on cost-effectiveness, given the plummeting ICER values when compared to the “treat-all” strategy. Drug cost was a strong determinant for cost-effectiveness in most models. Conclusions: In advanced ovarian cancer, drug use should be prioritized for upfront maintenance and for patients with BRCA mutation or BRCAness at recurrence. Additional economic evaluations are anticipated for novel indications.
Collapse
Affiliation(s)
- Vivien Kin Yi Chan
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Runqing Yang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Ian Chi Kei Wong
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Research Department of Policy and Practice, School of Pharmacy, University College London, London, United Kingdom
- Laboratory of Data Discovery for Health (D4H), Hong Kong Science Park, Hong Kong SAR, China
| | - Xue Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D4H), Hong Kong Science Park, Hong Kong SAR, China
- HKU-Shenzhen Hospital, Shenzhen, China
- *Correspondence: Xue Li,
| |
Collapse
|
9
|
Hu J, Liang P, Jin D, Fan R, Xie X, Liu C, Jiang Q, Gao L. Poly (ADP-ribose) polymerase inhibitors (PARPi) for advanced malignancies with multiple DNA-repair genetic aberrations. Expert Rev Anticancer Ther 2022; 22:717-723. [PMID: 35679134 DOI: 10.1080/14737140.2022.2088513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Poly (ADP-ribose) polymerase inhibitors (PARPi) have been approved for the treatment of advanced tumors with defects in genes involved in homologous recombination repair (HRR), including cancers of the prostate, pancreas, breast, and ovary. In these advanced tumors, PARPi afford 'synthetic lethality' by blocking the PARP-associated repair pathway in cancer cells with HRR genetic mutations, resulting in chromosome instability and cellular apoptosis. According to the synthetic lethality theory, patients with a greater burden of genetic alterations, in proportion (relative quantity) or category, would have more satisfactory outcomes after PARPi administration. However, this issue remains obscure based on the existing sporadic evidence. AREAS COVERED We summarize the therapeutic effects of PARPi in advanced tumors with multiple HRR genetic mutations, and attempted to compare these results with those obtained for cancers with a single mutation. EXPERT OPINION Limited evidence has provided a possibly encouraging response to PARPi among patients carrying multiple HRR genetic mutations compared with those with a single mutation (although the treatment effect was negative in some patients). Further research is needed to understand the role of PARPi in tumor cells with multiple HRR genetic mutations.
Collapse
Affiliation(s)
- Jian Hu
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chong Qing, China
| | - Peihe Liang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chong Qing, China
| | - Dachun Jin
- Department of Urology, Daping Hospital/Army Medical Center of PLA, Army Medical University
| | - Runze Fan
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chong Qing, China
| | - Xiaodu Xie
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chong Qing, China
| | - Chuan Liu
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chong Qing, China
| | - Qing Jiang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chong Qing, China
| | - Liang Gao
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chong Qing, China
| |
Collapse
|
10
|
Wu K, Chen M, Peng X, Li Y, Tang G, Peng J, Cao X. Recent Progress of the research on the benzimidazole PARP-1 inhibitors. Mini Rev Med Chem 2022; 22:2438-2462. [PMID: 35319364 DOI: 10.2174/1389557522666220321150700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 11/22/2022]
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) is a multifunctional protein that plays an important role in DNA repair and genome integrity. PARP-1 inhibitors can be used as effective drugs not only to treat BRCA-1/2 deficient cancers because of the effect of synthetically lethal, but also to treat non-BRCA1/2 deficient tumours because of the effect of PARP capture. Therefore, the PARP inhibitors have become a focus of compelling research. Among these inhibitors, substituted benzimidazole derivatives were mainly concerned lead compounds. However, the commercial available benzimidazole PARP-1 inhibitors have some shortcomings such as serious toxicity in combination with chemotherapy drugs, in vivo cardiovascular side effects such as anemia. Therefore it's crucial for scientists to explore more structure-activity relationships of the benzimidazole PARP-1 inhibitors and access safer and more effective PARP inhibitors. As the binding region of PARP-1 and the substrates is usually characterized as NI site and AD site, the modification of benzimidazoles mainly occurs on the benzimidazole skeleton (NI site), and the side chain of benzimidazole on 2-C position (AD site). Herein, the recent progresses of the researches of benzamides PARP inhibitors were introduced. We noticed that even though many efforts were taken to the modification of NI sites, there were still lacks of optimistic and impressive results. However, the structure-activity relationships of the modification of AD sites have not thoroughly discovered yet. We hope that enlightened by the previous researches, more researches of AD site should be occurred and more effective benzimidazole PARP-1 inhibitors could be designed, synthesized, and applied to clinics.
Collapse
Affiliation(s)
- Kaiyue Wu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, College of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Miaojia Chen
- Department of Pharmacy, the first People\'s Hospital, Pingjiang, Yueyang, Hunan, China
| | - Xiaoyu Peng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, College of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yang Li
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, College of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, College of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Junmei Peng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, College of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xuan Cao
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, College of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
11
|
Crawford DR. Compassion and Empathy in Basic Medical Science Teaching: A Suggested Model. Cureus 2021; 13:e20205. [PMID: 35004025 PMCID: PMC8729821 DOI: 10.7759/cureus.20205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2021] [Indexed: 12/30/2022] Open
Abstract
Medical school education typically consists of two main student bodies: medical students and biomedical graduate students. For both groups, compassion and empathy represent a major component of future professional roles. For medical students, this takes the form of the all-important doctor-patient relationship and adherence to the Hippocratic Oath. For biomedical students, future research and teaching are often driven by the opportunity to contribute to treatments to help pain and suffering for those in need. For both groups, such positive contributions further extend to families, who often suffer emotional distress watching the health struggles of a loved one. Given the key role that compassion and empathy play here, including them as part of student educational development is important. Such focus, however, is limited - especially during the initial academic classroom years - with most time here dedicated to the learning of facts and foundational material. Given its importance in the future professional roles of these students, we posit that more can be done to introduce and reinforce the concept of compassion and empathy during the initial didactic course years. Modest but viable options exist for the introduction of these concepts as a part of basic teaching that will provide additional reinforcement of this all-important sensitivity for others. Here we present a model providing suggestions and recommendations for the integration of compassion and empathy in otherwise basic scientific teaching, and in a way that also includes progressive equality positions on social issues. While the focus here is medical school education since it represents this author’s expertise as well as a field where young trainees graduate to professional careers requiring compassion, it can potentially be applied to many other disciplines.
Collapse
|
12
|
Ren N, Zhang L, Yu J, Guan S, Dai X, Sun L, Ying M. Efficacy and Safety of PARP Inhibitor Combination Therapy in Recurrent Ovarian Cancer: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:638295. [PMID: 34485111 PMCID: PMC8414886 DOI: 10.3389/fonc.2021.638295] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Though it is known to all that PARP inhibitors (PARPis) are effective when used as maintenance alone for women with recurrent ovarian cancer (ROC), little is known about whether using them in combination with other drugs would contribute to a better efficacy. We performed a systematic review and meta-analysis to explore the efficacy and safety of PARPi combination therapy compared with monotherapy. MATERIALS AND METHODS We searched for randomized controlled trials (RCTs) that offered the date we needed in PubMed, Embase, Cochrane, and major conference. Data extraction and processing were completed by three investigators to compare OS, PFS, and ORR both in intervention and in control subset. Then, we calculated the pooled RR and 95% CI of all-grade and high-grade adverse effects to study its safety. And we evaluated the within-study heterogeneity by using subgroup and sensitivity analysis. RESULTS AND CONCLUSION A total of three eligible RCTs covering 343 women were included. In PFS analysis, PARP inhibitor (PARPi) combination therapy can significantly improve PFS for women with ROC when compared with the controls (HR: 0.46, 95% CI: 0.35 to 0.59), especially for those with mutated BRCA (HR: 0.29, 95% CI: 0.19 to 0.45). And in OS analysis, combination therapy is not inferior to monotherapy (HR: 0.90, 95% CI: 0.50 to 1.61). As for ORR, the effectiveness of combination therapy and monotherapy was almost the same (RR: 1.04, 95% CI: 0.82 to 1.31). Additionally, combination therapy seldom causes more adverse events, both in all-grade and in high grade. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/PROSPERO/, International Prospective Register of Systematic Reviews (PROSPERO) (identifier, CRD42018109933).
Collapse
Affiliation(s)
- Ning Ren
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Leyin Zhang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jieru Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Siqi Guan
- Department of Traditional Chinese Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xinyang Dai
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Leitao Sun
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Minli Ying
- Department of Gynaecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
13
|
Jeong KY, Park M. Poly adenosine diphosphate-ribosylation, a promising target for colorectal cancer treatment. World J Gastrointest Oncol 2021. [PMID: 34163574 DOI: 10.4251/wjgo.v13.i6.574.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The development of colorectal cancer (CRC) can result from changes in a variety of cellular systems within the tumor microenvironment. Particularly, it is primarily associated with genomic instability that is the gradual accumulation of genetic and epigenetic changes consisting of a characteristic set of mutations crucial for pathways in CRC progression. Based on this background, the potential to focus on poly [adenosine diphosphate (ADP)-ribose] polymerase (PARP)-1 and poly-ADP ribosylation (PARylation) as the main causes of malignant formation of CRC may be considered. One of the important functions of PARP-1 and PARylation is its deoxyribonucleic acid (DNA) repair function, which plays a pivotal role in the DNA damage response and prevention of DNA damage maintaining the redox homeostasis involved in the regulation of oxidation and superoxide. PARP-1 and PARylation can also alter epigenetic markers and chromatin structure involved in transcriptional regulation for the oncogenes or tumor suppressor genes by remodeling histone and chromatin enzymes. Given the high importance of these processes in CRC, it can be considered that PARP-1 and PARylation are at the forefront of the pathological changes required for CRC progression. Therefore, this review addresses the current molecular biological features for understanding the multifactorial function of PARP-1 and PARylation in CRC related to the aforementioned roles; furthermore, it presents a summary of recent approaches with PARP-1 inhibition in non-clinical and clinical studies targeting CRC. This understanding could help embrace the importance of targeting PARP-1 and PARylation in the treatment of CRC, which may present the potential to identify various research topics that can be challenged both non-clinically and clinically.
Collapse
Affiliation(s)
- Keun-Yeong Jeong
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea.
| | - Minhee Park
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea
| |
Collapse
|
14
|
Jeong KY, Park M. Poly adenosine diphosphate-ribosylation, a promising target for colorectal cancer treatment. World J Gastrointest Oncol 2021; 13:574-588. [PMID: 34163574 PMCID: PMC8204356 DOI: 10.4251/wjgo.v13.i6.574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/22/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023] Open
Abstract
The development of colorectal cancer (CRC) can result from changes in a variety of cellular systems within the tumor microenvironment. Particularly, it is primarily associated with genomic instability that is the gradual accumulation of genetic and epigenetic changes consisting of a characteristic set of mutations crucial for pathways in CRC progression. Based on this background, the potential to focus on poly [adenosine diphosphate (ADP)-ribose] polymerase (PARP)-1 and poly-ADP ribosylation (PARylation) as the main causes of malignant formation of CRC may be considered. One of the important functions of PARP-1 and PARylation is its deoxyribonucleic acid (DNA) repair function, which plays a pivotal role in the DNA damage response and prevention of DNA damage maintaining the redox homeostasis involved in the regulation of oxidation and superoxide. PARP-1 and PARylation can also alter epigenetic markers and chromatin structure involved in transcriptional regulation for the oncogenes or tumor suppressor genes by remodeling histone and chromatin enzymes. Given the high importance of these processes in CRC, it can be considered that PARP-1 and PARylation are at the forefront of the pathological changes required for CRC progression. Therefore, this review addresses the current molecular biological features for understanding the multifactorial function of PARP-1 and PARylation in CRC related to the aforementioned roles; furthermore, it presents a summary of recent approaches with PARP-1 inhibition in non-clinical and clinical studies targeting CRC. This understanding could help embrace the importance of targeting PARP-1 and PARylation in the treatment of CRC, which may present the potential to identify various research topics that can be challenged both non-clinically and clinically.
Collapse
Affiliation(s)
- Keun-Yeong Jeong
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea
| | - Minhee Park
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea
| |
Collapse
|
15
|
Caracciolo D, Riillo C, Di Martino MT, Tagliaferri P, Tassone P. Alternative Non-Homologous End-Joining: Error-Prone DNA Repair as Cancer's Achilles' Heel. Cancers (Basel) 2021; 13:cancers13061392. [PMID: 33808562 PMCID: PMC8003480 DOI: 10.3390/cancers13061392] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cancer onset and progression lead to a high rate of DNA damage, due to replicative and metabolic stress. To survive in this dangerous condition, cancer cells switch the DNA repair machinery from faithful systems to error-prone pathways, strongly increasing the mutational rate that, in turn, supports the disease progression and drug resistance. Although DNA repair de-regulation boosts genomic instability, it represents, at the same time, a critical cancer vulnerability that can be exploited for synthetic lethality-based therapeutic intervention. We here discuss the role of the error-prone DNA repair, named Alternative Non-Homologous End Joining (Alt-NHEJ), as inducer of genomic instability and as a potential therapeutic target. We portray different strategies to drug Alt-NHEJ and discuss future challenges for selecting patients who could benefit from Alt-NHEJ inhibition, with the aim of precision oncology. Abstract Error-prone DNA repair pathways promote genomic instability which leads to the onset of cancer hallmarks by progressive genetic aberrations in tumor cells. The molecular mechanisms which foster this process remain mostly undefined, and breakthrough advancements are eagerly awaited. In this context, the alternative non-homologous end joining (Alt-NHEJ) pathway is considered a leading actor. Indeed, there is experimental evidence that up-regulation of major Alt-NHEJ components, such as LIG3, PolQ, and PARP1, occurs in different tumors, where they are often associated with disease progression and drug resistance. Moreover, the Alt-NHEJ addiction of cancer cells provides a promising target to be exploited by synthetic lethality approaches for the use of DNA damage response (DDR) inhibitors and even as a sensitizer to checkpoint-inhibitors immunotherapy by increasing the mutational load. In this review, we discuss recent findings highlighting the role of Alt-NHEJ as a promoter of genomic instability and, therefore, as new cancer’s Achilles’ heel to be therapeutically exploited in precision oncology.
Collapse
|
16
|
Su D, Wu B, Shi L. Cost-Effectiveness of Genomic Test-Directed Olaparib for Metastatic Castration-Resistant Prostate Cancer. Front Pharmacol 2021; 11:610601. [PMID: 33574757 PMCID: PMC7870786 DOI: 10.3389/fphar.2020.610601] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose: The effectiveness of poly (adenosine diphosphate–ribose) polymerase (PARP) inhibitor olaparib for metastatic castration-resistant prostate cancer (MCRPC) with multiple loss-of-function alterations in genes that are involved in DNA repair has been demonstrated. We aimed to evaluate the cost-effectiveness of genomic test-directed olaparib on MCRPC from the US payer perspective. Methods: A partitioned survival model was adopted to project the disease course of MCRPC had at least one gene alteration in BRCA1, BRCA2 and ATM (Scenario A) and has alterations in any of all 15 prespecified genes (Scenario B) after next-generation sequencing test. The efficacy and toxicity data were gathered from the PROfound trial. Clinical probabilities related to survival were estimated from the reported survival probabilities in each PROfound group. Cost and health preference data were derived from the literature. The incremental cost-effectiveness ratio (ICER) was measured. Subgroup analysis and sensitivity analysis were performed for exploring the model uncertainties. Results: Olaparib yielded an additional 0.063 and 0.068 of quality-adjusted life year (QALY) with the augmented cost of $7,382 and saved the cost of $ 1,980 compared to standard care in scenario A and B, respectively, which yielded an ICER of $116,903/QALY and a cost-saving option. The lower weekly cost related to olaparib treatment led to the dominant findings in scenario B. The varied results between scenario A and B could be partly explained by different the number need to screen for identifying eligible patients who could be administered with olaparib, which sharply augmented the costs of the olaparib arm in scenario A. Subgroup analysis and sensitivity analysis revealed the results were generally robust in both of two scenarios. Conclusion: The genomic test-directed olaparib is a preferred option compared with standard care strategy for men with MCRPC who had any of all 15 prespecified genes.
Collapse
Affiliation(s)
- Dan Su
- Department of Pharmacy, the First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Bin Wu
- Medical Decision and Economic Group, Ren Ji Hospital, South Campus, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lizheng Shi
- Department of Global Health Management and Policy, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
17
|
Talreja VT, Noronha V, Joshi A, Patil V, Prabhash K. An exceptional response to olaparib in relapsed and refractory BRCA2 mutated non-small cell lung cancer in hereditary breast-ovarian cancer syndrome. South Asian J Cancer 2020; 9:6. [PMID: 31956609 PMCID: PMC6956583 DOI: 10.4103/sajc.sajc_157_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Vikas T Talreja
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Vijay Patil
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
18
|
Wu B, Shi L. Cost-Effectiveness of Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. J Natl Compr Canc Netw 2020; 18:1528-1536. [DOI: 10.6004/jnccn.2020.7587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 05/08/2020] [Indexed: 11/17/2022]
Abstract
Background: Maintenance therapy with the PARP inhibitor olaparib for metastatic pancreatic cancer (MPC) with a germline BRCA1 or BRCA2 mutation has been shown to be effective. We aimed to evaluate the cost-effectiveness of maintenance olaparib for MPC from the US payer perspective. Materials and Methods: A partitioned survival model was adopted to project the disease course of MPC. Efficacy and toxicity data were gathered from the Pancreas Cancer Olaparib Ongoing (POLO) trial. Transition probabilities were estimated from the reported survival probabilities in each POLO group. Cost and health preference data were derived from the literature. The incremental cost-utility ratio, incremental net-health benefit, and incremental monetary benefit were measured. Subgroup analysis, one-way analysis, and probabilistic sensitivity analysis were performed to explore the model uncertainties. Results: Maintenance olaparib had an incremental cost-utility ratio of $191,596 per additional progression-free survival (PFS) quality-adjusted life-year (QALY) gained, with a high cost of $132,287 and 0.691 PFS QALY gained, compared with results for a placebo. Subgroup analysis indicated that maintenance olaparib achieved at least a 16.8% probability of cost-effectiveness at the threshold of $200,000/QALY. One-way sensitivity analyses revealed that the results were sensitive to the hazard ratio of PFS and the cost of olaparib. When overall survival was considered, maintenance olaparib had an incremental cost-utility ratio of $265,290 per additional QALY gained, with a high cost of $128,266 and 0.483 QALY gained, compared with results for a placebo. Conclusions: Maintenance olaparib is potentially cost-effective compared with placebo for patients with a germline BRCA mutation and MPC. Economic outcomes could be improved by tailoring treatment based on individual patient factors.
Collapse
Affiliation(s)
- Bin Wu
- 1Medical Decision and Economic Group, Department of Pharmacy, Ren Ji Hospital, South Campus, School of Medicine, Shanghai Jiaotong University, Shanghai, China; and
| | - Lizheng Shi
- 2Department of Global Health Management and Policy, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
19
|
Sinha A, Saleh A, Endersby R, Yuan SH, Chokshi CR, Brown KR, Kuzio B, Kauppinen T, Singh SK, Baker SJ, McKinnon PJ, Katyal S. RAD51-Mediated DNA Homologous Recombination Is Independent of PTEN Mutational Status. Cancers (Basel) 2020; 12:cancers12113178. [PMID: 33138032 PMCID: PMC7693555 DOI: 10.3390/cancers12113178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary PTEN is an important tumor suppressor that is frequently mutated in malignancy. PTEN mutational loss has been associated with reduced RAD51 expression and homologous recombination deficiency (HRD), however; recent studies have failed to recapitulate these findings. Here, we show that RAD51 expression, foci formation and homologous recombination repair activity are unaltered in normal and tumorigenic PTEN-deficient cells and patient samples. Furthermore, we show that PTEN-deficient tumor cell lines do not synergize with the clinical PARP inhibitor olaparib, underscoring a need to discontinue its use in treating patients with PTEN-deficient tumors that do not otherwise exhibit HRD. Abstract PTEN mutation occurs in a variety of aggressive cancers and is associated with poor patient outcomes. Recent studies have linked mutational loss of PTEN to reduced RAD51 expression and function, a key factor involved in the homologous recombination (HR) pathway. However, these studies remain controversial, as they fail to establish a definitive causal link to RAD51 expression that is PTEN-dependent, while other studies have not been able to recapitulate the relationship between the PTEN expression and the RAD51/HR function. Resolution of this apparent conundrum is essential due to the clinically-significant implication that PTEN-deficient tumors may be sensitive to poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) commonly used in the clinical management of BRCA-mutated and other HR-deficient (HRD) tumors. Methods: Primary Pten-deficient (and corresponding wild-type) mouse embryonic fibroblasts (MEFs) and astrocytes and PTEN-null human tumor cell lines and primary cells were assessed for RAD51 expression (via the Western blot analysis) and DNA damage repair analyses (via alkali comet and γH2AX foci assays). RAD51 foci analysis was used to measure HR-dependent DNA repair. Xrcc2-deficient MEFs served as an HR-deficient control, while the stable knockdown of RAD51 (shRAD51) served to control for the relative RAD51/HR-mediated repair and the phospho-53BP1 foci analysis served to confirm and measure non-homologous end joining (NHEJ) activity in PTEN-deficient and shRAD51-expressing (HRD) lines. Cell proliferation studies were used to measure any potential added sensitivity of PTEN-null cells to the clinically-relevant PARPi, olaparib. RAD51 levels and DNA damage response signaling were assessed in PTEN-mutant brain tumor initiating cells (BTICs) derived from primary and recurrent glioblastoma multiforme (GBM) patients, while expression of RAD51 and its paralogs were examined as a function of the PTEN status in the RNA expression datasets isolated from primary GBM tumor specimens and BTICs. Results: Pten knockout primary murine cells display unaltered RAD51 expression, endogenous and DNA strand break-induced RAD51 foci and robust DNA repair activity. Defective HR was only observed in the cells lacking Xrcc2. Likewise, human glioblastoma multiforme (GBM) cell lines with known PTEN deficiency (U87, PTEN-mutated; U251 and U373, PTEN-null) show apparent expression of RAD51 and display efficient DNA repair activity. Only GBM cells stably expressing shRNAs against RAD51 (shRAD51) display dysfunctional DNA repair activity and reduced proliferative capacity, which is exacerbated by PARPi treatment. Furthermore, GBM patient-derived BTICs displayed robust RAD51 expression and intact DNA damage response signaling in spite of PTEN-inactivating mutations. RNA expression analysis of primary GBM tissue specimens and BTICs demonstrate stable levels of RAD51 and its paralogs (RAD51B, RAD51C, RAD51D, XRCC2, XRCC3, and DMC1), regardless of the PTEN mutational status. Conclusions: Our findings demonstrate definitively that PTEN loss does not alter the RAD51 expression, its paralogs, or the HR activity. Furthermore, deficiency in PTEN alone is not sufficient to impart enhanced sensitivity to PARPi associated with HRD. This study is the first to unequivocally demonstrate that PTEN deficiency is not linked to the RAD51 expression or the HR activity amongst primary neural and non-neural Pten-null cells, PTEN-deficient tumor cell lines, and primary PTEN-mutant GBM patient-derived tissue specimens and BTICs.
Collapse
Affiliation(s)
- Asha Sinha
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; (A.S.); (A.S.); (S.H.Y.); (B.K.); (T.K.)
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Ali Saleh
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; (A.S.); (A.S.); (S.H.Y.); (B.K.); (T.K.)
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Raelene Endersby
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Avenue, Perth, WA 6009, Australia;
- Centre for Child Health Research, University of Western Australia, 15 Hospital Avenue, Perth, WA 6009, Australia
| | - Shek H. Yuan
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; (A.S.); (A.S.); (S.H.Y.); (B.K.); (T.K.)
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Chirayu R. Chokshi
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, ON L8S 4L8, Canada; (C.R.C.); (S.K.S.)
| | - Kevin R. Brown
- Donnelly Centre, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada;
| | - Bozena Kuzio
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; (A.S.); (A.S.); (S.H.Y.); (B.K.); (T.K.)
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Tiina Kauppinen
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; (A.S.); (A.S.); (S.H.Y.); (B.K.); (T.K.)
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg, MB R3E 0Z3, Canada
| | - Sheila K. Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1200 Main Street West, Hamilton, ON L8S 4L8, Canada; (C.R.C.); (S.K.S.)
- Department of Surgery, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Suzanne J. Baker
- Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA;
| | - Peter J. McKinnon
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA;
| | - Sachin Katyal
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; (A.S.); (A.S.); (S.H.Y.); (B.K.); (T.K.)
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Correspondence: ; Tel.: +1-204-787-2765; Fax: +1-204-787-2190
| |
Collapse
|
20
|
Ciecielski KJ, Berninger A, Algül H. Precision Therapy of Pancreatic Cancer: From Bench to Bedside. Visc Med 2020; 36:373-380. [PMID: 33178734 PMCID: PMC7590788 DOI: 10.1159/000509232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/08/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC), with a mortality rate of 94% and a 5-year-survival rate of only 8%, is one of the deadliest cancer entities worldwide, and early diagnostic methods as well as effective therapies are urgently needed. SUMMARY This review summarizes current clinical procedure and recent developments of oncological therapy in the palliative setting of metastatic PDAC. It further gives examples of successful, as well as failed, targeted therapy approaches and finally discusses promising ongoing research into the decade-old question of the "undruggability" of KRAS. KEY MESSAGES Bench-driven concepts change the clinical landscape from "one size fits all" towards precision medicine. With growing insight into the molecular mechanisms of pancreatic cancer the era of targeted therapy in PDAC is gaining a new momentum.
Collapse
Affiliation(s)
| | | | - Hana Algül
- Comprehensive Cancer Center Munich (CCCM), Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
21
|
Huang TT, Brill E, Nair JR, Zhang X, Wilson KM, Chen L, Thomas CJ, Lee JM. Targeting the PI3K/mTOR Pathway Augments CHK1 Inhibitor-Induced Replication Stress and Antitumor Activity in High-Grade Serous Ovarian Cancer. Cancer Res 2020; 80:5380-5392. [PMID: 32998994 DOI: 10.1158/0008-5472.can-20-1439] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/04/2020] [Accepted: 09/18/2020] [Indexed: 01/08/2023]
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most lethal gynecologic malignancy in industrialized countries and has limited treatment options. Targeting ataxia-telangiectasia and Rad3-related/cell-cycle checkpoint kinase 1 (CHK1)-mediated S-phase and G2-M-phase cell-cycle checkpoints has been a promising therapeutic strategy in HGSOC. To improve the efficacy of CHK1 inhibitor (CHK1i), we conducted a high-throughput drug combination screening in HGSOC cells. PI3K/mTOR pathway inhibitors (PI3K/mTORi) showed supra-additive cytotoxicity with CHK1i. Combined treatment with CHK1i and PI3K/mTORi significantly attenuated cell viability and increased DNA damage, chromosomal breaks, and mitotic catastrophe compared with monotherapy. PI3K/mTORi decelerated fork speed by promoting new origin firing via increased CDC45, thus potentiating CHK1i-induced replication stress. PI3K/mTORi also augmented CHK1i-induced DNA damage by attenuating DNA homologous recombination repair activity and RAD51 foci formation. High expression of replication stress markers was associated with poor prognosis in patients with HGSOC. Our findings indicate that combined PI3K/mTORi and CHK1i induces greater cell death in HGSOC cells and in vivo models by causing lethal replication stress and DNA damage. This insight can be translated therapeutically by further developing combinations of PI3K and cell-cycle pathway inhibitors in HGSOC. SIGNIFICANCE: Dual inhibition of CHK1 and PI3K/mTOR pathways yields potent synthetic lethality by causing lethal replication stress and DNA damage in HGSOC, warranting further clinical development.
Collapse
Affiliation(s)
- Tzu-Ting Huang
- Women's Malignancies Branch, Center for Cancer Research, NCI, Bethesda, Maryland.
| | - Ethan Brill
- Women's Malignancies Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Jayakumar R Nair
- Women's Malignancies Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland
| | - Kelli M Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland
| | - Lu Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland.,Lymphoid Malignancies Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| |
Collapse
|
22
|
Lampert EJ, Zimmer A, Padget M, Cimino-Mathews A, Nair JR, Liu Y, Swisher EM, Hodge JW, Nixon AB, Nichols E, Bagheri MH, Levy E, Radke MR, Lipkowitz S, Annunziata CM, Taube JM, Steinberg SM, Lee JM. Combination of PARP Inhibitor Olaparib, and PD-L1 Inhibitor Durvalumab, in Recurrent Ovarian Cancer: a Proof-of-Concept Phase II Study. Clin Cancer Res 2020; 26:4268-4279. [PMID: 32398324 PMCID: PMC7442720 DOI: 10.1158/1078-0432.ccr-20-0056] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/06/2020] [Accepted: 05/08/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE Preclinical studies suggest PARP inhibition (PARPi) induces immunostimulatory micromilieu in ovarian cancer thus complementing activity of immune checkpoint blockade. We conducted a phase II trial of PARPi olaparib and anti-PD-L1 durvalumab and collected paired fresh core biopsies and blood samples to test this hypothesis. PATIENTS AND METHODS In a single-center, proof-of-concept phase II study, we enrolled women aged ≥18 with recurrent ovarian cancer. All patients were immune checkpoint inhibitor-naïve and had measurable disease per RECISTv1.1, ECOG performance status 0-2, and adequate organ and marrow function. Patients received olaparib 300 mg twice daily and durvalumab 1,500 mg intravenously every 4 weeks until disease progression, unacceptable toxicity, or withdrawal of consent. Primary endpoint was overall response rate (ORR). Secondary objectives were safety and progression-free survival (PFS). Translational objectives included biomarker evaluation for relationships with clinical response and immunomodulatory effects by treatment. RESULTS Thirty-five patients with ovarian cancer [median, four prior therapies (IQR, 2-5.5), predominantly platinum-resistant (86%), BRCA wild-type (77%)] received at least one full cycle of treatment. ORR was 14% [5/35; 95% confidence interval (CI), 4.8%-30.3%]. Disease control rate (PR+SD) was 71% (25/35; 95% CI, 53.7%-85.4%). Treatment enhanced IFNγ and CXCL9/CXCL10 expression, systemic IFNγ/TNFα production, and tumor-infiltrating lymphocytes, indicating an immunostimulatory environment. Increased IFNγ production was associated with improved PFS [HR, 0.37 (95% CI, 0.16-0.87), P = 0.023], while elevated VEGFR3 levels were associated with worse PFS (HR, 3.22 (95% CI, 1.23-8.40), P = 0.017]. CONCLUSIONS The PARPi and anti-PD-L1 combination showed modest clinical activity in recurrent ovarian cancer. Our correlative study results suggest immunomodulatory effects by olaparib/durvalumab in patients and indicate that VEGF/VEGFR pathway blockade would be necessary for improved efficacy of the combination.
Collapse
Affiliation(s)
- Erika J Lampert
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Alexandra Zimmer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michelle Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | - Jayakumar R Nair
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Elizabeth M Swisher
- Division of Gynecologic Oncology, Departments of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Erin Nichols
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Bethesda, Maryland
| | - Mohammad H Bagheri
- Department of Radiology and Imaging Sciences, Clinical Center, National Cancer Institute, Bethesda, Maryland
| | - Elliott Levy
- Interventional Radiology, NIH Clinical Center, Bethesda, Maryland
| | - Marc R Radke
- Division of Gynecologic Oncology, Departments of Obstetrics and Gynecology, University of Washington, Seattle, Washington
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Janis M Taube
- Department of Dermatopathology, The Johns Hopkins Medical Institution, Baltimore, Maryland
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
23
|
Mariniello A, Ghisoni E, Righi L, Catino A, Chiari R, Del Conte A, Barbieri F, Cecere F, Gelibter A, Giajlevra M, Parra HS, Zichi C, DI Maio M, Valabrega G, Novello S. Women With Synchronous or Metachronous Lung and Ovarian Cancer: A Multi-Institutional Report. In Vivo 2020; 33:2021-2026. [PMID: 31662533 DOI: 10.21873/invivo.11699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND/AIM Double diagnosis of lung cancer (LC) and ovarian cancer (OC) is rare. Here, we describe patients with synchronous/metachronous LC and OC to identify common clinical and pathological patterns. PATIENTS AND METHODS Clinical, pathological and molecular data of patients diagnosed and treated at 30 European Institutions from 2008 to 2018 were retrieved and analysed. Whenever tissue was available, centralized pathology revision was performed. RESULTS A total of 19 cases were found; one was excluded at pathology revision. Most LCs were adenocarcinomas (15/18) and most OCs were high-grade serous (15/18) carcinomas. Of the 9 patients analysed, 7 carried oncogene-addicted LC (4 EGFR, 1 B-RAF and 2 ALK) and five out of 7 carried BRCA mutations. One patient with a germline-BRCA1 mutation received olaparib, resulting in a durable response of both malignancies. Median overall survival was 33 months. CONCLUSION In our series, most synchronous/metachronous LCs and OCs showed genetic alterations. Further analyses with wide NGS panel could shed light on the biological mechanisms driving their occurrence.
Collapse
Affiliation(s)
- Annapaola Mariniello
- Department of Oncology, University of Torino at San Luigi University Hospital, Orbassano, Italy
| | - Eleonora Ghisoni
- Department of Oncology, University of Torino, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Luisella Righi
- Department of Oncology, University of Torino at San Luigi University Hospital, Orbassano, Italy
| | - Annamaria Catino
- Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Rita Chiari
- Department of Medical Oncology, University of Perugia at Santa Maria della Misericordia Hospital, Perugia, Italy
| | | | - Fausto Barbieri
- Department of Oncology and Hematology, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Fabiana Cecere
- Careggi University Hospital, Medical Oncology Unit, Department of Oncology, Florence, Italy
| | - Alain Gelibter
- Sapienza University of Rome at Policlinico Umberto I, Oncology Unit, Rome, Italy
| | - Matteo Giajlevra
- CHU Grenoble Alpes, Clinique Universitaire de Pneumologie, Pôle Thorax et Vaisseaux, Grenoble, France
| | | | - Clizia Zichi
- Department of Oncology, University of Torino at San Luigi University Hospital, Orbassano, Italy
| | - Massimo DI Maio
- Department of Oncology, University of Torino at Mauriziano Umberto I Hospital, Turin, Italy
| | - Giorgio Valabrega
- Department of Oncology, University of Torino, Turin, Italy .,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Silvia Novello
- Department of Oncology, University of Torino at San Luigi University Hospital, Orbassano, Italy
| |
Collapse
|
24
|
Matsubayashi H, Takaori K, Morizane C, Kiyozumi Y. Familial Pancreatic Cancer and Surveillance of High-Risk Individuals. Gut Liver 2020; 13:498-505. [PMID: 30917631 PMCID: PMC6743804 DOI: 10.5009/gnl18449] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/04/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022] Open
Abstract
Family history of pancreatic cancer (PC) is a risk factor for PC development, and the risk level correlates with the number of affected families. A case of PC with ≥1 PC cases in the first-degree relative is broadly defined as familial pancreatic cancer (FPC) and accounts for 5% to 10% of total PC cases. FPC possesses several epidemiological, genetic and clinicopathological aspects that are distinct from those of conventional PCs. In Western countries, FPC registries have been established since the 1990s, and high-risk individuals are screened to detect early PCs. For the pharmacotherapy of FPC, especially in cases with germline pathogenic BRCA mutations, regimens using platinum and poly (ADP-ribose) polymerase inhibitor have recently been studied for their effectiveness. To date, the concept of FPC has prevailed in Western countries, and it has begun to infiltrate into Eastern countries. As the genetic background and environmental conditions vary in association with ethnicity and living area, we need to establish our own FPC registries and accumulate data in Asian countries.
Collapse
Affiliation(s)
- Hiroyuki Matsubayashi
- Divisions of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan.,Divisions of Endoscopy, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kyoichi Takaori
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chigusa Morizane
- Division of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshimi Kiyozumi
- Divisions of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
25
|
Przybycinski J, Nalewajska M, Marchelek-Mysliwiec M, Dziedziejko V, Pawlik A. Poly-ADP-ribose polymerases (PARPs) as a therapeutic target in the treatment of selected cancers. Expert Opin Ther Targets 2019; 23:773-785. [PMID: 31394942 DOI: 10.1080/14728222.2019.1654458] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The implementation of poly-ADP-ribose polymerase (PARP) inhibitors for therapy has created potential treatments for a wide spectrum of malignancies involving DNA damage repair gene abnormalities. PARPs are a group of enzymes that are responsible for detecting and repairing DNA damage and therefore play a key role in maintaining cell function and integrity. PARP inhibitors are drugs that target DNA repair deficiencies. Inhibiting PARP activity in cancer cells causes cell death. Areas covered: This review summarizes the role of PARP inhibitors in the treatment of cancer. We performed a systematic literature search in February 2019 in the electronic databases PubMed and EMBASE. Our search terms were the following: PARP, PARP inhibitors, PARPi, Poly ADP ribose polymerase, cancer treatment. We discuss PARP inhibitors currently being investigated in cancer clinical trials, their safety profiles, clinical resistance, combined therapeutic approaches and future challenges. Expert Opinion: The future could bring novel PARP inhibitors with greater DNA trapping potential, better safety profiles and improved combined therapies involving hormonal, chemo-, radio- or immunotherapies. Progress may afford wider indications for PARP inhibitors in the treatment of cancer and the utilization for cancer prevention in high-risk mutation carriers. Research efforts should focus on identifying novel drugs that target DNA repair deficiencies.
Collapse
Affiliation(s)
- Jarosław Przybycinski
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University , Szczecin , Poland
| | - Magdalena Nalewajska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University , Szczecin , Poland
| | | | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University , Szczecin , Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University , Szczecin , Poland
| |
Collapse
|
26
|
Wortman BG, Nout RA, Bosse T, Creutzberg CL. Selecting Adjuvant Treatment for Endometrial Carcinoma Using Molecular Risk Factors. Curr Oncol Rep 2019; 21:83. [PMID: 31367798 PMCID: PMC6669195 DOI: 10.1007/s11912-019-0825-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To provide an overview of common molecular risk factors in endometrial cancer (EC) with the possibility to improve adjuvant treatment selection. RECENT FINDINGS Recent studies have discovered and confirmed four different molecular subclasses in EC, with each having a distinct prognosis; POLE-ultramutated, microsatellite unstable, copy-number low, and copy-number high. Subsequent studies have shown that combining both molecular with clinicopathological risk factors can potentially improve adjuvant treatment selection for women with high-intermediate risk EC. For high risk and advanced stage EC, several molecular alterations are being explored for targeted therapy. Molecular alterations are frequently found in endometrial cancer and have currently not been implemented in the treatment guidelines for EC. Assessment of molecular alterations can distinguish patients that require less or more intensified adjuvant treatment. Trials investigating targeted therapies in EC are ongoing and have shown some promising results, however, more evidence is needed and results of randomized trials have to be awaited.
Collapse
Affiliation(s)
- Bastiaan G Wortman
- Department of Radiation Oncology, Leiden University Medical Center, K1-P, Albinusdreef 2, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Remi A Nout
- Department of Radiation Oncology, Leiden University Medical Center, K1-P, Albinusdreef 2, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carien L Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, K1-P, Albinusdreef 2, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
27
|
McCann KE. Advances in the use of PARP inhibitors for BRCA1/2-associated breast cancer: talazoparib. Future Oncol 2019; 15:1707-1715. [DOI: 10.2217/fon-2018-0751] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Poly-ADP-ribosyl polymerase (PARP) enzymes PARP-1 and PARP-2 recognize DNA damage and set off a cascade of cellular mechanisms required for multiple types of DNA damage repair. PARP inhibitors are small molecule mimetics of nicotinamide which bind to PARP’s catalytic domain to inhibit poly-ADP-ribosylation (PARylation) of target proteins, including PARP-1 itself. PARP inhibitors olaparib, veliparib, talazoparib, niraparib and rucaparib have predominantly been studied in women with breast or ovarian cancers associated with deleterious germline mutations in BRCA1 and BRCA2 (gBRCA1/2+). The BRCA1 and BRCA2 proteins are involved in DNA repair by homologous recombination. This review will focus on talazoparib, a PARP inhibitor approved by the US FDA for the treatment of metastatic gBRCA1/2+ breast cancers in October 2018.
Collapse
Affiliation(s)
- Kelly E McCann
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
28
|
Kassab MA, Yu X. The role of dePARylation in DNA damage repair and cancer suppression. DNA Repair (Amst) 2019; 76:20-29. [PMID: 30807923 DOI: 10.1016/j.dnarep.2019.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/03/2019] [Indexed: 12/19/2022]
Abstract
Poly(ADP-ribosyl)ation (PARylation) is a reversible post-translational modification regulating various biological pathways including DNA damage repair (DDR). Rapid turnover of PARylation is critically important for an optimal DNA damage response and maintaining genomic stability. Recent studies show that PARylation is tightly regulated by a group of enzymes that can erase the ADP-ribose (ADPR) groups from target proteins. The aim of this review is to present a comprehensive understanding of dePARylation enzymes, their substrates and roles in DDR. Special attention will be laid on the role of these proteins in the development of cancer and their feasibility in anticancer therapeutics.
Collapse
Affiliation(s)
- Muzaffer Ahmad Kassab
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010, USA
| | - Xiaochun Yu
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
29
|
Quaas A, Heydt C, Waldschmidt D, Alakus H, Zander T, Goeser T, Kasper P, Bruns C, Brunn A, Roth W, Hartmann N, Bunck A, Schmidt M, Buettner R, Merkelbach-Bruse S. Alterations in ERBB2 and BRCA and microsatellite instability as new personalized treatment options in small bowel carcinoma. BMC Gastroenterol 2019; 19:21. [PMID: 30717682 PMCID: PMC6360678 DOI: 10.1186/s12876-019-0942-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 01/25/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Carcinomas of the small bowel are rare tumors usually with dismal prognosis. Most recently, some potentially treatable molecular alterations were described. We emphasize the growing evidence of individualized treatment options in small bowel carcinoma. METHODS We performed a DNA- based multi-gene panel using ultra-deep sequencing analysis (including 14 genes with up to 452 amplicons in total; KRAS, NRAS, HRAS, BRAF, DDR2, ERBB2, KEAP1, NFE2L2, PIK3CA, PTEN, RHOA, BRCA1, BRCA2 and TP53) as well as an RNA-based gene fusion panel including ALK, BRAF, FGFR1, FGFR2, FGFR3, MET, NRG1, NTRK1, NTRK2, NTRK3, RET and ROS1 on eleven formalin fixed and paraffin embedded small bowel carcinomas. Additionally, mismatch-repair-deficiency was analyzed by checking the microsatellite status using the five different mononucleotide markers BAT25, BAT26, NR-21, NR-22 and NR-27 and loss of mismatch repair proteins using four different markers (MLH1, MSH6, MSH2, PMS2). RESULTS In five out of eleven small bowel carcinomas we found potentially treatable genetic alterations. Three patients demonstrated pathogenic (class 5) BRCA1 or BRCA2 mutations - one germline-related in a mixed neuroendocrine-non neuroendocrine neoplasm (MiNEN). Two additional patients revealed an activating ERBB2 mutation or PIK3CA mutation. Furthermore two tumors were highly microsatellite-instable (MSI-high), in one case associated to Lynch-syndrome. We did not find any gene fusions. CONCLUSION Our results underscore, in particular, the relevance of potentially treatable molecular alterations (like ERBB2, BRCA and MSI) in small bowel carcinomas. Further studies are needed to proof the efficacy of these targeted therapies in small bowel carcinomas.
Collapse
Affiliation(s)
- Alexander Quaas
- Institute of Pathology, University of Cologne, Cologne, Germany.
| | - Carina Heydt
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Dirk Waldschmidt
- Department of Hepato- and Gastroenterology, University of Cologne, Cologne, Germany
| | - Hakan Alakus
- Department of Visceral Surgery, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department of Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Tobias Goeser
- Department of Hepato- and Gastroenterology, University of Cologne, Cologne, Germany
| | - Philipp Kasper
- Department of Hepato- and Gastroenterology, University of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of Visceral Surgery, University of Cologne, Cologne, Germany
| | - Anna Brunn
- Institute of Neuropathology, University of Cologne, Cologne, Germany
| | - Wilfried Roth
- Institute of Pathology, University of Mainz, Mainz, Germany
| | - Nils Hartmann
- Institute of Pathology, University of Mainz, Mainz, Germany
| | - Anne Bunck
- Department of Radiology, University of Cologne, Cologne, Germany
| | - Matthias Schmidt
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | | | | |
Collapse
|
30
|
Koyama T, Rhrissorrakrai K, Parida L. Analysis on GENIE reveals novel recurrent variants that affect molecular diagnosis of sizable number of cancer patients. BMC Cancer 2019; 19:114. [PMID: 30709382 PMCID: PMC6359859 DOI: 10.1186/s12885-019-5313-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/22/2019] [Indexed: 12/18/2022] Open
Abstract
Background Significant numbers of variants detected in cancer patients are often left labeled only as variants of unknown significance (VUS). In order to expand precision medicine to a wider population, we need to extend our knowledge of pathogenicity and drug response in the context of VUS’s. Methods In this study, we analyzed variants from AACR Project GENIE Consortium APG (Cancer Discov 7:818-831, 2017) and compared them to the COSMIC database Forbes et al. (Nucleic Acids Res 43:D805-811, 2015) to identify recurrent variants that would merit further study. We filtered out known hotspot variants, inactivating variants in tumor suppressors, and likely benign variants by comparing with COSMIC and ExAC Lee et al. (Science 337:967-971, 2012). Results We have identified 45,933 novel variants with unknown significance unique to GENIE. In our analysis, we found on average six variants per patient where two could be considered as pathogenic or likely pathogenic and the majority are VUS’s. More importantly, we have discovered 730 recurrent variants that appear more than 3 times in GENIE but less than 3 in COSMIC. If we combine the recurrences of GENIE and COSMIC for all variants, 2586 are newly identified as occurring more than 3 times than when using COSMIC alone. Conclusions Although it would be inappropriate to blindly accept these recurrent variants as pathogenic, they may warrant higher priority than other observed VUS’s. These newly identified recurrent variants might affect the molecular profiles of approximately 1 in 6 patients. Further analysis and characterization of these variants in both research and clinical contexts will improve patient treatments and the development of new therapeutics. Electronic supplementary material The online version of this article (10.1186/s12885-019-5313-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takahiko Koyama
- IBM TJ Watson Research Center, Yorktown Heights, NY, 10598, USA.
| | | | - Laxmi Parida
- IBM TJ Watson Research Center, Yorktown Heights, NY, 10598, USA
| |
Collapse
|
31
|
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: 1.8] [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
|
32
|
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
|
33
|
Quaas A, Waldschmidt D, Alakus H, Zander T, Heydt C, Goeser T, Daheim M, Kasper P, Plum P, Bruns C, Brunn A, Roth W, Hartmann N, Bunck A, Schmidt M, Göbel H, Tharun L, Buettner R, Merkelbach-Bruse S. Therapy susceptible germline-related BRCA 1-mutation in a case of metastasized mixed adeno-neuroendocrine carcinoma (MANEC) of the small bowel. BMC Gastroenterol 2018; 18:75. [PMID: 29855275 PMCID: PMC5984468 DOI: 10.1186/s12876-018-0803-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 05/23/2018] [Indexed: 01/04/2023] Open
Abstract
Background Adenocarcinomas or combined adeno-neuroendocrine carcinomas (MANEC) of small bowel usually have a dismal prognosis with limited systemic therapy options. This is the first description of a patient showing a germline-related BRCA1 mutated MANEC of his ileum. The tumor presented a susceptibility to a combined chemotherapy and the PARP1-inhibitor olaparib. Case presentation A 74-year old male patient presented with a metastasized MANEC of his ileum. Due to clinical symptoms his ileum-tumor and the single brain metastasis were removed. We verified the same pathogenic (class 5) BRCA1 mutation in different tumor locations. There was no known personal history of a previous malignant tumor. Nevertheless we identified his BRCA1 mutation as germline-related. A systemic treatment was started including Gemcitabine followed by selective internal radiotherapy (SIRT) to treat liver metastases and in the further course Capecitabine but this treatment finally failed after 9 months and all liver metastases showed progression. The treatment failure was the reason to induce an individualized therapeutic approach using combined chemotherapy of carboplatin, paclitaxel and the Poly (ADP-ribose) polymerase- (PARP)-inhibitor olaparib analogous to the treatment protocol of Oza et al. All liver metastases demonstrated with significant tumor regression after 3 months and could be removed. In his most current follow up from December 2017 (25 months after his primary diagnosis) the patient is in a very good general condition without evidence for further metastases. Conclusion We present first evidence of a therapy susceptible germline-related BRCA1 mutation in small bowel adeno-neuroendocrine carcinoma (MANEC). Our findings offer a personalized treatment option. The germline background was unexpected in a 74-year old man with no previously known tumor burden. We should be aware of the familiar background in tumors of older patients as well.
Collapse
Affiliation(s)
- A Quaas
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany. .,Gastrointestinal Cancer Group Cologne, Cologne, Germany.
| | - D Waldschmidt
- Department of Hepato- and Gastroenterology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - H Alakus
- Department of Visceral Surgery, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany.,Gastrointestinal Cancer Group Cologne, Cologne, Germany
| | - T Zander
- Department of Oncology and Hematology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany.,Gastrointestinal Cancer Group Cologne, Cologne, Germany
| | - C Heydt
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - T Goeser
- Department of Hepato- and Gastroenterology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - M Daheim
- Department of Hepato- and Gastroenterology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - P Kasper
- Department of Hepato- and Gastroenterology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - P Plum
- Department of Visceral Surgery, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - C Bruns
- Department of Visceral Surgery, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - A Brunn
- Institute of Neuropathology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - W Roth
- Institute of Pathology, University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - N Hartmann
- Institute of Pathology, University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - A Bunck
- Department of Radiology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - M Schmidt
- Department of Nuclear-Medicine, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - H Göbel
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - L Tharun
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - R Buettner
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - S Merkelbach-Bruse
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| |
Collapse
|
34
|
Sizemore ST, Mohammad R, Sizemore GM, Nowsheen S, Yu H, Ostrowski MC, Chakravarti A, Xia F. Synthetic Lethality of PARP Inhibition and Ionizing Radiation is p53-dependent. Mol Cancer Res 2018; 16:1092-1102. [PMID: 29592899 DOI: 10.1158/1541-7786.mcr-18-0106] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
PARP inhibitors (PARPi) are potentially effective therapeutic agents capable of inducing synthetic lethality in tumors with deficiencies in homologous recombination (HR)-mediated DNA repair such as those carrying BRCA1 mutations. However, BRCA mutations are rare, the majority of tumors are proficient in HR repair, and thus most tumors are resistant to PARPi. Previously, we observed that ionizing radiation (IR) initiates cytoplasmic translocation of BRCA1 leading to suppression of HR-mediated DNA repair and induction of synthetic PARPi lethality in wild-type BRCA1 and HR-proficient tumor cells. The tumor suppressor p53 was identified as a key factor that regulates DNA damage-induced BRCA1 cytoplasmic sequestration following IR. However, the role of p53 in IR-induced PARPi sensitization remains unclear. This study elucidates the role of p53 in IR-induced PARPi cytotoxicity in HR-proficient cancer cells and suggests p53 status may help define a patient population that might benefit from this treatment strategy. Sensitization to PARPi following IR was determined in vitro and in vivo utilizing human breast and glioma tumor cells carrying wild-type BRCA1 and p53, and in associated cells in which p53 function was modified by knockdown or mutation. In breast and glioma cells with proficient HR repair, IR-induced BRCA1 cytoplasmic sequestration, HR repair inhibition, and subsequent PARPi sensitization in vitro and in vivo was dependent upon functional p53.Implications: Implications: p53 status determines PARP inhibitor sensitization by ionizing radiation in multiple BRCA1 and HR-proficient tumor types and may predict which patients are most likely to benefit from combination therapy. Mol Cancer Res; 16(7); 1092-102. ©2018 AACR.
Collapse
Affiliation(s)
- Steven T Sizemore
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Rahman Mohammad
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Gina M Sizemore
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Clinic, Rochester, Minnesota
| | - Hao Yu
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Fen Xia
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
35
|
Murai J, Feng Y, Yu GK, Ru Y, Tang SW, Shen Y, Pommier Y. Resistance to PARP inhibitors by SLFN11 inactivation can be overcome by ATR inhibition. Oncotarget 2018; 7:76534-76550. [PMID: 27708213 PMCID: PMC5340226 DOI: 10.18632/oncotarget.12266] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 12/20/2022] Open
Abstract
Poly(ADP-ribose) polymerase inhibitors (PARPIs) kill cancer cells by trapping PARP1 and PARP2. Talazoparib, the most potent PARPI inhibitor (PARPI), exhibits remarkable selectivity among the NCI-60 cancer cell lines beyond BRCA inactivation. Our genomic analyses reveal high correlation between response to talazoparib and Schlafen 11 (SLFN11) expression. Causality was established in four isogenic SLFN11-positive and -negative cell lines and extended to olaparib. Response to the talazoparib-temozolomide combination was also driven by SLFN11 and validated in 36 small cell lung cancer cell lines, and in xenograft models. Resistance in SLFN11-deficient cells was caused neither by impaired drug penetration nor by activation of homologous recombination. Rather, SLFN11 induced irreversible and lethal replication inhibition, which was independent of ATR-mediated S-phase checkpoint. The resistance to PARPIs by SLFN11 inactivation was overcome by ATR inhibition, mechanistically because SLFN11-deficient cells solely rely on ATR activation for their survival under PARPI treatment. Our study reveals that SLFN11 inactivation, which is common (~45%) in cancer cells, is a novel and dominant resistance determinant to PARPIs.
Collapse
Affiliation(s)
- Junko Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ying Feng
- BioMarin Pharmaceutical Inc., Novato, CA, USA
| | | | - Yuanbin Ru
- BioMarin Pharmaceutical Inc., Novato, CA, USA
| | - Sai-Wen Tang
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Current affiliation: Division of Blood and Marrow Transplantation, Department of Medicine, Stranford University School of Medicine, Stanford, CA, USA
| | - Yuqiao Shen
- BioMarin Pharmaceutical Inc., Novato, CA, USA
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
Chae YK, Anker JF, Carneiro BA, Chandra S, Kaplan J, Kalyan A, Santa-Maria CA, Platanias LC, Giles FJ. Genomic landscape of DNA repair genes in cancer. Oncotarget 2018; 7:23312-21. [PMID: 27004405 PMCID: PMC5029628 DOI: 10.18632/oncotarget.8196] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 03/02/2016] [Indexed: 01/09/2023] Open
Abstract
DNA repair genes are frequently mutated in cancer, yet limited data exist regarding the overall genomic landscape and functional implications of these alterations in their entirety. We created comprehensive lists of DNA repair genes and indirect caretakers. Mutation, copy number variation (CNV), and expression frequencies of these genes were analyzed in COSMIC. Mutation co-occurrence, clinical outcomes, and mutation burden were analyzed in TCGA. We report the 20 genes most frequently with mutations (n > 19,689 tumor samples for each gene), CNVs (n > 1,556), or up- or down-regulated (n = 7,998). Mutual exclusivity was observed as no genes displayed both high CNV gain and loss or high up- and down-regulation, and CNV gain and loss positively correlated with up- and down-regulation, respectively. Co-occurrence of mutations differed between cancers, and mutations in many DNA repair genes were associated with higher total mutation burden. Mutation and CNV frequencies offer insights into which genes may play tumor suppressive or oncogenic roles, such as NEIL2 and RRM2B, respectively. Mutual exclusivities within CNV and expression frequencies, and correlations between CNV and expression, support the functionality of these genomic alterations. This study provides comprehensive lists of candidate genes as potential biomarkers for genomic instability, novel therapeutic targets, or predictors of immunotherapy efficacy.
Collapse
Affiliation(s)
- Young Kwang Chae
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jonathan F Anker
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benedito A Carneiro
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sunandana Chandra
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason Kaplan
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Aparna Kalyan
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Cesar A Santa-Maria
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leonidas C Platanias
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Hematology-Oncology, Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Francis J Giles
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.,Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
37
|
Eetezadi S, Evans JC, Shen YT, De Souza R, Piquette-Miller M, Allen C. Ratio-Dependent Synergism of a Doxorubicin and Olaparib Combination in 2D and Spheroid Models of Ovarian Cancer. Mol Pharm 2018; 15:472-485. [DOI: 10.1021/acs.molpharmaceut.7b00843] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Sina Eetezadi
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - James C. Evans
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Yen-Ting Shen
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Raquel De Souza
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Micheline Piquette-Miller
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
38
|
Pazopanib Maintenance Therapy in East Asian Women With Advanced Epithelial Ovarian Cancer: Results From AGO-OVAR16 and an East Asian Study. Int J Gynecol Cancer 2018; 28:2-10. [DOI: 10.1097/igc.0000000000000602] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
ObjectiveThe recent phase 3 trial AGO-OVAR16 demonstrated that pazopanib maintenance improved median progression-free survival in patients with ovarian cancer whose disease did not progress during first-line treatment. However, this improvement was not seen in the subset of East Asian patients. The current analysis evaluated the efficacy and safety of pazopanib maintenance in East Asian patients from AGO-OVAR16 and a separate East Asian study.Materials and MethodsEast Asian patients from AGO-OVAR16 (n = 209) and the East Asian study (N = 145) were randomized 1:1 to receive pazopanib 800 mg/d or placebo for up to 24 months. The primary end point for each study was progression-free survival by RECIST (Response Evaluation Criteria in Solid Tumors) based on investigator assessment. Clinical and genetics data were analyzed separately by study or pooled according to separate predetermined statistical plans.ResultsPazopanib maintenance had a detrimental effect on median progression-free survival versus placebo in East Asian patients from the combined studies (n = 354; 17.9 vs 21.5 months; hazard ratio, 1.114; 95% confidence interval, 0.818–1.518; P = 0.4928). Pazopanib maintenance showed a disadvantage in overall survival in East Asian patients from AGO-OVAR16 versus placebo (hazard ratio, 1.706; 95% confidence interval, 1.010–2.883; P = 0.0465); overall survival analysis was not performed in the East Asian study because of insufficient event numbers. Pazopanib-treated patients had a significantly higher incidence of grade 3 or higher hypertension (27%) and neutropenia (13%) versus placebo.ConclusionsThe treatment effect of maintenance pazopanib in East Asian patients seemed to differ from that in non-Asian patients. In study-specific and pooled analyses, none of the potential factors analyzed could satisfactorily explain the different efficacy results of pazopanib in East Asian patients.
Collapse
|
39
|
Al-Hader A, Al-Rohil RN, Han H, Von Hoff D. Pancreatic acinar cell carcinoma: A review on molecular profiling of patient tumors. World J Gastroenterol 2017; 23:7945-7951. [PMID: 29259370 PMCID: PMC5725289 DOI: 10.3748/wjg.v23.i45.7945] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/17/2017] [Accepted: 10/26/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic carcinomas with acinar differentiation are rare, accounting for 1%-2% of adult pancreatic tumors; they include pancreatic acinar cell carcinoma (PACC), pancreatoblastoma, and carcinomas of mixed differentiation. Patients with PACC have a prognosis better than pancreatic ductal adenocarcinomas but worse than pancreatic neuroendocrine tumors. Reports of overall survival range from 18 to 47 mo. A literature review on PACCs included comprehensive genomic profiling and whole exome sequencing on a series of more than 70 patients as well as other diagnostic studies including immunohistochemistry. Surgical resection of PACC is the preferred treatment for localized and resectable tumors. The efficacy of adjuvant treatment is unclear. Metastatic PACCs are generally not curable and treated with systemic chemotherapy. They are moderately responsive to chemotherapy with different regimens showing various degrees of response in case reports/series. Most of these regimens were developed to treat patients with pancreatic ductal adenocarcinomas or colorectal adenocarcinomas. Review of PACC's molecular profiling showed a number of gene alterations such as: SMAD4, BRAF, BRCA2, TP53, RB1, MEN1, JAK-1, BRCA-1, BRCA-2, and DNA mismatch repair abnormalities. PACCs had multiple somatic mutations with some targetable with available drugs. Therefore, molecular profiling of PACC should be an option for patients with refractory PACC.
Collapse
Affiliation(s)
- Ahmad Al-Hader
- Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202-3082, United States
| | - Rami N Al-Rohil
- Department of Pathology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Haiyong Han
- Molecular Medicine Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, United States
| | - Daniel Von Hoff
- Molecular Medicine Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, United States
| |
Collapse
|
40
|
Kim JY, Yu JH, Nam SJ, Kim SW, Lee SK, Park WY, Noh DY, Nam DH, Park YH, Han W, Lee JE. Genetic and Clinical Characteristics of Phyllodes Tumors of the Breast. Transl Oncol 2017; 11:18-23. [PMID: 29145046 PMCID: PMC5684533 DOI: 10.1016/j.tranon.2017.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/12/2017] [Accepted: 10/23/2017] [Indexed: 12/11/2022] Open
Abstract
PURPOSE: Phyllodes tumors (PTs) of the breast are rare, accounting for less than 1% of all breast tumors. Among PTs, malignant PTs (MPTs) have malignant characteristics and distant metastases occur in about 20% to 30% of MPTs. However, there is no effective treatment for MPTs with distant metastasis, resulting in an abject prognosis. We performed targeted deep sequencing on PTs to identify the associations between genetic alterations and clinical prognosis. METHODS: We performed targeted deep sequencing to evaluate the genetic characteristics of PTs and analyzed the relationships between clinical and genetic characteristics. RESULTS: A total of 17 PTs were collected between 2001 and 2012. Histologic review was performed by pathologists. The samples included three benign PTs, one borderline PT, and 13 MPTs. The most frequently detected genetic alteration occurred in the TERT promoter region (70.6%), followed by MED12 (64.7%). EGFR amplification and TP53 alteration were detected in four MPTs without genetic alterations in MED12 and TERT promoter regions. Genetic alterations of RARA and ZNF703 were repeatedly found in PTs with local recurrence, and genetic alterations of SETD2, BRCA2, and TSC1 were detected in PTs with distant metastasis. Especially, MPT harboring PTEN and RB1 copy number deletion showed rapid disease progression. CONCLUSIONS: In this study, we provide genetic characterization and potential therapeutic target for this rare, potentially lethal disease. Further large-scale comprehensive genetic study and functional validation are warranted.
Collapse
Affiliation(s)
- Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,06351, Korea
| | - Jong Han Yu
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Seok Jin Nam
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Seok Won Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Se Kyung Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, 06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Korea; Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea; Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Korea; Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Jeong Eon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Korea; Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea.
| |
Collapse
|
41
|
Lanz HL, Saleh A, Kramer B, Cairns J, Ng CP, Yu J, Trietsch SJ, Hankemeier T, Joore J, Vulto P, Weinshilboum R, Wang L. Therapy response testing of breast cancer in a 3D high-throughput perfused microfluidic platform. BMC Cancer 2017; 17:709. [PMID: 29096610 PMCID: PMC5668957 DOI: 10.1186/s12885-017-3709-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 10/27/2017] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Breast cancer is the most common invasive cancer among women. Currently, there are only a few models used for therapy selection, and they are often poor predictors of therapeutic response or take months to set up and assay. In this report, we introduce a microfluidic OrganoPlate® platform for extracellular matrix (ECM) embedded tumor culture under perfusion as an initial study designed to investigate the feasibility of adapting this technology for therapy selection. METHODS The triple negative breast cancer cell lines MDA-MB-453, MDA-MB-231 and HCC1937 were selected based on their different BRCA1 and P53 status, and were seeded in the platform. We evaluate seeding densities, ECM composition (Matrigel®, BME2rgf, collagen I) and biomechanical (perfusion vs static) conditions. We then exposed the cells to a series of anti-cancer drugs (paclitaxel, olaparib, cisplatin) and compared their responses to those in 2D cultures. Finally, we generated cisplatin dose responses in 3D cultures of breast cancer cells derived from 2 PDX models. RESULTS The microfluidic platform allows the simultaneous culture of 96 perfused micro tissues, using limited amounts of material, enabling drug screening of patient-derived material. 3D cell culture viability is improved by constant perfusion of the medium. Furthermore, the drug response of these triple negative breast cancer cells was attenuated by culture in 3D and differed from that observed in 2D substrates. CONCLUSIONS We have investigated the use of a high-throughput organ-on-a-chip platform to select therapies. Our results have raised the possibility to use this technology in personalized medicine to support selection of appropriate drugs and to predict response to therapy in a real time fashion.
Collapse
Affiliation(s)
| | - Anthony Saleh
- Mimetas BV, Leiden, The Netherlands
- NIH, Bethesda, Maryland USA
| | | | | | | | - Jia Yu
- Mayo Clinic, Rochester, Minnesota USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Ohmoto A, Yachida S. Current status of poly(ADP-ribose) polymerase inhibitors and future directions. Onco Targets Ther 2017; 10:5195-5208. [PMID: 29138572 PMCID: PMC5667784 DOI: 10.2147/ott.s139336] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Inhibitors of poly(ADP-ribose) polymerases (PARPs), which play a key role in DNA damage/repair pathways, have been developed as antitumor agents based on the concept of synthetic lethality. Synthetic lethality is the idea that cell death would be efficiently induced by simultaneous loss of function of plural key molecules, for example, by exposing tumor cells with inactivating gene mutation of BRCA-mediated DNA repair to chemically induced inhibition of PARPs. Indeed, three PARP inhibitors, olaparib, rucaparib and niraparib have already been approved in the US or Europe, mainly for the treatment of BRCA-mutant ovarian cancer. Clinical trials of various combinations of PARP inhibitors with cytotoxic or molecular-targeted agents are also underway. In particular, expanded applications of PARP inhibitors are anticipated following recent reports that defects in homologous recombination repair (HRR) are associated with mutations in repair genes other than BRCA1/BRCA2, such as ATM, ATR, PALB2, RAD51, CHEK1 and CHEK2, as well as with epigenetic loss of BRCA1 function through promoter methylation or overexpression of the BRCA2-interacting transcriptional repressor EMSY. Current topics of interest include selection of the best agent in each clinical context, identification of new treatment targets for HRR-proficient cases, and development of PARP inhibitor-based regimens that are less toxic and that prolong overall survival as well as progression-free survival. In addition, potential long-term side effects and suitable biomarkers for predicting efficacy and mechanisms of clinical resistance are in discussion. This review summarizes representative preclinical and clinical data for PARP inhibitors and discusses their potential for future applications to treat various malignancies.
Collapse
Affiliation(s)
- Akihiro Ohmoto
- Laboratory of Clinical Genomics, National Cancer Center Research Institute, Tokyo
| | - Shinichi Yachida
- Laboratory of Clinical Genomics, National Cancer Center Research Institute, Tokyo.,Department of Cancer Genome Informatics, Graduate School of Medicine, Faculty of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
43
|
"Back to a false normality": new intriguing mechanisms of resistance to PARP inhibitors. Oncotarget 2017; 8:23891-23904. [PMID: 28055979 PMCID: PMC5410353 DOI: 10.18632/oncotarget.14409] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 12/23/2016] [Indexed: 01/07/2023] Open
Abstract
Several evidences have shown that BRCA mutations increased tumor-cells sensitivity to PARP inhibitors by synthetic lethality leading to an accelerated development of several compounds targeting the PARP enzymes system as anticancer agents for clinical setting. Most of such compounds have been investigated in ovarian and breast cancer, showing promising efficacy in BRCA-mutated patients. Recently clinical studies of PARP-inhibitors have been extended across different tumor types harboring BRCA-mutations, including also “BRCA-like” sporadic tumors with homologous recombination deficiency (HRD). This review summarizes the biological background underlying PARP-inhibition, reporting the results of the most relevant clinical trials carried out in patients treated with PARP inhibitors alone or in combination with chemotherapy. Molecular mechanisms responsible for the occurrence of both primary and acquired resistance have been elucidated, in order to support the development of new treatment strategies.
Collapse
|
44
|
Abstract
Approximately half of high-grade serous epithelial ovarian cancers incur alterations in genes of homologous recombination (BRCA1, BRCA2, RAD51C, Fanconi anemia genes), and the rest incur alterations in other DNA repair pathways at high frequencies. Such cancer-specific gene alterations can confer selective sensitivity to DNA damaging agents such as cisplatin and carboplatin, topotecan, etoposide, doxorubicin, and gemcitabine. Originally presumed to inhibit DNA repair, PARP inhibitors that have recently been approved by the FDA for the treatment of advanced ovarian cancer also act as DNA damaging agents by inducing PARP-DNA complexes. These DNA damaging agents induce different types of DNA lesions that require various DNA repair genes for the repair, but commonly induce replication fork slowing or stalling, also referred to as replication stress. Replication stress activates DNA repair checkpoint proteins (ATR, CHK1), which prevent further DNA damage. Hence, targeting DNA repair genes or DNA repair checkpoint genes augments the anti-tumor activity of DNA damaging agents. This review describes the rational basis for using DNA repair and DNA repair checkpoint inhibitors as single agents. The review also presents the strategies combining these inhibitors with DNA damaging agents for ovarian cancer therapy based on specific gene alterations.
Collapse
|
45
|
Lee JM, Cimino-Mathews A, Peer CJ, Zimmer A, Lipkowitz S, Annunziata CM, Cao L, Harrell MI, Swisher EM, Houston N, Botesteanu DA, Taube JM, Thompson E, Ogurtsova A, Xu H, Nguyen J, Ho TW, Figg WD, Kohn EC. Safety and Clinical Activity of the Programmed Death-Ligand 1 Inhibitor Durvalumab in Combination With Poly (ADP-Ribose) Polymerase Inhibitor Olaparib or Vascular Endothelial Growth Factor Receptor 1-3 Inhibitor Cediranib in Women's Cancers: A Dose-Escalation, Phase I Study. J Clin Oncol 2017; 35:2193-2202. [PMID: 28471727 DOI: 10.1200/jco.2016.72.1340] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Data suggest that DNA damage by poly (ADP-ribose) polymerase inhibition and/or reduced vascular endothelial growth factor signaling by vascular endothelial growth factor receptor inhibition may complement antitumor activity of immune checkpoint blockade. We hypothesize the programmed death-ligand 1 (PD-L1) inhibitor, durvalumab, olaparib, or cediranib combinations are tolerable and active in recurrent women's cancers. Patients and Methods This phase I study tested durvalumab doublets in parallel 3 + 3 dose escalations. Durvalumab was administered at 10 mg/kg every 2 weeks or 1,500 mg every 4 weeks with either olaparib tablets twice daily or cediranib on two schedules. The primary end point was the recommended phase II dose (RP2D). Response rate and pharmacokinetic analysis were secondary end points. Results Between June 2015 and May 2016, 26 women were enrolled. The RP2D was durvalumab 1,500 mg every 4 weeks with olaparib 300 mg twice a day, or cediranib 20 mg, 5 days on/2 days off. No dose-limiting toxicity was recorded with durvalumab plus olaparib. The cediranib intermittent schedule (n = 6) was examined because of recurrent grade 2 and non-dose-limiting toxicity grade 3 and 4 adverse events (AEs) on the daily schedule (n = 8). Treatment-emergent AEs included hypertension (two of eight), diarrhea (two of eight), pulmonary embolism (two of eight), pulmonary hypertension (one of eight), and lymphopenia (one of eight). Durvalumab plus intermittent cediranib grade 3 and 4 AEs were hypertension (one of six) and fatigue (one of six). Exposure to durvalumab increased cediranib area under the curve and maximum plasma concentration on the daily, but not intermittent, schedules. Two partial responses (≥15 months and ≥ 11 months) and eight stable diseases ≥ 4 months (median, 8 months [4 to 14.5 months]) were seen in patients who received durvalumab plus olaparib, yielding an 83% disease control rate. Six partial responses (≥ 5 to ≥ 8 months) and three stable diseases ≥ 4 months (4 to ≥ 8 months) were seen in 12 evaluable patients who received durvalumab plus cediranib, for a 50% response rate and a 75% disease control rate. Response to therapy was independent of PD-L1 expression. Conclusion To our knowledge, this is the first reported anti-PD-L1 plus olaparib or cediranib combination therapy. The RP2Ds of durvalumab plus olaparib and durvalumab plus intermittent cediranib are tolerable and active. Phase II studies with biomarker evaluation are ongoing.
Collapse
Affiliation(s)
- Jung-Min Lee
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Ashley Cimino-Mathews
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Cody J Peer
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Alexandra Zimmer
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Stanley Lipkowitz
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Christina M Annunziata
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Liang Cao
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Maria I Harrell
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Elizabeth M Swisher
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Nicole Houston
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Dana-Adriana Botesteanu
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Janis M Taube
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Elizabeth Thompson
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Aleksandra Ogurtsova
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Haiying Xu
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Jeffers Nguyen
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Tony W Ho
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - William D Figg
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| | - Elise C Kohn
- Jung-Min Lee, Cody J. Peer, Alexandra Zimmer, Stanley Lipkowitz, Christina M. Annunziata, Liang Cao, Nicole Houston, Dana-Adriana Botesteanu, Jeffers Nguyen, William D. Figg, and Elise C. Kohn, Center for Cancer Research, National Cancer Institute, Bethesda; Ashley Cimino-Mathews, Janis M. Taube, Elizabeth Thompson, Aleksandra Ogurtsova, and Haiying Xu, The Johns Hopkins Medical Institution, Baltimore; and Tony W. Ho, AstraZeneca, Gaithersburg, MD; and Maria I. Harrell and Elizabeth M. Swisher, University of Washington, Seattle, WA
| |
Collapse
|
46
|
Konecny GE, Kristeleit RS. PARP inhibitors for BRCA1/2-mutated and sporadic ovarian cancer: current practice and future directions. Br J Cancer 2016; 115:1157-1173. [PMID: 27736844 PMCID: PMC5104889 DOI: 10.1038/bjc.2016.311] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 08/02/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors cause targeted tumour cell death in homologous recombination (HR)-deficient cancers, including BRCA-mutated tumours, by exploiting synthetic lethality. PARP inhibitors are being evaluated in late-stage clinical trials of ovarian cancer (OC). Recently, olaparib was the first PARP inhibitor approved in the European Union and United States for the treatment of advanced BRCA-mutated OC. This paper reviews the role of BRCA mutations for tumorigenesis and PARP inhibitor sensitivity, and summarises the clinical development of PARP inhibitors for the treatment of patients diagnosed with OC. Among the five key PARP inhibitors currently in clinical development, olaparib has undergone the most extensive clinical investigation. PARP inhibitors have demonstrated durable antitumour activity in BRCA-mutated advanced OC as a single agent in the treatment and maintenance setting, particularly in platinum-sensitive disease. PARP inhibitors are well tolerated; however, further careful assessment of moderate and late-onset toxicity is mandatory in the maintenance and adjuvant setting, respectively. PARP inhibitors are also being evaluated in combination with chemotherapeutic and novel targeted agents to potentiate antitumour activities. Current research is extending the use of PARP inhibitors beyond BRCA mutations to other sensitising molecular defects that result in HR-deficient cancer, and is defining an HR-deficiency signature. Trials are underway to determine whether such a signature will predict sensitivity to PARP inhibitors in women with sporadic OC.
Collapse
Affiliation(s)
- G E Konecny
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, 2825 Santa Monica Blvd., Suite 200, Santa Monica, CA 90404–2429, USA
| | - R S Kristeleit
- Department of Oncology, University College London Cancer Institute, University College London, Paul Gorman Building, Huntley Street, London, WC1E 6BT, UK
| |
Collapse
|
47
|
Les actualités marquantes du congrès Targeted Anticancer Therapies — TAT 2016. ONCOLOGIE 2016. [DOI: 10.1007/s10269-016-2651-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
48
|
Rabenau K, Hofstatter E. DNA Damage Repair and the Emerging Role of Poly(ADP-ribose) Polymerase Inhibition in Cancer Therapeutics. Clin Ther 2016; 38:1577-88. [PMID: 27368114 DOI: 10.1016/j.clinthera.2016.06.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 11/19/2022]
Abstract
PURPOSE As a result of improved understanding of DNA repair mechanisms, poly(ADP-ribose) polymerase inhibitors (PARPi) are increasingly recognized to play an important therapeutic role in the treatment of cancer. The aim of this article is to provide a review of PARPi function in DNA damage repair and synthetic lethality and to demonstrate how these mechanisms can be exploited to provide new PARPi-based therapies to patients with solid tumors. METHODS Literature from a range of sources, including PubMed and MEDLINE, were searched to identify recent reports regarding DNA damage repair and PARPi. FINDINGS DNA damage repair is central to cellular viability. The family of poly(ADP-ribose) polymerase proteins play multiple intracellular roles in DNA repair, but function primarily in the resolution of repair of single-strand DNA breaks. Insights through the discovery of germline BRCA1/2 mutations led to the understanding of synthetic lethality and the potential therapeutic role of PARPi in the treatment of cancer. Further understanding of DNA damage repair and the concept of BRCA-like tumors have catalyzed PARPi clinical investigation in multiple oncologic settings. IMPLICATIONS PARPi hold great promise in the treatment of solid tumors, both as monotherapy and in combination with other cancer therapeutics. Multiple PARPi clinical trials are currently underway. Further understanding of aberrant DNA repair mechanisms in the germline and in the tumor genome will allow clinicians and researchers to apply PARPi most strategically in the era of personalized medicine.
Collapse
Affiliation(s)
- Karen Rabenau
- Yale University School of Medicine/Yale Cancer Center, New Haven, CT
| | - Erin Hofstatter
- Yale University School of Medicine/Yale Cancer Center, New Haven, CT.
| |
Collapse
|
49
|
Jenner ZB, Sood AK, Coleman RL. Evaluation of rucaparib and companion diagnostics in the PARP inhibitor landscape for recurrent ovarian cancer therapy. Future Oncol 2016; 12:1439-56. [PMID: 27087632 PMCID: PMC4976841 DOI: 10.2217/fon-2016-0002] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/04/2016] [Indexed: 12/28/2022] Open
Abstract
Rucaparib camsylate (CO-338; 8-fluoro-2-{4-[(methylamino)methyl]phenyl}-1,3,4,5-tetrahydro-6H-azepino[5,4,3-cd]indol-6-one ((1S,4R)-7,7-dimethyl-2-oxobicyclo[2.2.1]hept-1-yl)methanesulfonic acid salt) is a PARP1, 2 and 3 inhibitor. Phase I studies identified a recommended Phase II dose of 600 mg orally twice daily. ARIEL2 Part 1 established a tumor genomic profiling test for homologous recombination loss of heterozygosity quantification using a next-generation sequencing companion diagnostic (CDx). Rucaparib received US FDA Breakthrough Therapy designation for treatment of platinum-sensitive BRCA-mutated advanced ovarian cancer patients who received greater than two lines of platinum-based therapy. Comparable to rucaparib development, other PARP inhibitors, such as olaparib, niraparib, veliparib and talazoparib, are developing CDx tests for targeted therapy. PARP inhibitor clinical trials and CDx assays are discussed in this review, as are potential PARP inhibitor combination therapies and likely resistance mechanisms.
Collapse
Affiliation(s)
- Zachary B Jenner
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- McGovern Medical School, formerly The University of Texas Health Science Center at Houston (UTHealth) Medical School, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
50
|
Lim D, Ngeow J. Evaluation of the methods to identify patients who may benefit from PARP inhibitor use. Endocr Relat Cancer 2016; 23:R267-85. [PMID: 27226207 DOI: 10.1530/erc-16-0116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 05/23/2016] [Indexed: 12/17/2022]
Abstract
The effectiveness of poly (ADP-ribose) polymerase inhibitors (PARPi) in treating cancers associated with BRCA1/2 mutations hinges upon the concept of synthetic lethality and exemplifies the principles of precision medicine. Currently, most clinical trials are recruiting patients based on pathological subtypes or have included BRCA mutation analysis (germ line and/or somatic) as part of the selection criteria. Mounting evidence, however, suggests that these drugs may also be efficacious in tumors with defects in other genes involved in the homologous recombination repair pathway. Advances in molecular profiling techniques together with increased research efforts have led to a better understanding of the molecular aberrations underlying this BRCA-like phenotype and helped broaden the concept of BRCAness. Hence, it is likely that the list of predictive biomarkers for PARPi therapy will increase in future. There is currently no gold standard method of testing for PARPi response and no universal guidelines are in place on how to incorporate biomarker testing into routine clinical diagnostics. In this review, we explore the concept of BRCAness and highlight the different methods that have been used to identify patients who may benefit from the use of these anticancer agents. The identification of predictive biomarkers is crucial in improving patient selection and expanding the clinical applications of PARPi therapy.
Collapse
Affiliation(s)
- Diana Lim
- Department of PathologyNational University Health System, Singapore, Singapore
| | - Joanne Ngeow
- Lee Kong Chian School of MedicineNanyang Technological University, Singapore, Singapore Cancer Genetics ServiceDivision of Medical Oncology, National Cancer Centre, Singapore, Singapore
| |
Collapse
|