351
|
La Belle A, Khatib J, Schiemann WP, Vinayak S. Role of Platinum in Early-Stage Triple-Negative Breast Cancer. Curr Treat Options Oncol 2017; 18:68. [PMID: 29110096 DOI: 10.1007/s11864-017-0506-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OPINION STATEMENT Triple-negative breast cancer (TNBC) is both a clinically and genomically heterogeneous disease, with distinct molecular subtypes; however, most epidemiologic and clinical studies to date have defined it under a "one disease" umbrella. This is an important point, since one therapeutic approach for all TNBCs is unlikely to be successful given the underlying biological diversity. In this review, we explore the role of platinums in the treatment of TNBC, as well as the potential for biomarkers to predict patient response to these agents. The results of neoadjuvant TNBC trials, with addition of platinum to anthracycline/taxane-based chemotherapies, have been very encouraging given increases in pathologic complete response (pCR) rates. However, we do not have any evidence yet that these agents would lead to improvement in disease-free and overall survival. Moreover, addition of platinums increases toxicity and can compromise current standard chemotherapy doses, which further impedes their use in all TNBC patients. Therefore, the addition of platinums to standard chemotherapy should be used with caution and in discussion with patients after a careful assessment of risks and benefits. Clinical trials addressing the role of platinums in TNBC further remain of significant value.
Collapse
Affiliation(s)
- Alyssa La Belle
- Case Comprehensive Cancer Center, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH, 44106, USA
| | - Jude Khatib
- Department of Internal Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH, 44106, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH, 44106, USA.
| | - Shaveta Vinayak
- Case Comprehensive Cancer Center, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH, 44106, USA. .,Division of Hematology and Oncology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, LKS 5079, Cleveland, OH, 44106, USA.
| |
Collapse
|
352
|
Abstract
Triple negative is a term applied to breast cancers that do not meaningfully express the estrogen or progesterone hormone receptors or overexpress the human epidermal growth factor receptor 2 tyrosine kinase. At present, the only proven method for systemic management of triple-negative breast cancer for both early-stage and metastatic settings is cytotoxic chemotherapy. Here, we provide a comprehensive review of management strategies that are best supported by available data. We also review recent advances most likely to affect treatment of triple-negative breast cancer in the coming years with particular emphasis on targeted agents, biologics, and immunotherapy.
Collapse
Affiliation(s)
- Vijayakrishna K Gadi
- University of Washington, Fred Hutchinson Cancer Research Center, and Seattle Cancer Care Alliance, Seattle, WA
| | - Nancy E Davidson
- University of Washington, Fred Hutchinson Cancer Research Center, and Seattle Cancer Care Alliance, Seattle, WA
| |
Collapse
|
353
|
Jhan JR, Andrechek ER. Triple-negative breast cancer and the potential for targeted therapy. Pharmacogenomics 2017; 18:1595-1609. [PMID: 29095114 PMCID: PMC5694022 DOI: 10.2217/pgs-2017-0117] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is composed of several well-recognized subtypes including estrogen receptor, progesterone receptor and HER2 triple-negative breast cancer (TNBC). Without available targeted therapy options, standard of care for TNBC remains chemotherapy. It is of interest to note that TNBC tumors generally have better responses to chemotherapy compared with other subtypes. However, patients without complete response account for approximately 80% of TNBC. Mounting evidence suggests significant heterogeneity within the TNBC subtype, and studies have focused on genetic targets with high rates of altered expression. Recent studies suggest clear possibilities for benefits from targeted therapy in TNBC. In this review, we summarize studies of targeted therapy, including within mouse models, and discuss their applications in the development of combinatorial treatments to treat TNBC.
Collapse
Affiliation(s)
- Jing-Ru Jhan
- Department of Physiology, Michigan State University, 2194 Biomedical Physical Sciences Building, 567 Wilson Rd., East Lansing, MI 48824, USA
| | - Eran R Andrechek
- Department of Physiology, Michigan State University, 2194 Biomedical Physical Sciences Building, 567 Wilson Rd., East Lansing, MI 48824, USA
| |
Collapse
|
354
|
Milette S, Sicklick JK, Lowy AM, Brodt P. Molecular Pathways: Targeting the Microenvironment of Liver Metastases. Clin Cancer Res 2017; 23:6390-6399. [PMID: 28615370 PMCID: PMC5668192 DOI: 10.1158/1078-0432.ccr-15-1636] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/27/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Abstract
Curative treatment for metastatic solid cancers remains elusive. The liver, which is nourished by a rich blood supply from both the arterial and portal venous systems, is the most common site of visceral metastases, particularly from cancers arising in the gastrointestinal tract, with colorectal cancer being the predominant primary site in Western countries. A mounting body of evidence suggests that the liver microenvironment (LME) provides autocrine and paracrine signals originating from both parenchymal and nonparenchymal cells that collectively create both pre- and prometastatic niches for the development of hepatic metastases. These resident cells and their molecular mediators represent potential therapeutic targets for the prevention and/or treatment of liver metastases (LM). This review summarizes: (i) the current therapeutic options for treating LM, with a particular focus on colorectal cancer LM; (ii) the role of the LME in LM at each of its phases; (iii) potential targets in the LME identified through preclinical and clinical investigations; and (iv) potential therapeutic approaches for targeting elements of the LME before and/or after the onset of LM as the basis for future clinical trials. Clin Cancer Res; 23(21); 6390-9. ©2017 AACR.
Collapse
Affiliation(s)
- Simon Milette
- Departments of Surgery, Medicine, and Oncology McGill University and the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jason K. Sicklick
- Division of Surgical Oncology, Department of Surgery, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Andrew M. Lowy
- Division of Surgical Oncology, Department of Surgery, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Pnina Brodt
- Departments of Surgery, Medicine, and Oncology McGill University and the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
355
|
Simon R. Critical Review of Umbrella, Basket, and Platform Designs for Oncology Clinical Trials. Clin Pharmacol Ther 2017; 102:934-941. [PMID: 28795401 DOI: 10.1002/cpt.814] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/14/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022]
Abstract
The successful development of new drugs with a companion diagnostic based on genomic alteration of an oncogene has led to rethinking of all phases on clinical development of cancer drugs. We critically review some of the new clinical trial designs for biomarker-based cancer drug development. We try to clarify the objectives of the new designs and examine completed trials using these designs to evaluate what has been learned about these designs.
Collapse
|
356
|
Tang S, Pan H, Wei W, Yang H, Liu J, Yang R. GOLPH3: a novel biomarker that correlates with poor survival and resistance to chemotherapy in breast cancer. Oncotarget 2017; 8:105155-105169. [PMID: 29285241 PMCID: PMC5739628 DOI: 10.18632/oncotarget.21927] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/04/2017] [Indexed: 12/02/2022] Open
Abstract
The association between Golgi phosphoprotein 3 (GOLPH3) and clinical pathological characteristics, as well as the clinical outcomes of both neoadjuvant and adjuvant chemotherapies in breast cancer, remain largely unknown. In this study, we investigated the biological role and clinical significance of GOLPH3 in breast cancer. We found that GOLPH3 expression in tumor tissue was higher than that in adjacent noncancerous tissue (ANT) and fibroadenoma. GOLPH3 silencing reduced the migration, invasion, and proliferation of breast cancer cells and promoted apoptosis of the cells. Importantly, patients with high GOLPH3 expression had worse disease-free survival (DFS) and overall survival (OS), and GOLPH3 expression was correlated with clinical pathological characteristics such as molecular subtype, tumor-node-metastasis classification, and age but was not associated with surgery type. Patients with high GOLPH3 expression had poor DFS and OS in every molecular subtype, and an increase in tumor invasion and lymph node metastasis. The risk of recurrence increased with age in patients with high GOLPH3 expression, and surgery type had no influence on patient survival. This is the first study to investigate the correlation between GOLPH3 and response to chemotherapy in breast cancer. Patients with high GOLPH3 expression showed resistance to neoadjuvant and adjuvant chemotherapies, and GOLPH3 overexpression indicated a high risk of recurrence in patients who received adjuvant chemotherapy. These data suggest that GOLPH3 may be a novel biomarker that correlates with poor survival and resistance to chemotherapy in breast cancer.
Collapse
Affiliation(s)
- Shicong Tang
- Department of Breast Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Hong Pan
- Department of Thoracic Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Wei Wei
- Department of Breast Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Huawei Yang
- Department of Breast Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jianlun Liu
- Department of Breast Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Rirong Yang
- Department of Immunology, School of Preclinical Medicine, Guangxi Medical University, Guangxi, People's Republic of China
| |
Collapse
|
357
|
Zhou JX, Feng LJ, Zhang X. Risk of severe hematologic toxicities in cancer patients treated with PARP inhibitors: a meta-analysis of randomized controlled trials. Drug Des Devel Ther 2017; 11:3009-3017. [PMID: 29075104 PMCID: PMC5648323 DOI: 10.2147/dddt.s147726] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Purpose Hematologic toxicities, including neutropenia, thrombocytopenia, and anemia, are major adverse effects of PARP inhibitors (PARPis), but the incidence rate and overall risk has not been systematically studied. Therefore, we conducted a meta-analysis of published clinical trials to investigate the incidence and relative risks (RRs) of severe (high-grade) hematologic events in cancer patients treated with PARPis. Methods PubMed, Embase, and oncology conference proceedings were searched for relevant studies. Eligible studies were Phase II and III randomized controlled trials (RCTs) of PARPis in cancer patients with adequate safety data on hematologic toxicities. The summary incidence, RRs, and 95% confidence intervals (CIs) were calculated. Results A total of 2,479 patients from 12 RCTs revealed that the incidence of PARPi-associated severe hematologic toxicities was, respectively: neutropenia: 32.9% (95% CI, 20.5%–48.3%); thrombocytopenia: 15.9% (95% CI, 9.5%–25.4%), and anemia: 9.1% (95% CI, 5.1%–15.7%). Olaparib was associated with an increased risk of severe neutropenia. Veliparib was associated with an increased risk of severe neutropenia and thrombocytopenia. Niraparib was associated with an increased risk of severe thrombocytopenia, anemia, and neutropenia. When stratified by combination therapy, significantly increased risk of hematologic toxicities was observed for patients treated with PARPis monotherapy and PARPis combined with single-agent chemotherapy. Conclusion Treatment with PARPis olaparib, veliparib, and niraparib is associated with a significant increase in the risk of hematologic toxicities in cancer patients, and frequent clinical monitoring should be emphasized when managing these PARPis.
Collapse
Affiliation(s)
- Jian Xin Zhou
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing
| | - Li Jin Feng
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai
| | - Xi Zhang
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
358
|
Alternative Splicing in Breast Cancer and the Potential Development of Therapeutic Tools. Genes (Basel) 2017; 8:genes8100217. [PMID: 28981467 PMCID: PMC5664086 DOI: 10.3390/genes8100217] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Alternative splicing is a key molecular mechanism now considered as a hallmark of cancer that has been associated with the expression of distinct isoforms during the onset and progression of the disease. The leading cause of cancer-related deaths in women worldwide is breast cancer, and even when the role of alternative splicing in this type of cancer has been established, the function of this mechanism in breast cancer biology is not completely decoded. In order to gain a comprehensive view of the role of alternative splicing in breast cancer biology and development, we summarize here recent findings regarding alternative splicing events that have been well documented for breast cancer evolution, considering its prognostic and therapeutic value. Moreover, we analyze how the response to endocrine and chemical therapies could be affected due to alternative splicing and differential expression of variant isoforms. With all this knowledge, it becomes clear that targeting alternative splicing represents an innovative approach for breast cancer therapeutics and the information derived from current studies could guide clinical decisions with a direct impact in the clinical advances for breast cancer patients nowadays.
Collapse
|
359
|
Wang S, Yang FJ, Wang X, Zhou Y, Dai B, Han B, Ma HC, Ding YT, Shi XL. PARP-1 promotes tumor recurrence after warm ischemic liver graft transplantation via neutrophil recruitment and polarization. Oncotarget 2017; 8:88918-88933. [PMID: 29179487 PMCID: PMC5687657 DOI: 10.18632/oncotarget.21493] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/26/2017] [Indexed: 12/21/2022] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP-1) is a crucial contributor to exacerbate ischemia and reperfusion (IR) injury and cancer process. However, there is little research into whether PARP-1 affects the hepatocellular carcinoma (HCC) recurrence after liver transplantation. In this study, we investigated the influence of PARP-1 on hepatic neutrophil mobilizing and phenotype shifting which may lead to HCC recurrence after liver transplantation. We found that rats received the grafts with warm ischemic injury had higher risk of HCC recurrence, which was markedly prevented by pharmacological inhibition of PARP-1 after liver transplantation. In mouse models, the up-regulation of PARP-1 was closely related to the greater tumor burden and increased hepatic susceptibility to recurrence after IR injury. The reason was that high hepatic PARP-1 led to increased liver CXCL1 levels, which in turn promoted recruitment of neutrophils. Both blocking CXCL1/CXCR2 signaling pathway and depleting neutrophils decreased tumor burden. Moreover, these infiltrating neutrophils were programmed to a proangiogenic phenotype under the influence of PARP-1 in vivo after hepatic IR injury. In conclusion, IR-induced PARP-1 up-regulation increased the hepatic recruitment of neutrophils through regulation of CXCL1/CXCR2 signaling and polarized hepatic neutrophils to proangiogenic phenotype, which further promoted HCC recurrence after transplantation.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Fa-Ji Yang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xun Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuan Zhou
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Bo Dai
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Bing Han
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hu-Cheng Ma
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yi-Tao Ding
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiao-Lei Shi
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
360
|
Marotti JD, de Abreu FB, Wells WA, Tsongalis GJ. Triple-Negative Breast Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2133-2138. [DOI: 10.1016/j.ajpath.2017.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/01/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023]
|
361
|
Senft D, Leiserson MDM, Ruppin E, Ronai ZA. Precision Oncology: The Road Ahead. Trends Mol Med 2017; 23:874-898. [PMID: 28887051 PMCID: PMC5718207 DOI: 10.1016/j.molmed.2017.08.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 02/06/2023]
Abstract
Current efforts in precision oncology largely focus on the benefit of genomics-guided therapy. Yet, advances in sequencing techniques provide an unprecedented view of the complex genetic and nongenetic heterogeneity within individual tumors. Herein, we outline the benefits of integrating genomic and transcriptomic analyses for advanced precision oncology. We summarize relevant computational approaches to detect novel drivers and genetic vulnerabilities, suitable for therapeutic exploration. Clinically relevant platforms to functionally test predicted drugs/drug combinations for individual patients are reviewed. Finally, we highlight the technological advances in single cell analysis of tumor specimens. These may ultimately lead to the development of next-generation cancer drugs, capable of tackling the hurdles imposed by genetic and phenotypic heterogeneity on current anticancer therapies.
Collapse
Affiliation(s)
- Daniela Senft
- Tumor Initiation and Maintenance Program, NCI designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Mark D M Leiserson
- Microsoft Research New England, Cambridge, MA 02142, USA; Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Eytan Ruppin
- School of Computer Sciences and Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel; Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA
| | - Ze'ev A Ronai
- Tumor Initiation and Maintenance Program, NCI designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; Technion Integrated Cancer Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, 31096, Israel.
| |
Collapse
|
362
|
Barnard K, Klimberg VS. An Update on Randomized Clinical Trials in Breast Cancer. Surg Oncol Clin N Am 2017; 26:587-620. [DOI: 10.1016/j.soc.2017.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
363
|
Chagpar AB. On baseball and breast cancer. Am J Surg 2017; 215:353-356. [PMID: 28958652 DOI: 10.1016/j.amjsurg.2017.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 10/18/2022]
Abstract
Dedicated to the memory of Dr. William Hunter Harridge and Nonie Lowry, this manuscript discusses the ups and downs associated with novel therapies in breast cancer, and applies the lessons of persistence learned from baseball to our quest for more optimal treatments, particularly in triple negative breast cancer.
Collapse
Affiliation(s)
- Anees B Chagpar
- Department of Surgery, Yale University School of Medicine, United States.
| |
Collapse
|
364
|
Hassan S, Esch A, Liby T, Gray JW, Heiser LM. Pathway-Enriched Gene Signature Associated with 53BP1 Response to PARP Inhibition in Triple-Negative Breast Cancer. Mol Cancer Ther 2017; 16:2892-2901. [PMID: 28958991 DOI: 10.1158/1535-7163.mct-17-0170] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/29/2017] [Accepted: 09/18/2017] [Indexed: 12/30/2022]
Abstract
Effective treatment of patients with triple-negative (ER-negative, PR-negative, HER2-negative) breast cancer remains a challenge. Although PARP inhibitors are being evaluated in clinical trials, biomarkers are needed to identify patients who will most benefit from anti-PARP therapy. We determined the responses of three PARP inhibitors (veliparib, olaparib, and talazoparib) in a panel of eight triple-negative breast cancer cell lines. Therapeutic responses and cellular phenotypes were elucidated using high-content imaging and quantitative immunofluorescence to assess markers of DNA damage (53BP1) and apoptosis (cleaved PARP). We determined the pharmacodynamic changes as percentage of cells positive for 53BP1, mean number of 53BP1 foci per cell, and percentage of cells positive for cleaved PARP. Inspired by traditional dose-response measures of cell viability, an EC50 value was calculated for each cellular phenotype and each PARP inhibitor. The EC50 values for both 53BP1 metrics strongly correlated with IC50 values for each PARP inhibitor. Pathway enrichment analysis identified a set of DNA repair and cell cycle-associated genes that were associated with 53BP1 response following PARP inhibition. The overall accuracy of our 63 gene set in predicting response to olaparib in seven breast cancer patient-derived xenograft tumors was 86%. In triple-negative breast cancer patients who had not received anti-PARP therapy, the predicted response rate of our gene signature was 45%. These results indicate that 53BP1 is a biomarker of response to anti-PARP therapy in the laboratory, and our DNA damage response gene signature may be used to identify patients who are most likely to respond to PARP inhibition. Mol Cancer Ther; 16(12); 2892-901. ©2017 AACR.
Collapse
Affiliation(s)
- Saima Hassan
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon. .,Division of Surgical Oncology, Department of Surgery, Centre Hospitalier de l'Université de Montréal (CHUM), Centre de Recherche du CHUM, l'Université de Montréal, Québec, Canada
| | - Amanda Esch
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon
| | - Tiera Liby
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon
| | - Joe W Gray
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon
| | - Laura M Heiser
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
365
|
Novel Early Phase Clinical Trial Design in Oncology. Pharmaceut Med 2017. [DOI: 10.1007/s40290-017-0205-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
366
|
Ventz S, Alexander BM, Parmigiani G, Gelber RD, Trippa L. Designing Clinical Trials That Accept New Arms: An Example in Metastatic Breast Cancer. J Clin Oncol 2017; 35:3160-3168. [DOI: 10.1200/jco.2016.70.1169] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose The majority of randomized oncology trials are two-arm studies that test the efficacy of new therapies against a standard of care, thereby assigning a large proportion of patients to nonexperimental therapies. In contrast, multiarm studies efficiently share a common control arm while evaluating multiple experimental therapies. A major bottleneck for traditional multiarm trials is the requirement that all therapies—often drugs from different companies—have to be available at the same time when the trial starts. We evaluate the potential gains of a platform design—the rolling-arms design—that adds and removes arms on a rolling basis. Methods We define the rolling-arms design with the goal of minimizing the complexity of random assignment and data analyses of a platform trial. We then evaluate its potential advantages in hormone receptor–positive, human epidermal growth factor receptor 2–negative advanced breast cancer. Multiple pharmaceutical companies currently test CDK4/6 inhibitors in combination with letrozole in independent two-arm trials. We conducted a simulation study to quantify the reduction in sample size, number of patients treated with the standard of care, and the average time to treatment discovery if these therapies had been tested in a rolling-arms trial. Results A rolling-arms platform design with two to five experimental treatments can reduce the overall sample size requirement by up to 30% compared with standard two-arm studies. It assigns up to 60% fewer patients to the control arm compared with five independent trials that test distinct treatments. Moreover, under realistic scenarios, effective experimental treatments are discovered up to 15 months earlier compared with separate two-arm trials. Conclusion The rolling-arms platform design is applicable to a broad variety of diseases, and under realistic scenarios, it is substantially more efficient than standard two-arm randomized trials.
Collapse
Affiliation(s)
- Steffen Ventz
- Steffen Ventz, University of Rhode Island, Kingstown, RI; Brian M. Alexander, Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Dana-Farber Cancer Institute; Brian M. Alexander and Richard D. Gelber, Harvard Medical School; Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Harvard TH Chan School of Public Health; Richard D. Gelber, Frontier Science Foundation, Boston, MA
| | - Brian M. Alexander
- Steffen Ventz, University of Rhode Island, Kingstown, RI; Brian M. Alexander, Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Dana-Farber Cancer Institute; Brian M. Alexander and Richard D. Gelber, Harvard Medical School; Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Harvard TH Chan School of Public Health; Richard D. Gelber, Frontier Science Foundation, Boston, MA
| | - Giovanni Parmigiani
- Steffen Ventz, University of Rhode Island, Kingstown, RI; Brian M. Alexander, Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Dana-Farber Cancer Institute; Brian M. Alexander and Richard D. Gelber, Harvard Medical School; Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Harvard TH Chan School of Public Health; Richard D. Gelber, Frontier Science Foundation, Boston, MA
| | - Richard D. Gelber
- Steffen Ventz, University of Rhode Island, Kingstown, RI; Brian M. Alexander, Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Dana-Farber Cancer Institute; Brian M. Alexander and Richard D. Gelber, Harvard Medical School; Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Harvard TH Chan School of Public Health; Richard D. Gelber, Frontier Science Foundation, Boston, MA
| | - Lorenzo Trippa
- Steffen Ventz, University of Rhode Island, Kingstown, RI; Brian M. Alexander, Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Dana-Farber Cancer Institute; Brian M. Alexander and Richard D. Gelber, Harvard Medical School; Giovanni Parmigiani, Richard D. Gelber, and Lorenzo Trippa, Harvard TH Chan School of Public Health; Richard D. Gelber, Frontier Science Foundation, Boston, MA
| |
Collapse
|
367
|
Cristóbal I, Torrejón B, Martínez-Useros J, Madoz-Gurpide J, Rojo F, García-Foncillas J. PP2A regulates signaling through hormonal receptors in breast cancer with important therapeutic implications. Biochim Biophys Acta Rev Cancer 2017; 1868:435-438. [PMID: 28916342 DOI: 10.1016/j.bbcan.2017.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/10/2017] [Accepted: 08/31/2017] [Indexed: 11/27/2022]
Abstract
The functional inhibition of protein phosphatase 2A (PP2A) has emerged in the last years as a common alteration in breast cancer that determines poor outcome and contributes to disease progression and aggressiveness. Furthermore, expression of estrogen receptor (ER) is a high relevant molecular event with key therapeutic implications in breast cancer, and androgen receptor (AR) signaling is involved in the pathogenesis of breast cancer and represents a novel target with crescent importance in this disease. In this review, we summarize the role of the tumor suppressor PP2A in modulating ER and AR signaling in breast cancer, the molecular mechanisms involved, and its biological and therapeutic impact.
Collapse
Affiliation(s)
- Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain.
| | - Blanca Torrejón
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Javier Martínez-Useros
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | | | - Federico Rojo
- Pathology Department, IIS "Fundación Jiménez Diaz", Madrid, Spain.
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz, UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain.
| |
Collapse
|
368
|
Re-inventing drug development: A case study of the I-SPY 2 breast cancer clinical trials program. Contemp Clin Trials 2017; 62:168-174. [PMID: 28899813 DOI: 10.1016/j.cct.2017.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 09/04/2017] [Accepted: 09/07/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND In this case study, we profile the I-SPY 2 TRIAL (Investigation of Serial studies to Predict Your Therapeutic Response with Imaging And molecular anaLysis 2), a unique breast cancer clinical trial led by researchers at 20 leading cancer centers across the US, and examine its potential to serve as a model of drug development for other disease areas. This multicenter collaboration launched in 2010 to reengineer the drug development process to be more efficient and patient-centered. METHODS We conduct several interviews with the I-SPY leadership as well as a literature review of relevant publications to assess the I-SPY 2 initiative. RESULTS To date, six drugs have graduated from I-SPY 2, identified as excellent candidates for phase 3 trials in their corresponding tumor subtype, and several others have been or are still being evaluated. These trials are also more efficient, typically involving fewer subjects and reaching conclusions more quickly, and candidates have more than twice the predicted likelihood of success in a smaller phase 3 setting compared to traditional trials. CONCLUSIONS We observe that I-SPY 2 possesses several novel features that could be used as a template for more efficient and cost effective drug development, namely its adaptive trial design; precompetitive network of stakeholders; and flexible infrastructure to accommodate innovation.
Collapse
|
369
|
PARP Inhibitors in Breast Cancer: Latest Evidence. CURRENT BREAST CANCER REPORTS 2017. [DOI: 10.1007/s12609-017-0251-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
370
|
Siu LL, Ivy SP, Dixon EL, Gravell AE, Reeves SA, Rosner GL. Challenges and Opportunities in Adapting Clinical Trial Design for Immunotherapies. Clin Cancer Res 2017; 23:4950-4958. [PMID: 28864723 PMCID: PMC5669041 DOI: 10.1158/1078-0432.ccr-16-3079] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/18/2017] [Accepted: 07/06/2017] [Indexed: 12/26/2022]
Abstract
Immunotherapy adds an exciting new dimension to the treatment of cancer, joining other approaches as a key pillar in the oncotherapeutics armamentarium. Immuno-oncology agents harbor unique mechanisms of antitumor activity by leveraging the host immune system, which may result in response patterns, resistance kinetics, and toxicity profiles that differ from other systemic therapies. These features have led to many discussions on ways to optimally integrate immunotherapy into cancer clinical trials. This overview provides an introduction to the four CCR Focus articles that ensue, with special thoughts paid to clinical trial endpoints, biomarker development and validation, combination strategies, and limitations that arise with increasing use of these agents. In addition, this overview examines design concepts that may be applied to invigorate clinical trials and to maximize their impact in the immuno-oncology era.
Collapse
Affiliation(s)
- Lillian L Siu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.
| | - S Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | | | | | - Steven A Reeves
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Gary L Rosner
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| |
Collapse
|
371
|
Severson TM, Wolf DM, Yau C, Peeters J, Wehkam D, Schouten PC, Chin SF, Majewski IJ, Michaut M, Bosma A, Pereira B, Bismeijer T, Wessels L, Caldas C, Bernards R, Simon IM, Glas AM, Linn S, van ‘t Veer L. The BRCA1ness signature is associated significantly with response to PARP inhibitor treatment versus control in the I-SPY 2 randomized neoadjuvant setting. Breast Cancer Res 2017; 19:99. [PMID: 28851423 PMCID: PMC5574249 DOI: 10.1186/s13058-017-0861-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/25/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Patients with BRCA1-like tumors correlate with improved response to DNA double-strand break-inducing therapy. A gene expression-based classifier was developed to distinguish between BRCA1-like and non-BRCA1-like tumors. We hypothesized that these tumors may also be more sensitive to PARP inhibitors than standard treatments. METHODS A diagnostic gene expression signature (BRCA1ness) was developed using a centroid model with 128 triple-negative breast cancer samples from the EU FP7 RATHER project. This BRCA1ness signature was then tested in HER2-negative patients (n = 116) from the I-SPY 2 TRIAL who received an oral PARP inhibitor veliparib in combination with carboplatin (V-C), or standard chemotherapy alone. We assessed the association between BRCA1ness and pathologic complete response in the V-C and control arms alone using Fisher's exact test, and the relative performance between arms (biomarker × treatment interaction, likelihood ratio p < 0.05) using a logistic model and adjusting for hormone receptor status (HR). RESULTS We developed a gene expression signature to identify BRCA1-like status. In the I-SPY 2 neoadjuvant setting the BRCA1ness signature associated significantly with response to V-C (p = 0.03), but not in the control arm (p = 0.45). We identified a significant interaction between BRCA1ness and V-C (p = 0.023) after correcting for HR. CONCLUSIONS A genomic-based BRCA1-like signature was successfully translated to an expression-based signature (BRC1Aness). In the I-SPY 2 neoadjuvant setting, we determined that the BRCA1ness signature is capable of predicting benefit of V-C added to standard chemotherapy compared to standard chemotherapy alone. TRIAL REGISTRATION I-SPY 2 TRIAL beginning December 31, 2009: Neoadjuvant and Personalized Adaptive Novel Agents to Treat Breast Cancer (I-SPY 2), NCT01042379 .
Collapse
Affiliation(s)
- Tesa M. Severson
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | - Philip C. Schouten
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Ian J. Majewski
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Current address: Division of Cancer and Haematology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Magali Michaut
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Astrid Bosma
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Tycho Bismeijer
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Faculty of EEMCS, Delft University of Technology, Delft, The Netherlands
| | | | - René Bernards
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Sabine Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
372
|
Wolf DM, Yau C, Sanil A, Glas A, Petricoin E, Wulfkuhle J, Severson TM, Linn S, Brown-Swigart L, Hirst G, Buxton M, DeMichele A, Hylton N, Symmans F, Yee D, Paoloni M, Esserman L, Berry D, Rugo H, Olopade O, van 't Veer L. DNA repair deficiency biomarkers and the 70-gene ultra-high risk signature as predictors of veliparib/carboplatin response in the I-SPY 2 breast cancer trial. NPJ Breast Cancer 2017; 3:31. [PMID: 28948212 PMCID: PMC5572474 DOI: 10.1038/s41523-017-0025-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/24/2017] [Accepted: 06/05/2017] [Indexed: 12/17/2022] Open
Abstract
Veliparib combined with carboplatin (VC) was an experimental regimen evaluated in the biomarker-rich neoadjuvant I-SPY 2 trial for breast cancer. VC showed improved efficacy in the triple negative signature. However, not all triple negative patients achieved pathologic complete response and some HR+HER2− patients responded. Pre-specified analysis of five DNA repair deficiency biomarkers (BRCA1/2 germline mutation; PARPi-7, BRCA1ness, and CIN70 expression signatures; and PARP1 protein) was performed on 116 HER2− patients (VC: 72 and concurrent controls: 44). We also evaluated the 70-gene ultra-high risk signature (MP1/2), one of the biomarkers used to define subtype in the trial. We used logistic modeling to assess biomarker performance. Successful biomarkers were combined using a simple voting scheme to refine the ‘predicted sensitive’ group and Bayesian modeling used to estimate the pathologic complete response rates. BRCA1/2 germline mutation status associated with VC response, but its low prevalence precluded further evaluation. PARPi-7, BRCA1ness, and MP1/2 specifically associated with response in the VC arm but not the control arm. Neither CIN70 nor PARP1 protein specifically predicted VC response. When we combined the PARPi-7 and MP1/2 classifications, the 42% of triple negative patients who were PARPi7-high and MP2 had an estimated pCR rate of 75% in the VC arm. Only 11% of HR+/HER2− patients were PARPi7-high and MP2; but these patients were also more responsive to VC with estimated pathologic complete response rates of 41%. PARPi-7, BRCA1ness and MP1/2 signatures may help refine predictions of VC response, thereby improving patient care. Several predictive gene signatures can help identify breast cancer patients likely to respond to veliparib, an investigational PARP inhibitor, combined with the chemotherapy agent carboplatin. A team led by Denise Wolf, Christina Yau, and Laura van ‘t Veer from the University of California, San Francisco, used data from the I-SPY 2 trial to assess the predictive value of six different biomarkers in determining which women with early stage and locally advanced, aggressive breast cancer would have no signs of disease after veliparib—carboplatin treatment. They found three biomarkers with predictive value: a 7-gene expression signature that predicts breast cancer cell line sensitivity to another PARP inhibitor called olaparib; a 77-gene expression signature that detects molecular features shared with BRCA1-mutant tumours; and a 70-gene signature of recurrence risk called MammaPrint.
Collapse
Affiliation(s)
- Denise M Wolf
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94115 USA
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94115 USA
| | | | - Annuska Glas
- Agendia, Inc., 1098XH Amsterdam, The Netherlands
| | - Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, Virginia 22030 USA
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Fairfax, Virginia 22030 USA
| | - Tesa M Severson
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sabine Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lamorna Brown-Swigart
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94115 USA
| | - Gillian Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94115 USA
| | - Meredith Buxton
- QuantumLeap Healthcare Collaborative, San Francisco, CA 94143 USA
| | - Angela DeMichele
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Nola Hylton
- Department of Radiology, University of California, San Francisco, San Francisco, CA 94115 USA
| | - Fraser Symmans
- Division of Pathology, University of Texas, MD Anderson, Houston, TX 77030 USA
| | - Doug Yee
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455 USA
| | - Melissa Paoloni
- QuantumLeap Healthcare Collaborative, San Francisco, CA 94143 USA
| | - Laura Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94115 USA
| | - Don Berry
- Berry Consultants, LLC, Austin, TX 78746 USA
| | - Hope Rugo
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94115 USA
| | | | - Laura van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94115 USA
| |
Collapse
|
373
|
Systemic Therapies for Nonmetastatic Breast Cancer: The Role of Neoadjuvant and Adjuvant Chemotherapy and the Use of Endocrine Therapy. Clin Obstet Gynecol 2017; 59:756-771. [PMID: 27741213 DOI: 10.1097/grf.0000000000000237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Breast cancer is a heterogenous disease, comprised of at least 3 major subtypes: hormone receptor-positive/HER2-(HR+), HER2+, and HR-/HER2-(triple negative) breast cancers. The medical management of each subype is distinct. In this article, we review contemporary data supporting the use of chemotherapy, endocrine therapy and biologic therapies, especially HER2-directed agents, in the adjuvant and neoadjuvant setting in patients with newly diagnosed nonmetastatic (stage I-III) breast cancer.
Collapse
|
374
|
Dhawan MS, Bartelink IH, Aggarwal RR, Leng J, Zhang JZ, Pawlowska N, Terranova-Barberio M, Grabowsky JA, Gewitz A, Chien AJ, Moasser M, Kelley RK, Maktabi T, Thomas S, Munster PN. Differential Toxicity in Patients with and without DNA Repair Mutations: Phase I Study of Carboplatin and Talazoparib in Advanced Solid Tumors. Clin Cancer Res 2017; 23:6400-6410. [PMID: 28790114 DOI: 10.1158/1078-0432.ccr-17-0703] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/24/2017] [Accepted: 08/04/2017] [Indexed: 11/16/2022]
Abstract
Purpose: The PARP inhibitor (PARPi) talazoparib may potentiate activity of chemotherapy and toxicity in cells vulnerable to DNA damage.Experimental Design: This phase I study evaluated the safety, tolerability, pharmacokinetics, and efficacy of talazoparib and carboplatin. Pharmacokinetic modeling explored associations between DNA vulnerability and hematologic toxicity.Results: Twenty-four patients (eight males; 16 females) with solid tumors were enrolled in four cohorts at 0.75 and 1 mg daily talazoparib and weekly carboplatin (AUC 1 and 1.5, every 2 weeks or every 3 weeks), including 14 patients (58%) with prior platinum treatment. Dose-limiting toxicities included grade 3 fatigue and grade 4 thrombocytopenia; the MTD was not reached. Grade 3/4 toxicities included fatigue (13%), neutropenia (63%), thrombocytopenia (29%), and anemia (38%). After cycle 2's dose, delays/reductions were required in all patients. One complete and two partial responses occurred in germline BRCA1/2 (gBRCA1/2) patients. Four patients showed stable disease beyond 4 months, three of which had known mutations in DNA repair pathways. Pharmacokinetic toxicity modeling suggests that after three cycles of carboplatin AUC 1.5 every 3 weeks and talazoparib 1 mg daily, neutrophil counts decreased 78% [confidence interval (CI), 87-68] from baseline in gBRCA carriers and 63% (CI, 72-55) in noncarriers (P < 0.001). Pharmacokinetic toxicity modeling suggests an intermittent, pulse dosing schedule of PARP inhibition, differentiated by gBRCA mutation status, may improve the benefit/risk ratio of combination therapy.Conclusions: Carboplatin and talazoparib showed efficacy in DNA damage mutation carriers, but hematologic toxicity was more pronounced in gBRCA carriers. Carboplatin is best combined with intermittent talazoparib dosing differentiated by germline and somatic DNA damage mutation carriers. Clin Cancer Res; 23(21); 6400-10. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | - Jim Leng
- University of California, San Francisco, San Francisco, CA
| | - Jenna Z Zhang
- University of California, San Francisco, San Francisco, CA
| | - Nela Pawlowska
- University of California, San Francisco, San Francisco, CA
| | | | | | - Andrew Gewitz
- University of California, San Francisco, San Francisco, CA
| | - Amy J Chien
- University of California, San Francisco, San Francisco, CA
| | - Mark Moasser
- University of California, San Francisco, San Francisco, CA
| | - Robin K Kelley
- University of California, San Francisco, San Francisco, CA
| | - Tayeba Maktabi
- University of California, San Francisco, San Francisco, CA
| | - Scott Thomas
- University of California, San Francisco, San Francisco, CA
| | | |
Collapse
|
375
|
Connor JT, DeMichele A, Wittes J. University of Pennsylvania ninth annual conference on statistical issues in clinical trials: Where are we with adaptive clinical trial designs? (afternoon panel discussion). Clin Trials 2017; 14:470-482. [PMID: 28776417 DOI: 10.1177/1740774517723605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
376
|
Nishikawa T, Matsumoto K, Tamura K, Yoshida H, Imai Y, Miyasaka A, Onoe T, Yamaguchi S, Shimizu C, Yonemori K, Shimoi T, Yunokawa M, Xiong H, Nuthalapati S, Hashiba H, Kiriyama T, Leahy T, Komarnitsky P, Fujiwara K. Phase 1 dose-escalation study of single-agent veliparib in Japanese patients with advanced solid tumors. Cancer Sci 2017; 108:1834-1842. [PMID: 28665051 PMCID: PMC5581522 DOI: 10.1111/cas.13307] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/26/2017] [Indexed: 12/26/2022] Open
Abstract
Veliparib (ABT‐888) is a potent, orally bioavailable poly(ADP‐ribose) polymerase‐1 and ‐2 inhibitor. This phase 1 study evaluated the tolerability, pharmacokinetic profile, safety, and preliminary antitumor activity of single‐agent veliparib in Japanese patients with advanced solid tumors. Eligible patients were assigned to treatment with veliparib at 200 or 400 mg dose; veliparib was self‐administered orally twice daily on days 1–28 of 28‐day cycles. Dose escalation, following a 3 + 3 design, defined dose‐limiting toxicities, the maximum tolerated dose, and the recommended phase 2 dose. Sixteen patients were enrolled (median age, 59 years). Fourteen patients had high‐grade serous ovarian cancer, one had primary peritoneal cancer, and one had BRCA‐mutated breast cancer. The most frequent treatment‐emergent adverse events were nausea and vomiting (93.8% each), decreased appetite (62.5%), abdominal pain, diarrhea, and malaise (31.3% each). A grade ≥3 toxicity was observed in 50% of patients; one patient each in the 200 mg (n = 4) and 400 mg (n = 12) cohorts experienced serious adverse events. Dose‐limiting toxicities were observed for one patient at the 400 mg dose. No toxicities leading to death were reported. The recommended phase 2 dose was defined as 400 mg twice daily. The veliparib pharmacokinetic profile was consistent with that reported for the Western population. Two patients, both with ovarian cancer, had a RECIST partial response. Veliparib monotherapy showed manageable tolerability and safety profiles and a predictable pharmacokinetic profile at a 400 mg twice‐daily dose, and supports the inclusion of Japanese patients in the multinational phase 3 study (NCT02470585).
Collapse
Affiliation(s)
- Tadaaki Nishikawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan.,Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Koji Matsumoto
- Department of Medical Oncology, Hyogo Cancer Center, Hyogo, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Yoshida
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Yuichi Imai
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Aki Miyasaka
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Takuma Onoe
- Department of Medical Oncology, Hyogo Cancer Center, Hyogo, Japan
| | - Satoshi Yamaguchi
- Department of Gynecologic Oncology, Hyogo Cancer Center, Hyogo, Japan
| | - Chikako Shimizu
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsunori Shimoi
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Mayu Yunokawa
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hao Xiong
- AbbVie, Inc., North Chicago, Illinois, USA
| | | | | | | | | | | | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| |
Collapse
|
377
|
Maxfield KE, Buckman-Garner S, Parekh A. The Role of Public-Private Partnerships in Catalyzing the Critical Path. Clin Transl Sci 2017; 10:431-442. [PMID: 28776943 PMCID: PMC6402188 DOI: 10.1111/cts.12488] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/20/2017] [Indexed: 01/29/2023] Open
Affiliation(s)
- Kimberly E Maxfield
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - ShaAvhrée Buckman-Garner
- Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ameeta Parekh
- Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
378
|
Curigliano G, Burstein HJ, Winer EP, Gnant M, Dubsky P, Loibl S, Colleoni M, Regan MM, Piccart-Gebhart M, Senn HJ, Thürlimann B, André F, Baselga J, Bergh J, Bonnefoi H, Brucker SY, Cardoso F, Carey L, Ciruelos E, Cuzick J, Denkert C, Di Leo A, Ejlertsen B, Francis P, Galimberti V, Garber J, Gulluoglu B, Goodwin P, Harbeck N, Hayes DF, Huang CS, Huober J, Khaled H, Jassem J, Jiang Z, Karlsson P, Morrow M, Orecchia R, Osborne KC, Pagani O, Partridge AH, Pritchard K, Ro J, Rutgers EJT, Sedlmayer F, Semiglazov V, Shao Z, Smith I, Toi M, Tutt A, Viale G, Watanabe T, Whelan TJ, Xu B. De-escalating and escalating treatments for early-stage breast cancer: the St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol 2017; 28:1700-1712. [PMID: 28838210 PMCID: PMC6246241 DOI: 10.1093/annonc/mdx308] [Citation(s) in RCA: 777] [Impact Index Per Article: 97.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The 15th St. Gallen International Breast Cancer Conference 2017 in Vienna, Austria reviewed substantial new evidence on loco-regional and systemic therapies for early breast cancer. Treatments were assessed in light of their intensity, duration and side-effects, seeking where appropriate to escalate or de-escalate therapies based on likely benefits as predicted by tumor stage and tumor biology. The Panel favored several interventions that may reduce surgical morbidity, including acceptance of 2 mm margins for DCIS, the resection of residual cancer (but not baseline extent of cancer) in women undergoing neoadjuvant therapy, acceptance of sentinel node biopsy following neoadjuvant treatment of many patients, and the preference for neoadjuvant therapy in HER2 positive and triple-negative, stage II and III breast cancer. The Panel favored escalating radiation therapy with regional nodal irradiation in high-risk patients, while encouraging omission of boost in low-risk patients. The Panel endorsed gene expression signatures that permit avoidance of chemotherapy in many patients with ER positive breast cancer. For women with higher risk tumors, the Panel escalated recommendations for adjuvant endocrine treatment to include ovarian suppression in premenopausal women, and extended therapy for postmenopausal women. However, low-risk patients can avoid these treatments. Finally, the Panel recommended bisphosphonate use in postmenopausal women to prevent breast cancer recurrence. The Panel recognized that recommendations are not intended for all patients, but rather to address the clinical needs of the majority of common presentations. Individualization of adjuvant therapy means adjusting to the tumor characteristics, patient comorbidities and preferences, and managing constraints of treatment cost and access that may affect care in both the developed and developing world.
Collapse
Affiliation(s)
- G Curigliano
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | - H J Burstein
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - E P Winer
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M Gnant
- Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - P Dubsky
- Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
- Klinik St. Anna, Luzern, Switzerland
| | - S Loibl
- German Breast Group, Neu-Isenburg, Germany
| | - M Colleoni
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | - M M Regan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - M Piccart-Gebhart
- Department of Medical Oncology, Institut Jules Bordet, UniversitÕ Libre de Bruxelles, Brussels, Belgium
| | - H-J Senn
- Tumor and Breast Center ZeTuP, St. Gallen
| | - B Thürlimann
- Breast Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - F André
- Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - J Baselga
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - J Bergh
- Karolinska Institute and University Hospital, Stockholm, Sweden
| | - H Bonnefoi
- University of Bordeaux, Bordeaux, France
| | - S Y Brucker
- Universitäts-Frauenklinik Tübingen, Tübingen, Germany
| | - F Cardoso
- Champalimaud Cancer Centre, Lisbon, Portugal
| | - L Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, USA
| | - E Ciruelos
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - J Cuzick
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - C Denkert
- Institut für Pathologie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - A Di Leo
- Azienda Usl Toscana Centro, Prato, Italy
| | | | - P Francis
- Peter McCallum Cancer Centre, Melbourne, Australia
| | - V Galimberti
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | - J Garber
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - B Gulluoglu
- Marmara University School of Medicine, Istanbul, Turkey
| | - P Goodwin
- University of Toronto, Mount Sinai Hospital, Toronto, Canada
| | - N Harbeck
- University of Munich, München, Germany
| | - D F Hayes
- Comprehensive Cancer Center, University of Michigan, Ann-Arbor, USA
| | - C-S Huang
- National Taiwan University Hospital, Taipei, Taiwan
| | | | - H Khaled
- The National Cancer Institute, Cairo University, Cairo, Egypt
| | - J Jassem
- Medical University of Gdansk, Gdansk, Poland
| | - Z Jiang
- Hospital Affiliated to Military Medical Science, Beijing, China
| | - P Karlsson
- Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrensky University Hospital, Gothenburg, Sweden
| | - M Morrow
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - R Orecchia
- Breast Cancer Program, Istituto Europeo di Oncologia, Milano, Italy
| | | | - O Pagani
- Institute of Oncology Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - A H Partridge
- Breast Oncology Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - K Pritchard
- Sunnybrook Odette Cancer Center, University of Toronto, Toronto, Canada
| | - J Ro
- National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, Korea
| | - E J T Rutgers
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - F Sedlmayer
- LKH Salzburg, Paracelsus Medical University Clinics, Salzburg, Austria
| | - V Semiglazov
- N.N. Petrov Research Institute of Oncology, St. Petersburg, Russian Federation
| | - Z Shao
- Fudan University Cancer Hospital, Shanghai, China
| | - I Smith
- The Royal Marsden, Sutton, Surrey, UK
| | - M Toi
- Graduate School of Medicine Kyoto University, Sakyo-ku, Kyoto City, Japan
| | - A Tutt
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
| | - G Viale
- University of Milan, Milan, Italy
- Istituto Europeo di Oncologia, Milan, Italy
| | - T Watanabe
- Hamamatsu Oncology Center, Hamamatsu, Japan
| | | | - B Xu
- National Cancer Center, Chaoyang District, Beijing, China
| |
Collapse
|
379
|
Somlo G, Frankel PH, Arun BK, Ma CX, Garcia AA, Cigler T, Cream LV, Harvey HA, Sparano JA, Nanda R, Chew HK, Moynihan TJ, Vahdat LT, Goetz MP, Beumer JH, Hurria A, Mortimer J, Piekarz R, Sand S, Herzog J, Van Tongeren LR, Ferry-Galow KV, Chen AP, Ruel C, Newman EM, Gandara DR, Weitzel JN. Efficacy of the PARP Inhibitor Veliparib with Carboplatin or as a Single Agent in Patients with Germline BRCA1- or BRCA2-Associated Metastatic Breast Cancer: California Cancer Consortium Trial NCT01149083. Clin Cancer Res 2017; 23:4066-4076. [PMID: 28356425 PMCID: PMC5540749 DOI: 10.1158/1078-0432.ccr-16-2714] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/05/2016] [Accepted: 03/24/2017] [Indexed: 11/16/2022]
Abstract
Purpose: We aimed to establish the MTD of the poly (ADP-ribose) (PAR) polymerase inhibitor, veliparib, in combination with carboplatin in germline BRCA1- and BRCA2- (BRCA)-associated metastatic breast cancer (MBC), to assess the efficacy of single-agent veliparib, and of the combination treatment after progression, and to correlate PAR levels with clinical outcome.Experimental Design: Phase I patients received carboplatin (AUC of 5-6, every 21 days), with escalating doses (50-20 mg) of oral twice-daily (BID) veliparib. In a companion phase II trial, patients received single-agent veliparib (400 mg BID), and upon progression, received the combination at MTD. Peripheral blood mononuclear cell PAR and serum veliparib levels were assessed and correlated with outcome.Results: Twenty-seven phase I trial patients were evaluable. Dose-limiting toxicities were nausea, dehydration, and thrombocytopenia [MTD: veliparib 150 mg po BID and carboplatin (AUC of 5)]. Response rate (RR) was 56%; 3 patients remain in complete response (CR) beyond 3 years. Progression-free survival (PFS) and overall survival (OS) were 8.7 and 18.8 months. The PFS and OS were 5.2 and 14.5 months in the 44 patients in the phase II trial, with a 14% RR in BRCA1 (n = 22) and 36% in BRCA2 (n = 22). One of 30 patients responded to the combination therapy after progression on veliparib. Higher baseline PAR was associated with clinical benefit.Conclusions: Safety and efficacy are encouraging with veliparib alone and in combination with carboplatin in BRCA-associated MBC. Lasting CRs were observed when the combination was administered first in the phase I trial. Further investigation of PAR level association with clinical outcomes is warranted. Clin Cancer Res; 23(15); 4066-76. ©2017 AACR.
Collapse
Affiliation(s)
- George Somlo
- City of Hope Comprehensive Cancer Center, Duarte, California.
| | - Paul H Frankel
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Banu K Arun
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia X Ma
- Washington University School of Medicine, St. Louis, Missouri
| | - Agustin A Garcia
- University of Southern California/Norris Cancer Center, Los Angeles, California
| | | | - Leah V Cream
- Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | | | - Joseph A Sparano
- Montefiore Medical Center, Moses & Weuker Divisions, Department of Oncology, Bronx, New York
| | - Rita Nanda
- The University of Chicago, Chicago, Illinois
| | - Helen K Chew
- University of California, Davis Cancer Center, Sacramento, California
| | | | | | | | - Jan H Beumer
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Arti Hurria
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Joanne Mortimer
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Richard Piekarz
- Investigational Drug Branch, Cancer Therapy Evaluation Program, DCTD, NCI, Bethesda, Maryland
| | - Sharon Sand
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Josef Herzog
- City of Hope Comprehensive Cancer Center, Duarte, California
| | | | - Katherine V Ferry-Galow
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratories, Frederick, Maryland
| | - Alice P Chen
- Investigational Drug Branch, Cancer Therapy Evaluation Program, DCTD, NCI, Bethesda, Maryland
| | | | - Edward M Newman
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - David R Gandara
- University of California, Davis Cancer Center, Sacramento, California
| | | |
Collapse
|
380
|
Wang SS, Zimmermann M, Zhang H, Lin TY, Malfatti M, Haack K, Turteltaub KW, Cimino GD, de Vere White R, Pan CX, Henderson PT. A diagnostic microdosing approach to investigate platinum sensitivity in non-small cell lung cancer. Int J Cancer 2017; 141:604-613. [PMID: 28437852 PMCID: PMC5497716 DOI: 10.1002/ijc.30747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/21/2017] [Indexed: 12/29/2022]
Abstract
The platinum-based drugs cisplatin, carboplatin and oxaliplatin are often used for chemotherapy, but drug resistance is common. The prediction of resistance to these drugs via genomics is a challenging problem since hundreds of genes are involved. A possible alternative is to use mass spectrometry to determine the propensity for cells to form drug-DNA adducts-the pharmacodynamic drug-target complex for this class of drugs. The feasibility of predictive diagnostic microdosing was assessed in non-small cell lung cancer (NSCLC) cell culture and a pilot clinical trial. Accelerator mass spectrometry (AMS) was used to quantify [14 C]carboplatin-DNA monoadduct levels in the cell lines induced by microdoses and therapeutic doses of carboplatin, followed by correlation with carboplatin IC50 values for each cell line. The adduct levels in cell culture experiments were linearly proportional to dose (R2 = 0.95, p < 0.0001) and correlated with IC50 across all cell lines for microdose and therapeutically relevant carboplatin concentrations (p = 0.02 and p = 0.01, respectively). A pilot microdosing clinical trial was conducted to define protocols and gather preliminary data. Plasma pharmacokinetics (PK) and [14 C]carboplatin-DNA adducts in white blood cells and tumor tissues from six NSCLC patients were quantified via AMS. The blood plasma half-life of [14 C]carboplatin administered as a microdose was consistent with the known PK of therapeutic dosing. The optimal [14 C]carboplatin formulation for the microdose was 107 dpm/kg of body weight and 1% of the therapeutic dose for the total mass of carboplatin. No microdose-associated toxicity was observed in the patients. Additional accruals are required to significantly correlate adduct levels with response.
Collapse
Affiliation(s)
- Si-Si Wang
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA
| | - Maike Zimmermann
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA
| | - Hongyong Zhang
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA
| | - Tzu-yin Lin
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA
| | | | - Kurt Haack
- Lawrence Livermore National Laboratory, Livermore, CA
| | | | | | | | - Chong-xian Pan
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA
- Department of Urology, University of California Davis, Sacramento, CA
- VA Northern California Health Care System, Mather, CA
| | - Paul T. Henderson
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA
| |
Collapse
|
381
|
Yang L, Zhang Y, Shan W, Hu Z, Yuan J, Pi J, Wang Y, Fan L, Tang Z, Li C, Hu X, Tanyi JL, Fan Y, Huang Q, Montone K, Dang CV, Zhang L. Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Sci Transl Med 2017; 9:eaal1645. [PMID: 28747513 PMCID: PMC5705017 DOI: 10.1126/scitranslmed.aal1645] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 04/12/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022]
Abstract
Strategies to enhance response to poly(adenosine diphosphate-ribose) polymerase inhibitor (PARPi) in primary and acquired homologous recombination (HR)-proficient tumors would be a major advance in cancer care. We used a drug synergy screen that combined a PARPi, olaparib, with 20 well-characterized epigenetic drugs and identified bromodomain and extraterminal domain inhibitors (BETis; JQ1, I-BET762, and OTX015) as drugs that acted synergistically with olaparib in HR-proficient cancer cells. Functional assays demonstrated that repressed BET activity reduces HR and thus enhances PARPi-induced DNA damage in cancer cells. We also found that inhibition or depletion of BET proteins impairs transcription of BRCA1 and RAD51, two genes essential for HR. Moreover, BETi treatment sensitized tumors to PARP inhibition in preclinical animal models of HR-proficient breast and ovarian cancers. Finally, we showed that the BRD4 gene was focally amplified across 20 types of common cancers. Combination with BETi could greatly expand the utility of PARP inhibition to patients with HR-proficient cancer.
Collapse
Affiliation(s)
- Lu Yang
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Obstetrics and Gynecology, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Youyou Zhang
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Weiwei Shan
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Zhongyi Hu
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiao Yuan
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingjiang Pi
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yueying Wang
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lingling Fan
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Zhaoqing Tang
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chunsheng Li
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowen Hu
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janos L Tanyi
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Kathleen Montone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chi V Dang
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
382
|
Goldenholz DM, Goldenholz SR, Moss R, French J, Lowenstein D, Kuzniecky R, Haut S, Cristofaro S, Detyniecki K, Hixson J, Karoly P, Cook M, Strashny A, Theodore WH, Pieper C. Does accounting for seizure frequency variability increase clinical trial power? Epilepsy Res 2017; 137:145-151. [PMID: 28781216 DOI: 10.1016/j.eplepsyres.2017.07.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/28/2017] [Accepted: 07/21/2017] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Seizure frequency variability is associated with placebo responses in randomized controlled trials (RCT). Increased variability can result in drug misclassification and, hence, decreased statistical power. We investigated a new method that directly incorporated variability into RCT analysis, ZV. METHODS Two models were assessed: the traditional 50%-responder rate (RR50), and the variability-corrected score, ZV. Each predicted seizure frequency upper and lower limits using prior seizures. Accuracy was defined as percentage of time-intervals when the observed seizure frequencies were within the predicted limits. First, we tested the ZV method on three datasets (SeizureTracker: n=3016, Human Epilepsy Project: n=107, and NeuroVista: n=15). An additional independent SeizureTracker validation dataset was used to generate a set of 200 simulated trials each for 5 different sample sizes (total N=100 to 500 by 100), assuming 20% dropout and 30% drug efficacy. "Power" was determined as the percentage of trials successfully distinguishing placebo from drug (p<0.05). RESULTS Prediction accuracy across datasets was, ZV: 91-100%, RR50: 42-80%. Simulated RCT ZV analysis achieved >90% power at N=100 per arm while RR50 required N=200 per arm. SIGNIFICANCE ZV may increase the statistical power of an RCT relative to the traditional RR50.
Collapse
Affiliation(s)
- Daniel M Goldenholz
- Clinical Epilepsy Section, NINDS, NIH, United States; Division of Epilepsy, Beth Israel Deaconess Medical Center.
| | | | | | | | | | | | - Sheryl Haut
- Department of Neurology, Montefiore Medical Center/Albert Einstein College of Medicine, United States.
| | | | | | - John Hixson
- Department of Neurology, UCSF, United States.
| | | | | | - Alex Strashny
- Department of Neurology, Centers for Disease Control, United States.
| | | | - Carl Pieper
- Duke University Medical Center, Dept. of Biostatistics and Bioinformatics, United States.
| |
Collapse
|
383
|
Kourie HR, El Rassy E, Clatot F, de Azambuja E, Lambertini M. Emerging treatments for HER2-positive early-stage breast cancer: focus on neratinib. Onco Targets Ther 2017; 10:3363-3372. [PMID: 28744140 PMCID: PMC5513878 DOI: 10.2147/ott.s122397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Over the last decades, a better understanding of breast cancer heterogeneity provided tools for a biologically based personalization of anticancer treatments. In particular, the overexpression of the human epidermal growth factor receptor 2 (HER2) by tumor cells provided a specific target in these HER2-positive tumors. The development of the monoclonal antibody trastuzumab, and its approval in 1998 for the treatment of patients with metastatic disease, radically changed the natural history of this aggressive subtype of breast cancer. These findings provided strong support for the continuous research in targeting the HER2 pathway and implementing the development of new anti-HER2 targeted agents. Besides trastuzumab, a series of other anti-HER2 agents have been developed and are currently being explored for the treatment of breast cancer patients, including those diagnosed with early-stage disease. Among these agents, neratinib, an oral tyrosine kinase inhibitor that irreversibly inhibits HER1, HER2, and HER4 at the intracellular level, has shown promising results, including when administered to patients previously exposed to trastuzumab-based treatment. This article aims to review the available data on the role of the HER2 pathway in breast cancer and on the different targeted agents that have been studied or are currently under development for the treatment of patients with early-stage HER2-positive disease with a particular focus on neratinib.
Collapse
Affiliation(s)
- Hampig Raphael Kourie
- Department of Oncology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Elie El Rassy
- Department of Oncology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Florian Clatot
- Department of Medical Oncology and IRON/U1245, Centre Henri Becquerel, Rouen, France.,Breast Cancer Translational Research Laboratory
| | - Evandro de Azambuja
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Matteo Lambertini
- Breast Cancer Translational Research Laboratory.,Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
384
|
Affiliation(s)
- Janet Woodcock
- From the Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Lisa M LaVange
- From the Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| |
Collapse
|
385
|
Colman H. Toward more informative biomarker-based clinical trials in glioblastoma. Neuro Oncol 2017; 19:880-881. [PMID: 28521009 DOI: 10.1093/neuonc/nox063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Howard Colman
- Department of Neurosurgery and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
386
|
Dey N, De P, Leyland-Jones B. PI3K-AKT-mTOR inhibitors in breast cancers: From tumor cell signaling to clinical trials. Pharmacol Ther 2017; 175:91-106. [DOI: 10.1016/j.pharmthera.2017.02.037] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
387
|
Oualla K, El-Zawahry HM, Arun B, Reuben JM, Woodward WA, Gamal El-Din H, Lim B, Mellas N, Ueno NT, Fouad TM. Novel therapeutic strategies in the treatment of triple-negative breast cancer. Ther Adv Med Oncol 2017; 9:493-511. [PMID: 28717401 PMCID: PMC5502951 DOI: 10.1177/1758834017711380] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous subtype of breast cancer that is defined by negative estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) status. Treating patients with TNBC remains clinically challenging, as patients are not candidates for endocrine or HER2-directed therapy. As a result, chemotherapy with traditional agents such as anthracyclines and taxanes remains the only available option with moderate success. Recent discoveries have revealed that TNBC is a heterogeneous disease at the clinical, histological and molecular levels. The use of biomarkers to identify distinct subsets of TNBC that derive the greatest benefit from presently approved as well as novel therapeutics has become the main focus of current research. The aim of this review is to explore the clinical and biological complexity of TNBC as well as identify novel therapeutic options that target the various molecular subsets of TNBC.
Collapse
Affiliation(s)
- Karima Oualla
- Medical Oncology Department, Hassan II University Hospital, Fes, Morocco
| | - Heba M. El-Zawahry
- Department of Medical Oncology, The National Cancer Institute, Cairo University, Cairo, Egypt
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M. Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy A. Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heba Gamal El-Din
- Department of Surgical Oncology, The National Cancer Institute, Cairo University, Cairo, Egypt
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Nawfel Mellas
- Medical Oncology Department, Hassan II University Hospital, Fes, Morocco
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Tamer M. Fouad
- Department of Medical Oncology, The National Cancer Institute, Cairo University, Kasr El-Aini Road, Cairo, 11796, Egypt
| |
Collapse
|
388
|
Xu S, Zhao C, Jia Z, Wang X, Han Y, Yang Z. Down-regulation of PARP1 by miR-891b sensitizes human breast cancer cells to alkylating chemotherapeutic drugs. Arch Gynecol Obstet 2017; 296:543-549. [PMID: 28660502 DOI: 10.1007/s00404-017-4444-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 06/22/2017] [Indexed: 11/30/2022]
Abstract
PURPOSE Breast cancer is the most common invasive type of cancer among women. Role of different microRNAs (miRNAs) and poly(ADP-ribose) polymerases (PARPs) in breast cancer has been well established. This study aimed to explore the effects of miR-891b on sensitizing breast cancer cells to alkylating chemotherapeutic drugs through PARPs. METHODS The expression of miR-891b and PARP1 in human breast cancer cells HCC1806 was overexpressed by transfection with their mimics or expressing vector. Then, the transfected cells were exposed to 40 µM N-methyl-N-nitro-N-nitrosoguanidine (MNNG) for 1 h. The correlation between miR-891b and PARP1 was detected by RT-qPCR, western blot, and dual-luciferase reporter assay. Besides, MTT assay and Annexin V assay were done to measure cell proliferation and apoptosis, respectively. RESULTS PARP1 was a target of miR-891b, and it was negatively regulated by miR-891b. MiR-891b increased the sensitivity of the HCC1806 cells to the cytotoxic effects of MNNG through suppressing cell proliferation and increasing the percentage of apoptotic cells. Restoration of PARP1 activity in the HCC1806 cells led to loss of miR-891b mediated sensitivity of the HCC1806 cells to MNNG. CONCLUSION MiR-891b increases the sensitivity of the breast cancer cells (HCC1806) to the cytotoxic effects of the chemotherapeutic agent MNNG by suppressing the expression of PARP1.
Collapse
Affiliation(s)
- Shujian Xu
- Department of Breast Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China.,Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Cui Zhao
- Department of Rehabilitation Medicine, The Affiliated Hospital to Binzhou Medical University, Binzhou, 256603, China
| | - Zhongming Jia
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Xilong Wang
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Yong Han
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China
| | - Zhenlin Yang
- Department of Thyroid and Breast Surgery, The Affiliated Hospital to Binzhou Medical University, No. 661, Huangheer Road, Bincheng District, Binzhou, 256603, China.
| |
Collapse
|
389
|
Echavarria I, López-Tarruella S, Márquez-Rodas I, Jerez Y, Martin M. Neratinib for the treatment of HER2-positive early stage breast cancer. Expert Rev Anticancer Ther 2017. [PMID: 28649882 DOI: 10.1080/14737140.2017.1338954] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Despite the advances in the treatment of HER2-positive breast cancer, resistance to actual chemotherapeutic regimens eventually occurs. Neratinib, an orally available pan-inhibitor of the ERBB family, represents an interesting new option for early-stage HER2-positive breast cancer. Areas covered: In this article, the development of neratinib, with a special focus on its potential value in the treatment of early-stage HER2-positive breast cancer, has been reviewed. For this purpose, a literature search was conducted, including preclinical studies, early-phase trials in advanced cancer with neratinib in monotherapy and in combination, and phase II and large phase III trials in the early setting. Management of neratinib-induced toxicity, future perspectives for the drug, and ongoing trials are also discussed in this review. Expert commentary: Neratinib is emerging as a promising oral drug for the treatment of HER2-positive breast cancer. Although FDA and EMA approval is derived from the extended adjuvant treatment, this setting may not be the ideal scenario to obtain the beneficial effects of neratinib. Confirmatory data in the neoadjuvant setting and subgroup analysis from the ExTENET trial might bring some light into the best setting for neratinib therapy. Data from confirmatory trials in the metastatic setting are also required.
Collapse
Affiliation(s)
- Isabel Echavarria
- a Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) , Madrid , Spain
| | - Sara López-Tarruella
- b Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CiberOnc , Madrid , Spain
| | - Iván Márquez-Rodas
- b Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CiberOnc , Madrid , Spain
| | - Yolanda Jerez
- b Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CiberOnc , Madrid , Spain
| | - Miguel Martin
- c Instituto de Investigación Sanitaria Gregorio Maranon (IiSGM), Universidad Complutense, CiberOnc, GEICAM , Madrid , Spain
| |
Collapse
|
390
|
Denkert C, Liedtke C, Tutt A, von Minckwitz G. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet 2017; 389:2430-2442. [PMID: 27939063 DOI: 10.1016/s0140-6736(16)32454-0] [Citation(s) in RCA: 612] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 10/31/2016] [Accepted: 11/09/2016] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer is a heterogeneous disease and specific therapies have not been available for a long time. Therefore, conventional chemotherapy is still considered the clinical state of the art. Different subgroups of triple-negative breast cancer have been identified on the basis of protein expression, mRNA signatures, and genomic alterations. Important elements of triple-negative breast cancer biology include high proliferative activity, an increased immunological infiltrate, a basal-like and a mesenchymal phenotype, and deficiency in homologous recombination, which is in part associated with loss of BRCA1 or BRCA2 function. A minority of triple-negative tumours express luminal markers, such as androgen receptors, and have a lower proliferative activity. These biological subgroups are overlapping and currently cannot be combined into a unified model of triple-negative breast cancer biology. Nevertheless, the molecular analysis of this disease has identified potential options for targeted therapeutic intervention. This has led to promising clinical strategies, including modified chemotherapy approaches targeting the DNA damage response, angiogenesis inhibitors, immune checkpoint inhibitors, or even anti-androgens, all of which are being evaluated in phase 1-3 clinical studies. This Series paper focuses on the most relevant clinical questions, summarises the results of recent clinical trials, and gives an overview of ongoing studies and trial concepts that will lead to a more refined therapy for this tumour type.
Collapse
Affiliation(s)
- Carsten Denkert
- Institute of Pathology, Charité Universitätsmedizin Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Germany.
| | | | - Andrew Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research and Breast Cancer Now Research Unit, King's College, London, UK
| | | |
Collapse
|
391
|
Yin G, Chen N, Lee JJ. Bayesian Adaptive Randomization and Trial Monitoring with Predictive Probability for Time-to-event Endpoint. STATISTICS IN BIOSCIENCES 2017; 10:420-438. [PMID: 30559900 DOI: 10.1007/s12561-017-9199-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
There has been much development in Bayesian adaptive designs in clinical trials. In the Bayesian paradigm, the posterior predictive distribution characterizes the future possible outcomes given the currently observed data. Based on the interim time-to-event data, we develop a new phase II trial design by combining the strength of both Bayesian adaptive randomization and the predictive probability. By comparing the mean survival times between patients assigned to two treatment arms, more patients are assigned to the better treatment on the basis of adaptive randomization. We continuously monitor the trial using the predictive probability for early termination in the case of superiority or futility. We conduct extensive simulation studies to examine the operating characteristics of four designs: the proposed predictive probability adaptive randomization design, the predictive probability equal randomization design, the posterior probability adaptive randomization design, and the group sequential design. Adaptive randomization designs using predictive probability and posterior probability yield a longer overall median survival time than the group sequential design, but at the cost of a slightly larger sample size. The average sample size using the predictive probability method is generally smaller than that of the posterior probability design.
Collapse
Affiliation(s)
- Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong,
| | - Nan Chen
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, U.S.A.,
| | - J Jack Lee
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, U.S.A.,
| |
Collapse
|
392
|
Bettaieb A, Paul C, Plenchette S, Shan J, Chouchane L, Ghiringhelli F. Precision medicine in breast cancer: reality or utopia? J Transl Med 2017. [PMID: 28623955 PMCID: PMC5474301 DOI: 10.1186/s12967-017-1239-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Many cancers, including breast cancer, have demonstrated prognosis and support advantages thanks to the discovery of targeted therapies. The advent of these new approaches marked the rise of precision medicine, which leads to improve the diagnosis, prognosis and treatment of cancer. Precision medicine takes into account the molecular and biological specificities of the patient and their tumors that will influence the treatment determined by physicians. This new era of medicine is accessible through molecular genetics platforms, the development of high-speed sequencers and means of analysis of these data. Despite the spectacular results in the treatment of cancers including breast cancer, described in this review, not all patients however can benefit from this new strategy. This seems to be related to the many genetic mutations, which may be different from one patient to another or within the same patient. It comes to give new impetus to the research—both from a technological and biological point of view—to make the hope of precision medicine accessible to all.
Collapse
Affiliation(s)
- Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France. .,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France. .,Immunology and Immunotherapy of Cancer Laboratory, EA7269, Université de Bourgogne, EPHE 7 Bd Jeanne d'Arc, 21079, Dijon, France.
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France
| | - Stéphanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France
| | - Jingxuan Shan
- Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Lotfi Chouchane
- Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - François Ghiringhelli
- Département d'Oncologie Médicale, Centre Georges-François-Leclerc, 21000, Dijon, France.,Plateforme de Transfert en Biologie Cancérologique, Centre Georges-François-Leclerc, 21000, Dijon, France.,UMR 1231 Inserm-Université de Bourgogne Franche Comté, UFR des Sciences de Santé, 21000, Dijon, France.,Université de Bourgogne, 21000, Dijon, France
| |
Collapse
|
393
|
Goldberg RM, Wei L, Fernandez S. The Evolution of Clinical Trials in Oncology: Defining Who Benefits from New Drugs Using Innovative Study Designs. Oncologist 2017; 22:1015-1019. [PMID: 28620092 PMCID: PMC5599203 DOI: 10.1634/theoncologist.2017-0153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/17/2017] [Indexed: 11/17/2022] Open
Abstract
Historically, advocates of randomized drug treatment trials in oncology contributed to the changing paradigm for testing cancer treatments in the U.S., which led to global efforts to generate evidence of promise in preclinical model systems and translate those findings into improved patient outcomes. In the age of genomic medicine, the challenge to speed the evolution of how clinical trials are conducted in patients with cancer continues.
Collapse
Affiliation(s)
- Richard M Goldberg
- Department of Medicine, Morgantown, West Virginia, USA
- West Virginia University Cancer Institute, Morgantown, West Virginia, USA
- Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia, USA
| | - Lai Wei
- Department of Biomedical Informatics, Ohio State University Center for Biostatistics, Ohio State University, Columbus, Ohio, USA
| | - Soledad Fernandez
- Department of Biomedical Informatics, Ohio State University Center for Biostatistics, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
394
|
Earl H, Molica S, Rutkowski P. Spotlight on landmark oncology trials: the latest evidence and novel trial designs. BMC Med 2017; 15:111. [PMID: 28571584 PMCID: PMC5454584 DOI: 10.1186/s12916-017-0884-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 05/23/2017] [Indexed: 12/18/2022] Open
Abstract
The era of precision oncology is marked with prominent successes in the therapy of advanced soft tissue sarcomas, breast cancer, ovarian cancer and haematological neoplasms, among others. Moreover, recent trials of immune checkpoint inhibitors in melanoma, non-small cell lung carcinoma, and head and neck cancers have significantly influenced the therapeutic landscape by providing promising evidence for immunotherapy efficacy in the adjuvant setting in high-risk locoregional disease. To speed up the introduction of targeted therapy for cancer patients, novel phase II trials are being designed, and may likely form the basis for the 'landmark trials' of the future. A special article collection in BMC Medicine, "Spotlight on landmark oncology trials", features articles from invited experts on recent clinical practice-changing trials.
Collapse
Affiliation(s)
- Helena Earl
- University of Cambridge Department of Oncology, NIHR Cambridge Biomedical Research Centre, and Hon Consultant in Medical Oncology, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Stefano Molica
- Department Hematology-Oncology, Azienda Ospedaliera Pugliese-Ciaccio, 88100, Catanzaro, Italy
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute - Oncology Center, Roentgena 5, 02-781, Warsaw, Poland.
| |
Collapse
|
395
|
Bardia A, Mayer IA, Diamond JR, Moroose RL, Isakoff SJ, Starodub AN, Shah NC, O’Shaughnessy J, Kalinsky K, Guarino M, Abramson V, Juric D, Tolaney SM, Berlin J, Messersmith WA, Ocean AJ, Wegener WA, Maliakal P, Sharkey RM, Govindan SV, Goldenberg DM, Vahdat LT. Efficacy and Safety of Anti-Trop-2 Antibody Drug Conjugate Sacituzumab Govitecan (IMMU-132) in Heavily Pretreated Patients With Metastatic Triple-Negative Breast Cancer. J Clin Oncol 2017. [DOI: 10.1200/jco.2016.70.8297.2017.2.test] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Aditya Bardia
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Ingrid A. Mayer
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Jennifer R. Diamond
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Rebecca L. Moroose
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Steven J. Isakoff
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Alexander N. Starodub
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Nikita C. Shah
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Joyce O’Shaughnessy
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Kevin Kalinsky
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Michael Guarino
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Vandana Abramson
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Dejan Juric
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Sara M. Tolaney
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Jordan Berlin
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Wells A. Messersmith
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Allyson J. Ocean
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - William A. Wegener
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Pius Maliakal
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Robert M. Sharkey
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Serengulam V. Govindan
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - David M. Goldenberg
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Linda T. Vahdat
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| |
Collapse
|
396
|
Gonçalves A, Monneur A, Viens P, Bertucci F. The use of systemic therapies to prevent progression of inflammatory breast cancer: which targeted therapies to add on cytotoxic combinations? Expert Rev Anticancer Ther 2017; 17:593-606. [PMID: 28506194 DOI: 10.1080/14737140.2017.1330655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Inflammatory breast cancer is a rare but frequently fatal disease, essentially because of its high ability to develop distant metastases. Even though the prognosis of IBC was significantly improved by multimodal management, including the systematic use of cytotoxic-based induction, the prognosis remains largely dismal. Areas covered: This review presents the main achievements in the systemic treatment of IBC during the past 30 years. It focuses more specifically on recent results obtained with targeted therapies, including anti-HER2 and anti-angiogenic agents. Novel approaches under investigation are presented. Expert commentary: Current management of IBC is subtype-specific and the largest benefit has been achieved in HER2-positive disease. The identification of breakthrough therapeutic advances is eagerly awaited and will require the development of IBC-specific clinical trials. Future clinical investigations should not only aim to increase the pathological response rate but also to eradicate distant metastases, which ultimately lead to patient death.
Collapse
Affiliation(s)
- Anthony Gonçalves
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Audrey Monneur
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - Patrice Viens
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| | - François Bertucci
- a Department of Medical Oncology, Institut Paoli-Calmettes, Aix Marseille Univ , CNRS U7258, INSERM U1068, CRCM , Marseille , France
| |
Collapse
|
397
|
Chien AJ, Chambers J, Mcauley F, Kaplan T, Letourneau J, Hwang J, Kim MO, Melisko ME, Rugo HS, Esserman LJ, Rosen MP. Fertility preservation with ovarian stimulation and time to treatment in women with stage II-III breast cancer receiving neoadjuvant therapy. Breast Cancer Res Treat 2017; 165:151-159. [PMID: 28503722 DOI: 10.1007/s10549-017-4288-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 11/29/2022]
Abstract
PURPOSE To determine whether fertility preservation with ovarian stimulation (OS) results in treatment delay in breast cancer (BC) patients receiving neoadjuvant therapy (NAT). METHODS This is a retrospective study of women screened for the prospective neoadjuvant ISPY2 trial at the University of California San Francisco. All patients were <43, had stage II-III BC, and received neoadjuvant therapy. Time to initiation of NAT was compared between women who underwent OS (STIM) and women who did not (control). Patient and tumor characteristics, as well as oncologic outcomes, were compared between STIM and control groups. RESULTS 82 patients were included (34 STIM and 48 control). STIM patients were overall younger (mean = 35 vs. 36.9 years old, p = 0.06), and more likely to be childless (79.4 vs 31.2%, p < 0.0001) than controls. Mean time from diagnosis to initiation of NAT was 40 days, with no significant difference between STIM and control groups (mean 39.8 days vs 40.9 days, p = 0.75). Mean time from diagnosis to fertility consultation was 16.3 days. With median follow-up of 79 months, 16 (19.5%) patients have recurred or died from BC. Rates of pCR, recurrence, and death were similar in both groups. Six of 34 STIM patients have undergone embryo transfer, resulting in one patient with two live births. CONCLUSION Fertility preservation with OS can be performed in the neoadjuvant setting without delay in initiation of systemic therapy and should be discussed with all early-stage BC patients of reproductive age.
Collapse
Affiliation(s)
- A Jo Chien
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA, 94115, USA.
| | - Julia Chambers
- University of California San Francisco School of Medicine, Francisco, CA, USA
| | - Fiona Mcauley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA, 94115, USA
| | - Tessa Kaplan
- Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Joseph Letourneau
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Jimmy Hwang
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA, 94115, USA
| | - Mi-Ok Kim
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA, 94115, USA
| | - Michelle E Melisko
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA, 94115, USA
| | - Hope S Rugo
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA, 94115, USA
| | - Laura J Esserman
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA, 94115, USA
| | - Mitchell P Rosen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
398
|
Suh DH, Kim M, Kim K, Kim HJ, Lee KH, Kim JW. Major clinical research advances in gynecologic cancer in 2016: 10-year special edition. J Gynecol Oncol 2017; 28:e45. [PMID: 28382802 PMCID: PMC5391398 DOI: 10.3802/jgo.2017.28.e45] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 12/11/2022] Open
Abstract
In 2016, 13 topics were selected as major research advances in gynecologic oncology. For ovarian cancer, study results supporting previous ones regarding surgical preventive strategies were reported. There were several targeted agents that showed comparable responses in phase III trials, including niraparib, cediranib, and nintedanib. On the contrary to our expectations, dose-dense weekly chemotherapy regimen failed to prove superior survival outcomes compared with conventional triweekly regimen. Single-agent non-platinum treatment to prolong platinum-free-interval in patients with recurrent, partially platinum-sensitive ovarian cancer did not improve and even worsened overall survival (OS). For cervical cancer, we reviewed robust evidences of larger-scaled population-based study and cost-effectiveness of nonavalent vaccine for expanding human papillomavirus (HPV) vaccine coverage. Standard of care treatment of locally advanced cervical cancer (LACC) was briefly reviewed. For uterine corpus cancer, new findings about appropriate surgical wait time from diagnosis to surgery were reported. Advantages of minimally invasive surgery over conventional laparotomy were reconfirmed. There were 5 new gene regions that increase the risk of developing endometrial cancer. Regarding radiation therapy, Post-Operative Radiation Therapy in Endometrial Cancer (PORTEC)-3 quality of life (QOL) data were released and higher local control rate of image-guided adaptive brachytherapy was reported in LACC. In addition, 4 general oncology topics followed: chemotherapy at the end-of-life, immunotherapy with reengineering T-cells, actualization of precision medicine, and artificial intelligence (AI) to make personalized cancer therapy real. For breast cancer, adaptively randomized trials, extending aromatase inhibitor therapy, and ribociclib and palbociclib were introduced.
Collapse
Affiliation(s)
- Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Miseon Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jae Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
399
|
Du Y, Yamaguchi H, Hsu JL, Hung MC. PARP inhibitors as precision medicine for cancer treatment. Natl Sci Rev 2017. [DOI: 10.1093/nsr/nwx027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AbstractPersonalized or precision medicine is an emerging treatment approach tailored to individuals or certain groups of patients based on their unique characteristics. These types of therapies guided by biomarkers tend to be more effective than traditional approaches, especially in cancer. The inhibitor against poly (ADP-ribose) polymerase (PARP), olaparib (Lynparza, AstraZeneca), which was approved by the US Food and Drug Administration (FDA) in 2014, demonstrated efficacy specifically for ovarian cancer patients harboring mutations in BRCA genes, which encode proteins in DNA double-strand break repairs. However, the response to PARP inhibitors has been less encouraging in other cancer types that also carry defects in the BRCA genes. Thus, furthering our understanding of the underlying mechanism of PARP inhibitors and resistance is critical to improve their efficacy. In this review, we summarize the results of preclinical studies and the clinical application of PARP inhibitors, and discuss the future direction of PARP inhibitors as a potential marker-guided personalized medicine for cancer treatment.
Collapse
Affiliation(s)
- Yi Du
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
| | - Hirohito Yamaguchi
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
| | - Jennifer L. Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 40402
- Department of Biotechnology, Asia University, Taichung 41354
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 40402
- Department of Biotechnology, Asia University, Taichung 41354
| |
Collapse
|
400
|
|