301
|
Walsh EM, Shalaby A, O’Loughlin M, Keane N, Webber MJ, Kerin MJ, Keane MM, Glynn SA, Callagy GM. Outcome for triple negative breast cancer in a retrospective cohort with an emphasis on response to platinum-based neoadjuvant therapy. Breast Cancer Res Treat 2018; 174:1-13. [PMID: 30488345 PMCID: PMC6418073 DOI: 10.1007/s10549-018-5066-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
Purpose The rate of pathological complete response (pCR) for patients with triple negative breast cancer (TNBC) is increased when carboplatin is added to neo-adjuvant chemotherapy (NACT). However, while phase III trial data showing a survival benefit are awaited, carboplatin is not yet standard-of-care for TNBC. The aim of this study was to examine the rate of pCR and the outcome for those treated with carboplatin and to examine the predictors of response to therapy. Methods The retrospective series comprised 333 consecutive patients with TNBC (median follow-up time, 43 months). Adjuvant chemotherapy was given to 51% (n = 168) of patients and 29% (n = 97) received anthracycline–taxane NACT with carboplatin given to 9% (n = 31) of patients. Results Overall, 25% (n = 78) of patients experienced a breast cancer recurrence and 22% (n = 68) died from disease. A pCR breast and pCR breast/axilla was more common in those who received carboplatin (n = 18, 58% and n = 17, 55%, respectively) compared those who did not (n = 23, 36% and n = 18, 28%, respectively) (p = 0.041 and p = 0.011, respectively). By multivariable analysis, carboplatin and high tumor grade were independent predictors of pCR breast/axilla (ORnon-pCR = 0.17; 95% CI 0.06–0.54; p = 0.002; and ORnon-pCR = 0.05, 95% CI 0.01–0.27; p < 0.001, respectively). pCR breast/axilla was an independent predictor of DFS (HRnon-pCR=6.23; 95% CI 1.36–28.50; p = 0.018), metastasis-free survival (HRnon-pCR = 5.08; 95% CI 1.09–23.65; p = 0.038) and BCSS (HRnon-pCR = 8.52; 95% CI 1.09–66.64; p = 0.041). Conclusion Carboplatin therapy and high tumor grade are associated with a significant increase in the rate of pCR, which is an independent predictor of outcome. These data support the use of carboplatin in NACT for TNBC, while results from phase III studies are awaited. Electronic supplementary material The online version of this article (10.1007/s10549-018-5066-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elaine M. Walsh
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
- Department of Medical Oncology, University Hospital Galway, Galway, Ireland
| | - Aliaa Shalaby
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Mark O’Loughlin
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Nessa Keane
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Mark J Webber
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Michael J. Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Maccon M. Keane
- Department of Medical Oncology, University Hospital Galway, Galway, Ireland
| | - Sharon A. Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Grace M. Callagy
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| |
Collapse
|
302
|
Denduluri N, Miller K, O'Regan RM. Using a Neoadjuvant Approach for Evaluating Novel Therapies for Patients With Breast Cancer. Am Soc Clin Oncol Educ Book 2018; 38:47-55. [PMID: 30231324 DOI: 10.1200/edbk_200719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Preoperative systemic therapy, though primarily used to downstage breast cancers, can offer, using pathologic complete response (pCR) as an endpoint, a rapid assessment of efficacy of a given therapeutic approach, particularly in triple-negative (TNBC) and HER2-positive breast cancers. Recently, this approach resulted in the approval of pertuzumab for HER2-positive cancers, in a considerably quicker timeline than would have been possible with its assessment in the adjuvant setting. However, the use of preoperative systemic therapy remains controversial, as the higher response rates noted with newer approaches have not routinely translated into improved longer-term outcomes, nor have they been confirmed in larger adjuvant trials. Almost all trials have demonstrated that pCR is a robust prognostic marker in patients with TNBC and HER2-positive cancers, so part of this discrepancy may be due to inadequate power in the preoperative trials and/or due to the heterogeneous nature of breast cancers. PCR following preoperative chemotherapy is not prognostic in many hormone receptor (HR)-positive breast cancers, especially those with a luminal A phenotype, which typically has minimal response to chemotherapy. Given this lack of response to chemotherapy, there is considerable interest in the use of neoadjuvant endocrine therapy (NET). The rate of pCR to NET in HR-positive cancers is low, leading to the use of surrogate markers, including changes in Ki-67 and the preoperative endocrine prognostic index (PEPI) score, as biomarkers of efficacy. Overall, the use of neoadjuvant approaches offers a rapid assessment of efficacy of novel therapies and remains a useful research tool for drug evaluation.
Collapse
Affiliation(s)
- Neelima Denduluri
- From the Virginia Cancer Specialists, Fairfax, VA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Kathy Miller
- From the Virginia Cancer Specialists, Fairfax, VA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Ruth M O'Regan
- From the Virginia Cancer Specialists, Fairfax, VA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; University of Wisconsin Carbone Cancer Center, Madison, WI
| |
Collapse
|
303
|
Use of Multigene Prognostic Indices to Guide Clinical Decision-Making Regarding Adjuvant Systemic Therapy. CURRENT BREAST CANCER REPORTS 2018. [DOI: 10.1007/s12609-018-0297-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
304
|
Simonsen KL, Fracasso PM, Bernstein SH, Wind-Rotolo M, Gupta M, Comprelli A, Reilly TP, Cassidy J. The Fast Real-time Assessment of Combination Therapies in Immuno-ONcology (FRACTION) program: innovative, high-throughput clinical screening of immunotherapies. Eur J Cancer 2018; 103:259-266. [PMID: 30292142 DOI: 10.1016/j.ejca.2018.07.127] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/06/2018] [Accepted: 07/24/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND The unprecedented success of immuno-oncology (I-O) agents targeting the cytotoxic T lymphocyte-associated antigen 4 and programmed death-1/programmed death-ligand 1 pathways has stimulated the rapid development of other I-O agents against novel immune targets. Bristol-Myers Squibb has designed a novel phase II platform trial, the Fast Real-time Assessment of Combination Therapies in Immuno-ONcology (FRACTION) Program, to efficiently identify promising combinations for patients with specific malignancies. The concept and study design of the FRACTION Program-currently ongoing in patients with advanced non-small-cell lung cancer (FRACTION-Lung), gastric cancer (FRACTION-Gastric Cancer) and renal cell carcinoma (FRACTION-RCC)-are described. METHODS The FRACTION Program comprises open-label, phase II studies that use adaptive randomisation designs with rolling combination regimens. Master Protocols provide the overall study design framework, whereas Sub-Protocols introduced over time provide details on specific I-O combination therapies to which patients may be randomised. In a Master Protocol, patients are enrolled into different Study Tracks based on characteristics such as prior I-O therapy experience. Patients who progress may be rerandomised to other combination regimens from any ongoing Sub-Protocol. Primary objectives are to assess objective response rate, median duration of response and progression-free survival rate at 24 weeks; the secondary objective is to investigate safety and tolerability. Biomarker collection before and on treatment will facilitate identification of patient subsets who benefit most from each therapy. CONCLUSIONS The FRACTION Program allows for the evaluation of multiple I-O combinations through individual studies for specific tumours using an adaptive trial design and continuous enrolment.
Collapse
Affiliation(s)
- Katy L Simonsen
- Global Biometric Sciences - Oncology, Bristol-Myers Squibb, Princeton, NJ, USA.
| | | | | | | | - Manish Gupta
- Clinical Pharmacology and Pharmacometrics - Oncology, Bristol-Myers Squibb, Princeton, NJ, USA.
| | - Adriana Comprelli
- Oncology Clinical Operations Strategy, Bristol-Myers Squibb, Princeton, NJ, USA.
| | | | - Jim Cassidy
- Exploratory Clinical and Translational Research - Early Oncology Development, Bristol-Myers Squibb, Princeton, NJ, USA.
| |
Collapse
|
305
|
van Werkhoven CH, Harbarth S, Bonten MJM. Adaptive designs in clinical trials in critically ill patients: principles, advantages and pitfalls. Intensive Care Med 2018; 45:678-682. [PMID: 30377740 PMCID: PMC6483961 DOI: 10.1007/s00134-018-5426-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022]
Affiliation(s)
- C H van Werkhoven
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - S Harbarth
- Infection Control Program and WHO Collaborating Center, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - M J M Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
306
|
Fremd C, Jaeger D, Schneeweiss A. Targeted and immuno-biology driven treatment strategies for triple-negative breast cancer: current knowledge and future perspectives. Expert Rev Anticancer Ther 2018; 19:29-42. [PMID: 30351981 DOI: 10.1080/14737140.2019.1537785] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Accounting for about 15% of breast cancer patients, triple-negative breast cancer (TNBC) is responsible for 25% of disease related deaths, more frequent distant spread and visceral metastasis. However, improving survival in TNBC failed and primary resistance, immunological ignorance and tumor heterogeneity limit clinical activity of novel therapies. In view of recent molecular, genetic and immunologic insights, this review aims to describe the current status of immunological and targeted treatments from a hypothesis driven perspective. Areas covered: Recent preclinical studies and ongoing clinical trials for immune directed and targeted treatments of TNBC are summarized, including immune-checkpoint blockade, resistance mechanisms, inhibition of poly (ADP-ribose) polymerase (PARP), combinatorial strategies as well as preclinical, hypothesis generating studies. Expert commentary: Sustained responses have been observed with immune-checkpoint blockade and PARP inhibitors demonstrated remarkable efficacy in germline BRCA mutated TNBC. In order to generate clinical success of many other, to date ineffective, targeted and immune therapies, the integration of multidimensional, large amounts of data, will be essential and likely accelerate treatment progress of TNBC.
Collapse
Affiliation(s)
- Carlo Fremd
- a National Center for Tumor Diseases, Department of Medical Oncology , University of Heidelberg , Heidelberg , Germany
| | - Dirk Jaeger
- a National Center for Tumor Diseases, Department of Medical Oncology , University of Heidelberg , Heidelberg , Germany
| | - Andreas Schneeweiss
- a National Center for Tumor Diseases, Department of Medical Oncology , University of Heidelberg , Heidelberg , Germany
| |
Collapse
|
307
|
He X, Ji J, Dong R, Liu H, Dai X, Wang C, Esteva FJ, Yeung SCJ. Prognosis in different subtypes of metaplastic breast cancer: a population-based analysis. Breast Cancer Res Treat 2018; 173:329-341. [PMID: 30341462 DOI: 10.1007/s10549-018-5005-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Metaplastic breast cancer (MpBC) is a rare histological subtype of breast cancer recognized as a unique pathologic entity in 2000. However, the pathogenesis, optimal therapy, and prognosis of MpBC and the potential effect of systemic treatments on different subtypes of MpBC are not well defined. METHODS A retrospective population-based study was performed to identify breast cancer patients with MpBC and other triple-negative breast cancers (TNBC) between 2010 and 2014 using the surveillance, Epidemiology, and End Results (SEER) database. Chi-square test was used to analyze characteristics between subgroups. Kaplan-Meier analysis and Multivariate Cox regressions were used to evaluate overall survival (OS) of MpBC, TNBC, and MpBC subgroups. Competing risk analysis and multivariate regression model of competing risk were used to assess breast cancer-specific survival (BCSS) of MpBC and TNBC RESULTS: We identified a study cohort of 22,433 patients (1112 MpBC and 21,321 TNBC). MpBC correlated with older population, larger tumor size and less lymph node involvement, and TNBC phenotype. Patients with MpBC especially with triple-negative subtype (TN-MpBC) had worse survival than the overall TNBC population. However, the prognosis of MpBC without triple-negative subtype (non-TN MpBC) was not different from that of TNBC. In Kaplan-Meier analysis, chemotherapy was not associated with significant difference in OS of TN-MpBC. In non-TN MpBC group, the 3-year OS was 79.8% for patients receiving chemotherapy and 70.5% in patients without chemotherapy, and chemotherapy was associated (P = 0.033) with improved OS. Within the MpBC patients, radiotherapy was significantly (HR 1.544; 95% CI 1.148-2.078; P = 0.004) associated with improved OS and (HR 1.474; 95% CI 1.067-2.040; P = 0.019) BCSS. CONCLUSIONS Patients with TN-MpBC had worse prognosis than TNBC and chemotherapy was not associated with improved survival. In contrast, non-TN MpBC may derive survival benefit from chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Xuexin He
- Department of Medical Oncology, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jiefang Rd, Hangzhou, Zhejiang, 310010, China. .,Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jiali Ji
- Department of Medical Oncology, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jiefang Rd, Hangzhou, Zhejiang, 310010, China
| | - Rongrong Dong
- Department of Medical Oncology, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jiefang Rd, Hangzhou, Zhejiang, 310010, China
| | - Hong Liu
- Department of Medical Oncology, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jiefang Rd, Hangzhou, Zhejiang, 310010, China
| | - Xiaolan Dai
- Department of Pharmacology, Shantou University, Shantou, Guangdong, China
| | - Chongjian Wang
- Department of Biostatistics, Zhengzhou University, Zhengzhou, Henan, China
| | - Francisco J Esteva
- Division of Hematology/Oncology, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Sai-Ching Jim Yeung
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1468, Houston, TX, 77030, USA.
| |
Collapse
|
308
|
Escrivá-de-Romaní S, Arumí M, Zamora E, Bellet M. Neoadjuvant Model as a Platform for Research in Breast Cancer and Novel Targets under Development in this Field. Breast Care (Basel) 2018; 13:251-262. [PMID: 30319327 DOI: 10.1159/000492122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
For decades, the neoadjuvant setting has provided a useful scenario for research in breast cancer. Historically, neoadjuvant clinical trials, either hormone therapy-based or chemotherapy-based, have tried to recapitulate the results of their counterpart adjuvant studies, but with smaller patient numbers, more rapid outcomes (clinical response and/or pathologic complete response (pCR)), together with additional biologic information. As for neoadjuvant chemotherapy trials, the increase in pCR rates has been recently accepted as an appropriate surrogate marker to accelerate drug approval in high-risk breast cancer patients. In this setting, with the exception of luminal A tumors, pCR has been associated with improved long-term outcomes, particularly when the analysis is based on specific trials for each breast cancer subtype. For luminal tumors receiving neoadjuvant endocrine therapy, Ki67 at 2-4 weeks and the preoperative endocrine prognostic index score are the most accepted intermediate markers of efficacy, which will be validated in ongoing larger trials. In this review, we describe the different neoadjuvant designs: from the classical randomized trials in which treatment is delivered for 6 or more months to short non-therapeutic presurgical studies lasting just 2 or 3 weeks. We also review the main neoadjuvant trials, either ongoing or completed, for luminal, triple-negative, and HER2-positive breast cancer. The translational effort and research of biomarkers conducted in these studies will be particularly addressed.
Collapse
Affiliation(s)
- Santiago Escrivá-de-Romaní
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Miriam Arumí
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Esther Zamora
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Meritxell Bellet
- Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
309
|
Liposits G, Loh KP, Soto-Perez-de-Celis E, Dumas L, Battisti NML, Kadambi S, Baldini C, Banerjee S, Lichtman SM. PARP inhibitors in older patients with ovarian and breast cancer: Young International Society of Geriatric Oncology review paper. J Geriatr Oncol 2018; 10:337-345. [PMID: 30333088 DOI: 10.1016/j.jgo.2018.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 09/16/2018] [Accepted: 10/10/2018] [Indexed: 01/28/2023]
Abstract
Breast and ovarian cancer are common malignancies among older adults, causing significant morbidity and mortality. Although most cases of breast and ovarian cancer are sporadic, a significant proportion is caused by mutations in cancer susceptibility genes, most often breast cancer susceptibility genes (BRCA) 1 and 2. Furthermore, some breast and ovarian tumors are phenotypically similar to those with BRCA mutations, a phenomenon known as "BRCAness". BRCA mutations and "BRCAness" lead to defects in DNA repair, which may be a target for therapeutic agents such as Poly ADP-Ribose Polymerase (PARP) inhibitors. PARP inhibitors are novel medications which lead to double-strand breaks resulting in cell death due to synthetic lethality, and which have been shown to be effective in patients with advanced breast and ovarian cancers with or without BRCA mutations. Three different PARP inhibitors (olaparib, niraparib, and rucaparib) have been approved for the treatment of ovarian cancer and one (olaparib) for breast cancer harboring BRCA mutations. Here, we review the currently available evidence regarding the use of PARP inhibitors for the treatment of patients with breast and ovarian cancer, with a particular focus on the inclusion of older adults in clinical trials of these therapies. Additionally, we provide an overview of currently ongoing studies of PARP inhibitors in breast and ovarian cancer, and include recommendations for increasing the evidence-base for using these medications among older patients.
Collapse
Affiliation(s)
- Gabor Liposits
- Department of Oncology, Region Hospital West Jutland, Gl. Landevej 61, Herning, 7400, Denmark.
| | - Kah Poh Loh
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| | - Enrique Soto-Perez-de-Celis
- Enrique Soto-Perez-de-Celis Department of Geriatrics, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico.
| | - Lucy Dumas
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.
| | | | - Sindhuja Kadambi
- Division of Geriatrics/Aging, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France.
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust Sutton, United Kingdom.
| | - Stuart M Lichtman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, NY, USA.
| |
Collapse
|
310
|
Robert M, Patsouris A, Frenel JS, Gourmelon C, Augereau P, Campone M. Emerging PARP inhibitors for treating breast cancer. Expert Opin Emerg Drugs 2018; 23:211-221. [DOI: 10.1080/14728214.2018.1527900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Marie Robert
- René Gauducheau, Institut de Cancérologie de l’Ouest, St Herblain, France
| | - Anne Patsouris
- Paul Papin, Institut de Cancérologie de l’Ouest, Angers, France
| | | | - Carole Gourmelon
- René Gauducheau, Institut de Cancérologie de l’Ouest, St Herblain, France
| | - Paule Augereau
- Paul Papin, Institut de Cancérologie de l’Ouest, Angers, France
| | - Mario Campone
- René Gauducheau, Institut de Cancérologie de l’Ouest, St Herblain, France
- Medical oncology, Centre de Recherche en Cancérologie Nantes-Angers (CRNA), Saint-Herblain, France
| |
Collapse
|
311
|
Loriot Y, Meynard G, Klajer E, Bolognini C, Gassian N, Thiery-Vuillemin A. [DNA damage repair: An emerging strategy in metastatic prostate cancer]. Bull Cancer 2018; 105:944-954. [PMID: 30278883 DOI: 10.1016/j.bulcan.2018.05.017] [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] [Received: 03/10/2018] [Accepted: 05/25/2018] [Indexed: 12/17/2022]
Abstract
Genetic instability is one part of the oncogenic process. Gene mutations involved in DNA repair mechanisms can promote this genetic instability and participate in oncogenesis and metastatic progression. In prostate cancer, DNA repair abnormalities mainly correspond to somatic or constitutional mutations of the BRCA2 and ATM genes. Therapeutic management of metastatic castration-resistant prostate cancer (mCRPC) is currently based on new hormonal therapies (abiraterone, enzalutamide) and taxane-type chemotherapy (docetaxel or cabazitaxel). Preliminary data tend to indicate a specific activity of agents causing DNA breaks (platinum salts) and PARP inhibitors in patients with these DNA repair abnormalities. The frequency of DNA repair gene mutations in patients with prostate cancer (around 20%) and the antitumor response of PARP inhibitors make it a possible short-term therapeutic strategy with several registering clinical trials ongoing.
Collapse
Affiliation(s)
- Yohann Loriot
- Gustave-Roussy, département de médecine, Inserm U981, 114, rue Édouard-Vaillant, 94805 Villejuif, France
| | | | - Elodie Klajer
- CHU de Besançon, oncologie, 25030 Besançon cedex, France
| | | | - Noémie Gassian
- CHU de Besançon, oncologie, 25030 Besançon cedex, France
| | - Antoine Thiery-Vuillemin
- CHU de Besançon, oncologie, 25030 Besançon cedex, France; Inserm, UMR1098, 25020 Besançon cedex, France; Université de Franche-Comté, UMR1098, SFR IBCT, 25020 Besançon, France.
| |
Collapse
|
312
|
Bardia A, Parton M, Kümmel S, Estévez LG, Huang CS, Cortés J, Ruiz-Borrego M, Telli ML, Martin-Martorell P, López R, Beck JT, Ismail-Khan R, Chen SC, Hurvitz SA, Mayer IA, Carreon D, Cameron S, Liao S, Baselga J, Kim SB. Paclitaxel With Inhibitor of Apoptosis Antagonist, LCL161, for Localized Triple-Negative Breast Cancer, Prospectively Stratified by Gene Signature in a Biomarker-Driven Neoadjuvant Trial. J Clin Oncol 2018; 36:JCO2017748392. [PMID: 30235087 DOI: 10.1200/jco.2017.74.8392] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE There are currently no targeted therapies approved for triple-negative breast cancer (TNBC). A tumor necrosis factor α ( TNFα)-based gene expression signature (GS) predictive of sensitivity to LCL161, inhibitor of apoptosis antagonist, was translated into a clinical assay and evaluated in a neoadjuvant trial. PATIENTS AND METHODS Women with localized TNBC (T2/N0-2/M0) were prospectively stratified by GS status and randomly assigned (1:1) to receive oral LCL161 (1,800 mg once per week) and intravenous paclitaxel (80 mg/m2 once per week; combination arm) or paclitaxel alone (control arm) for 12 weeks, followed by surgery. The primary objective was to determine whether neoadjuvant LCL161 enhances efficacy of paclitaxel, defined by > 7.5% increase in the pathologic complete response (pCR, breast) rate, stratified by GS. RESULTS Of 209 patients enrolled (207 with valid GS scores), 30.4% had GS-positive TNBC. In the GS-positive group, pCR was higher in the combination versus the control arm (38.2% v 17.2%), with 88.8% posterior probability of > 7.5% increase in pCR. However, in the GS-negative group, the pCR was lower in the combination group (5.6% v 16.4%), with 0% posterior probability of > 7.5% increase in pCR. A higher incidence of grade 3 or 4 adverse events in the combination arm included neutropenia (24.5%) and diarrhea (5.7%). Overall, 19 patients (18.1%) in the combination arm discontinued treatment because of adverse events, including pyrexia (n = 5), pneumonia (n = 4), and pneumonitis (n = 4), versus five patients (4.9%) in the control arm. CONCLUSION This neoadjuvant trial provides evidence supporting a biomarker-driven targeted therapy approach for selected patients with GS-positive TNBC and demonstrates the utility of a neoadjuvant trial for biomarker validation and drug development, but also highlights toxicity risk. Future neoadjuvant clinical trials should carefully weigh these considerations for targeted therapy development in biomarker-defined TNBC.
Collapse
Affiliation(s)
- Aditya Bardia
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Marina Parton
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sherko Kümmel
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Laura G Estévez
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chiun-Sheng Huang
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Javier Cortés
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Manuel Ruiz-Borrego
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Melinda L Telli
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Paloma Martin-Martorell
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Rafael López
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - J Thaddeus Beck
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Roohi Ismail-Khan
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Shin-Cheh Chen
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sara A Hurvitz
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ingrid A Mayer
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Daniel Carreon
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Scott Cameron
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Serena Liao
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - José Baselga
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Bae Kim
- Aditya Bardia, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; Scott Cameron and Serena Liao, Novartis Institutes for BioMedical Research, Cambridge, MA; Marina Parton, The Royal Marsden Hospital, London, United Kingdom; Sherko Kümmel, Kliniken Essen-Mitte, Essen, Germany; Laura G. Estévez, Centro Integral Oncológico Clara Campal; Javier Cortés, University Hospital Ramón y Cajal, Madrid; Javier Cortés,Vall d'Hebron Institute of Oncology, Barcelona; Manuel Ruiz-Borrego, Hospital Universitario Virgen del Rocío, Seville; Paloma Martin-Martorell, Hospital Clínico Universitario de Valencia, Valencia; Rafael López, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Chiun-Sheng Huang, National Taiwan University Hospital, National Taiwan University College of Medicine; Shin-Cheh Chen, Chang Gung Memorial Hospital, Taipei, Republic of China; Melinda L. Telli, Stanford University School of Medicine, Stanford; Sara A. Hurvitz, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA; J. Thaddeus Beck, Highlands Oncology Group, Fayetteville, AR; Roohi Ismail-Khan, Moffitt Cancer Center, Tampa, FL; Ingrid A. Mayer, Vanderbilt-Ingram Cancer Center, Nashville, TN; Daniel Carreon, Novartis Pharmaceuticals Corporation, East Hanover, NJ; José Baselga, Memorial Sloan Kettering Cancer Center, New York, NY; and Sung-Bae Kim, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
313
|
Abstract
The number of breast cancer (BC) cases is growing worldwide, being most frequently diagnosed in the early-setting. Mammaprint™ is a 70-gene-expression signature, originally designed for selecting early BC patients with low risk of developing metastasis, so that they could be spared adjuvant chemotherapy. Its use as a prognostic biomarker has been extensively validated, both retrospectively and prospectively. However, its value as a predictive tool and as a clinically useful tool remains controversial. This review will describe how the test works, its application in the clinic and its limitations. Cost-effectiveness studies will be summarized. Finally, we will provide a perspective on the use of Mammaprint in the near future, as a valuable tool for personalizing the treatment of early BC patients.
Collapse
Affiliation(s)
- Mariana Brandão
- Institut Jules Bordet & L'Université Libre de Bruxelles (U.L.B.), 121, 1000, Brussels, Belgium
| | - Noam Pondé
- Institut Jules Bordet & L'Université Libre de Bruxelles (U.L.B.), 121, 1000, Brussels, Belgium
| | - Martine Piccart-Gebhart
- Institut Jules Bordet & L'Université Libre de Bruxelles (U.L.B.), 121, 1000, Brussels, Belgium
| |
Collapse
|
314
|
Triple-Negative Breast Cancers: Systematic Review of the Literature on Molecular and Clinical Features with a Focus on Treatment with Innovative Drugs. Curr Oncol Rep 2018; 20:76. [PMID: 30128845 DOI: 10.1007/s11912-018-0726-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Triple-negative breast cancer (TNBC) accounts for 15-20% of diagnosed breast tumours, with higher incidence in young and African-American women, and it is frequently associated with BRCA germline mutations. Chemotherapy is the only well-established therapeutic option in both early- and advanced-stages of the disease. TNBC tumours relapse earlier after standard anthracycline- and/or taxane-based chemotherapy treatments, generally within 1-3 years after the diagnosis, and often develop visceral metastases, representing the subtype with a worse prognosis among all breast cancers. In the present review, we will provide an updated overview of the available results of recent clinical trials for this disease and we will describe the implications of the known molecular pathways representing novel targets for development of future therapies for TNBC patients. RECENT FINDINGS Over the past decade, the advent of gene expression micro-array technology has led to the identification of different actionable targets including various genomic alterations, androgen receptor, PARP, PI3K, VEGF and other proteins of the angiogenic pathway. Thus, novel targeted drugs have been tested in clinical trials reporting promising results in specific TNBC molecular subgroups. Although cytotoxic chemotherapy remains the mainstay of treatment for TNBC patients, the identification of novel 'drugable' targets and pathways for developing personalized treatments represents a promising investigational approach in the management of the TNBC subtype.
Collapse
|
315
|
McCann KE, Hurvitz SA. Advances in the use of PARP inhibitor therapy for breast cancer. Drugs Context 2018; 7:212540. [PMID: 30116283 PMCID: PMC6089618 DOI: 10.7573/dic.212540] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 12/30/2022] Open
Abstract
Poly-ADP-ribose polymerase 1 (PARP-1) and PARP-2 are DNA damage sensors that are most active during S-phase of the cell cycle and that have wider-reaching roles in DNA repair than originally described. BRCA1 and BRCA2 (Breast Cancer) proteins are involved in homologous recombination repair (HRR), which requires a homologous chromosome or sister chromatid as a template to faithfully repair DNA double-strand breaks. The small-molecule NAD+ mimetics, olaparib, niraparib, rucaparib, veliparib, and talazoparib, inhibit the catalytic activity of PARP-1 and PARP-2 and are currently being studied in later-stage clinical trials. PARP inhibitor clinical trials have predominantly focused on patients with breast and ovarian cancer with deleterious germline BRCA1 and BRCA2 mutations (gBRCA1/2+) but are now expanding to include cancers with known, suspected, or more-likely-than-not defects in homologous recombination repair. In ovarian cancer, this group also includes women whose cancers are responsive to platinum therapy. Olaparib was FDA-approved in January 2018 for the treatment of gBRCA1/2+ metastatic breast cancers. gBRCA1+ predisposes women to develop triple-negative breast cancers, while women with gBRCA2+ tend to develop hormone-receptor-positive, human epidermal growth factor receptor 2 negative breast cancers. Although PARP inhibitor monotherapy strategies seem most effective in cancers with homologous recombination repair defects, combination strategies may allow expansion into a wider range of cancers. By interfering with DNA repair, PARP inhibitors essentially sensitize cells to DNA-damaging chemotherapies and radiation therapy. Certainly, one could also consider expanding the utility of PARP inhibitors beyond gBRCA1/2+ cancers by causing DNA damage with cytotoxic agents in the presence of a DNA repair inhibitor. Unfortunately, in numerous phase I clinical trials utilizing a combination of cytotoxic chemotherapy at standard doses with dose-escalation of PARP inhibitors, there has generally been failure to reach monotherapy dosages of PARP inhibitors due to myelosuppressive toxicities. Strategies utilizing angiogenesis inhibitors and immune checkpoint inhibitors are generally not hindered by additive toxicities, though the utility of combining PARP inhibitors with treatments that have not been particularly effective in breast cancers somewhat tempers enthusiasm. Finally, there are combination strategies that may serve to mitigate resistance to PARP inhibitors, namely, upregulation of the intracellular PhosphoInositide-3-kinase, AK thymoma (protein kinase B), mechanistic target of rapamycin (PI3K-AKT-mTOR) pathway, or perhaps are more simply meant to interfere with a cell growth pathway heavily implicated in breast cancers while administering relatively well-tolerated PARP inhibitor therapy.
Collapse
Affiliation(s)
- Kelly E McCann
- David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Sara A Hurvitz
- David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
316
|
Calfee CS, Delucchi KL, Sinha P, Matthay MA, Hackett J, Shankar-Hari M, McDowell C, Laffey JG, O'Kane CM, McAuley DF. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. THE LANCET RESPIRATORY MEDICINE 2018; 6:691-698. [PMID: 30078618 DOI: 10.1016/s2213-2600(18)30177-2] [Citation(s) in RCA: 480] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/12/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Precision medicine approaches that target patients on the basis of disease subtype have transformed treatment approaches to cancer, asthma, and other heterogeneous syndromes. Two distinct subphenotypes of acute respiratory distress syndrome (ARDS) have been identified in three US-based clinical trials, and these subphenotypes respond differently to positive end-expiratory pressure and fluid management. We aimed to investigate whether these subphenotypes exist in non-US patient populations and respond differently to pharmacotherapies. METHODS HARP-2 was a multicentre, randomised controlled trial of simvastatin (80 mg) versus placebo done in general intensive care units (ICUs) at 40 hospitals in the UK and Ireland within 48 h of onset of ARDS. The primary outcome was ventilator-free days, and secondary outcomes included non-pulmonary organ failure-free days and mortality. In a secondary analysis of HARP-2, we applied latent class analysis to baseline data without consideration of outcomes to identify subphenotypes, and we compared clinical outcomes across subphenotypes and treatment groups. FINDINGS 540 patients were recruited to HARP-2. One patient withdrew consent for the use of their data, so data from 539 patients were analysed. In our secondary analysis, a two-class (two subphenotype) model was an improvement over a one-class model (p<0·0001), with 353 (65%) patients in the hypoinflammatory subphenotype group and 186 (35%) in the hyperinflammatory subphenotype group. Additional classes did not improve model fit. Clinical and biological characteristics of the two subphenotypes were similar to previous studies. Patients with the hyperinflammatory subphenotype had fewer ventilator-free days (median 2 days [IQR 0-17] vs 18 [IQR 0-23]; p<0·0001), fewer non-pulmonary organ failure-free days (15 [0-25] vs 27 [21-28]; p<0·0001), and higher 28-day mortality (73 [39%] vs 59 [17%]; p<0·0001) than did those with the hypoinflammatory subphenotype. Although HARP-2 found no difference in 28-day survival between placebo and simvastatin, significantly different survival was identified across patients stratified by treatment and subphenotype (p<0·0001). Specifically, within the hyperinflammatory subphenotype, patients treated with simvastatin had significantly higher 28-day survival than did those given placebo (p=0·008). A similar pattern was observed for 90-day survival. INTERPRETATION Two subphenotypes of ARDS were identified in the HARP-2 cohort, with distinct clinical and biological features and disparate clinical outcomes. The hyperinflammatory subphenotype had improved survival with simvastatin compared with placebo. These findings support further pursuit of predictive enrichment strategies in critical care clinical trials. FUNDING UK Efficacy and Mechanism Evaluation Programme and National Institutes of Health.
Collapse
Affiliation(s)
- Carolyn S Calfee
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Anesthesia, University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Kevin L Delucchi
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Pratik Sinha
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Michael A Matthay
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Anesthesia, University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan Hackett
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Manu Shankar-Hari
- Guy's and St Thomas' NHS Foundation Trust, ICU Support Offices, St Thomas' Hospital, London, UK; School of Immunology and Microbial Sciences, Kings College London, London, UK
| | - Cliona McDowell
- Northern Ireland Clinical Trials Unit, The Royal Hospitals Belfast, Belfast, UK
| | - John G Laffey
- Anaesthesia, School of Medicine, and Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland; Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada; Departments of Anesthesia, Physiology and Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - Cecilia M O'Kane
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Daniel F McAuley
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK; Regional Intensive Care Unit, The Royal Hospitals Belfast, Belfast, UK
| | | |
Collapse
|
317
|
Xu W, Gao J, Hu F, Cheung SH. Response-adaptive treatment allocation for non-inferiority trials with heterogeneous variances. Comput Stat Data Anal 2018. [DOI: 10.1016/j.csda.2018.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
318
|
Moore-Smith L, Forero-Torres A, Stringer-Reasor E. Future Developments in Neoadjuvant Therapy for Triple-Negative Breast Cancer. Surg Clin North Am 2018; 98:773-785. [PMID: 30005773 DOI: 10.1016/j.suc.2018.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Breast cancer is the 2nd leading cause of cancer-related death in women in the United States. In general, advances in targeted treatment for breast cancer have improved over the last twenty years, except in the triple-negative breast cancer (TNBC) subtype. TNBC is an aggressive breast cancer subtype with limited treatment options as compared to hormone positive breast cancers. Recently, genomic profiling of TNBC shows promise in aiding clinicians to develop personalized targeted agents. Prioritizing novel molecular-based therapies in the neoadjuvant setting may help investigators understand mechanisms of resistance and ultimately improve patient outcomes in TNBC.
Collapse
Affiliation(s)
- Lakisha Moore-Smith
- Department of Medicine, Brookwood Baptist Health - Princeton, 833 Princeton Avenue, POB III Suite 200, Birmingham, AL 35211-1311, USA
| | - Andres Forero-Torres
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2517, Birmingham, AL 35294-3300, USA
| | - Erica Stringer-Reasor
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2501, Birmingham, AL 35294-3300, USA.
| |
Collapse
|
319
|
Shi Y, Jin J, Ji W, Guan X. Therapeutic landscape in mutational triple negative breast cancer. Mol Cancer 2018; 17:99. [PMID: 30007403 PMCID: PMC6046102 DOI: 10.1186/s12943-018-0850-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/03/2018] [Indexed: 02/08/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogeneous disease with aggressive behavior and poor prognosis. Genomic sequencing has detected a distinctive mutational portrait of both the germline and somatic alterations in TNBC, which is staggeringly different from other breast cancer subtypes. The clinical utility of sequencing germline BRCA1/2 genes has been well established in TNBC. However, for other predisposition genes, studies concerning the risk and penetrance to TNBC are relatively scarce. Very few recurrent mutations, including TP53 and PI3KCA mutations, together with a long tail of individually rare mutations occur in TNBC. These combined effects of genomic alterations drive TNBC progression. Given the complexity and heterogeneity of TNBC, clinical interpretation of the genomic alterations in TNBC may pave a new way for the treatment of TNBC. In this review, we summarized the germline and somatic mutation profiles of TNBC and discussed the current and upcoming therapeutic strategies targeting the mutant proteins or pathways to enable tailored-therapeutics.
Collapse
Affiliation(s)
- Yaqin Shi
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Juan Jin
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Wenfei Ji
- Department of Medical Oncology, Jinling Clinical College, Nanjing Medical University, Nanjing, 210002, China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China. .,Department of Medical Oncology, Jinling Clinical College, Nanjing Medical University, Nanjing, 210002, China. .,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
320
|
Wang X, Tan Y, Cao X, Kim JA, Chen T, Hu Y, Wexler M, Wang X. Epigenetic activation of HORMAD1 in basal-like breast cancer: role in Rucaparib sensitivity. Oncotarget 2018; 9:30115-30127. [PMID: 30046392 PMCID: PMC6059019 DOI: 10.18632/oncotarget.25728] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 06/22/2018] [Indexed: 12/27/2022] Open
Abstract
Basal-like breast cancer (BLBC) is an aggressive breast cancer subtype with features similar to the basal cells surrounding the mammary ducts. Treatment of patients with BLBC has been challenging due to the lack of well-defined molecular targets. Due to the clinical and pathological similarities of BLBC with BRCA-deficient breast cancers, the effectiveness of Poly (ADP-ribose) polymerase inhibitors (PARPi) has been tested in early phase clinical trials for patients with advanced BLBC, with limited clinical responses. Recently, it was reported that HORMAD1 overexpression sensitizes BLBC to HR-targeting agents by suppressing homologous recombination. Our independent analysis suggests that HORMAD1 is aberrantly overexpressed in about 80% of BLBC, and its expression in normal tissues is restricted to testis. Our experimental data suggests that HORMAD1 overexpression correlates with focal hypomethylation in BLBC. On the other hand, investigation of the Genomics of Drug Sensitivity in Cancer dataset revealed significantly reduced sensitivity of HORMAD1-overexpressing BLBC cell lines to Rucaparib, a commonly used PARPi. To further assess the role of HORMAD1 in PARPi sensitivity, we generated three HORMAD1-overexpressing xenograft models using the HORMAD1-low BLBC cell lines HCC1954, HCC1806, and BT20; we then subjected these xenograft models to Rucaparib treatment. Ectopic expression of HORMAD1 enhances tumor formations in two of these models, and significantly reduces sensitivity to Rucaparib in the HCC1954 model. Taken together, our data suggest that epigenetic activation of HORMAD1 by hypomethylation in BLBC may endow reduced sensitivity to Rucaparib treatment in some tumor models.
Collapse
Affiliation(s)
- Xian Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Department of Pathology, University of Pittsburgh, PA 15232, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ying Tan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xixi Cao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Ah Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tianmeng Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Department of Pathology, University of Pittsburgh, PA 15232, USA
| | - Yiheng Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Department of Pathology, University of Pittsburgh, PA 15232, USA
| | - Matthew Wexler
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Department of Pathology, University of Pittsburgh, PA 15232, USA
| | - Xiaosong Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Department of Pathology, University of Pittsburgh, PA 15232, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
321
|
Talisa VB, Yende S, Seymour CW, Angus DC. Arguing for Adaptive Clinical Trials in Sepsis. Front Immunol 2018; 9:1502. [PMID: 30002660 PMCID: PMC6031704 DOI: 10.3389/fimmu.2018.01502] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/18/2018] [Indexed: 12/13/2022] Open
Abstract
Sepsis is life-threatening organ dysfunction due to dysregulated response to infection. Patients with sepsis exhibit wide heterogeneity stemming from genetic, molecular, and clinical factors as well as differences in pathogens, creating challenges for the development of effective treatments. Several gaps in knowledge also contribute: (i) biomarkers that identify patients likely to benefit from specific treatments are unknown; (ii) therapeutic dose and duration is often poorly understood; and (iii) short-term mortality, a common outcome measure, is frequently criticized for being insensitive. To date, the majority of sepsis trials use traditional design features, and have largely failed to identify new treatments with incremental benefit over standard of care. Traditional trials are also frequently conducted as part of a drug evaluation process that is segmented into several phases, each requiring separate trials, with a long time delay from inception through design and execution to incorporation of results into clinical practice. By contrast, adaptive clinical trial designs facilitate the evaluation of several candidate treatments simultaneously, learn from emergent discoveries during the course of the trial, and can be structured efficiently to lead to more timely conclusions compared to traditional trial designs. Adoption of new treatments in clinical practice can be accelerated if these trials are incorporated in electronic health records as part of a learning health system. In this review, we discuss challenges in the evaluation of treatments for sepsis, and explore potential benefits and weaknesses of recent advances in adaptive trial methodologies to address these challenges.
Collapse
Affiliation(s)
| | - Sachin Yende
- Clinical Research, Investigation, and Systems Modeling of Acute Illness Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | | | | |
Collapse
|
322
|
Abstract
Progress in the treatment of triple-negative breast cancer remains an important challenge. Given the aggressive biology and high risk of distant recurrence, systemic chemotherapy is warranted in most patients. Neaodjuvant chemotherapy benefits patients with locally advanced disease by downsizing the tumor and increasing the probability of breast-conserving surgery. Clinical and pathologic responses provide important prognostic information, which makes neoadjuvant therapy an attractive approach for all patients with triple-negative breast cancer. Clinical research in the neoadjuvant setting is focused on improvement in pathologic complete response rates and outcomes of patients with residual disease.
Collapse
|
323
|
Godone RLN, Leitão GM, Araújo NB, Castelletti CHM, Lima-Filho JL, Martins DBG. Clinical and molecular aspects of breast cancer: Targets and therapies. Biomed Pharmacother 2018; 106:14-34. [PMID: 29945114 DOI: 10.1016/j.biopha.2018.06.066] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 12/23/2022] Open
Abstract
Breast Cancer is a complex disease characterized by the occurrence of multiple molecular alterations. Currently, some molecular markers are in use for breast cancer diagnostic, prognostic, and predictive purposes. Thus, genetic signatures are available for improving the decision-making. The biomarkers are also essential as therapeutic approaches, but many questions remain due to the lack of efficacy on breast cancer treatment, mainly for triple-negative breast cancer subtype. Since the genetic profile of breast cancer can also be related to different ethnic groups and geographic areas, the reference populations of the genetic assays and clinical trials need to include a broader population beyond the European and North American patients. In this review, we analyzed the current and potential molecular markers that could help to improve the strategies for breast cancer therapy.
Collapse
Affiliation(s)
- R L N Godone
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil
| | - G M Leitão
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Clinical Hospital of Pernambuco - Professor Romero Marques, Federal University of Pernambuco (UFPE), Brazil
| | - N B Araújo
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil
| | - C H M Castelletti
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Agronomic Institute of Pernambuco (IPA), Recife, Pernambuco, Brazil
| | - J L Lima-Filho
- Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Department of Biochemistry, Federal University of Pernambuco (UFPE), Brazil
| | - D B G Martins
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Department of Biochemistry, Federal University of Pernambuco (UFPE), Brazil.
| |
Collapse
|
324
|
Steenbruggen TG, van Ramshorst MS, Kok M, Linn SC, Smorenburg CH, Sonke GS. Neoadjuvant Therapy for Breast Cancer: Established Concepts and Emerging Strategies. Drugs 2018; 77:1313-1336. [PMID: 28616845 DOI: 10.1007/s40265-017-0774-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the last decade, the systemic treatment approach for patients with early breast cancer has partly shifted from adjuvant treatment to neoadjuvant treatment. Systemic treatment administration started as a 'one size fits all' approach but is currently customized according to each breast cancer subtype. Systemic treatment in a neoadjuvant setting is at least as effective as in an adjuvant setting and has several additional advantages. First, it enables response monitoring and provides prognostic information; second, it downstages the tumor, allowing for less extensive surgery, improved cosmetic outcomes, and reduced postoperative complications such as lymphedema; and third, it enables early development of new treatment strategies by using pathological complete remission as a surrogate outcome of event-free and overall survival. In this review we give an overview of the current standard of neoadjuvant systemic treatment strategies for the three main subtypes of breast cancer: hormone receptor-positive, triple-negative, and human epidermal growth factor receptor 2-positive. Additionally, we summarize drugs that are under investigation for use in the neoadjuvant setting.
Collapse
Affiliation(s)
- Tessa G Steenbruggen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Mette S van Ramshorst
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Marleen Kok
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Sabine C Linn
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Carolien H Smorenburg
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands.
| |
Collapse
|
325
|
Groheux D. Now Is the Time to Use 18F-FDG PET/CT to Optimize Neoadjuvant Treatment in Triple-Negative Breast Cancer! J Nucl Med 2018; 59:863-864. [DOI: 10.2967/jnumed.118.210922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/12/2018] [Indexed: 11/16/2022] Open
|
326
|
Fletcher JI, Ziegler DS, Trahair TN, Marshall GM, Haber M, Norris MD. Too many targets, not enough patients: rethinking neuroblastoma clinical trials. Nat Rev Cancer 2018; 18:389-400. [PMID: 29632319 DOI: 10.1038/s41568-018-0003-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuroblastoma is a rare solid tumour of infancy and early childhood with a disproportionate contribution to paediatric cancer mortality and morbidity. Combination chemotherapy, radiation therapy and immunotherapy remains the standard approach to treat high-risk disease, with few recurrent, actionable genetic aberrations identified at diagnosis. However, recent studies indicate that actionable aberrations are far more common in relapsed neuroblastoma, possibly as a result of clonal expansion. In addition, although the major validated disease driver, MYCN, is not currently directly targetable, multiple promising approaches to target MYCN indirectly are in development. We propose that clinical trial design needs to be rethought in order to meet the challenge of providing rigorous, evidence-based assessment of these new approaches within a fairly small patient population and that experimental therapies need to be assessed at diagnosis in very-high-risk patients rather than in relapsed and refractory patients.
Collapse
Affiliation(s)
- Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Toby N Trahair
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Glenn M Marshall
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia.
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
327
|
Wang X, Shi Y, Huang D, Guan X. Emerging therapeutic modalities of PARP inhibitors in breast cancer. Cancer Treat Rev 2018; 68:62-68. [PMID: 29870916 DOI: 10.1016/j.ctrv.2018.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/26/2022]
Abstract
Inhibition of Poly (ADP-ribose) polymerase (PARP) has shown marked benefit for breast cancer with homologous recombination deficiency, whether driven by defects in BRCA1, BRCA2, or other pathway components. Since the initial approval of olaparib, a mostly investigated PARP inhibitor (PARPi), the clinical development of PARPi in breast cancer treatment has been a major emphasis. Researches in investigating platinum-PARPi combination use compared with platinum monotherapy demonstrated promising benefit in metastatic BRCA mutated breast cancer or TNBC, while no such superiority was observed in the neoadjuvant setting of TNBC. Moreover, the utility of PARP inhibition in BRCA1/2 mutated breast cancer with different platinum-free interval was investigated. There was a clear association between clinical benefit with PARPi and platinum sensitivity, whereas partial efficacy of PARPi still occurs in platinum-resistant patients. In addition, proof-of-principle studies of immunotherapy combined with PARPi in breast cancer have obtained promising results, indicating the potential benefit of the combination therapy in patients with breast cancer. These efforts, contributing to maximize the utility of PARPi, may drive a new era of this agent after its first routine use. In this review, we summarized the utility of combining platinum-PARPi in BRCA mutated breast cancer or TNBC compared with platinum monotherapy and provided promising prospects of PARPi as maintenance therapy in breast cancer, as well as providing a strong rationale for testing immunotherapy combined with PARPi in breast cancer to expand the clinical utility of PARPi.
Collapse
Affiliation(s)
- Xin Wang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yaqin Shi
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Doudou Huang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China; Department of Medical Oncology, Jinling Clinical College, Nanjing Medical University, Nanjing 210002, China.
| |
Collapse
|
328
|
Papadimitriou M, Mountzios G, Papadimitriou CA. The role of PARP inhibition in triple-negative breast cancer: Unraveling the wide spectrum of synthetic lethality. Cancer Treat Rev 2018; 67:34-44. [PMID: 29753961 DOI: 10.1016/j.ctrv.2018.04.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 15-20% of all breast cancers and is characterized by a lack of immunohistochemical expression of estrogen receptors (ER), progesterone receptors (PR) and HER2. TNBC is associated with poor long-term outcomes compared with other breast cancer subtypes. Many of these tumors are also basal-like cancers which are characterized by an aggressive biological behavior with a distant recurrence peak observed early at 3 years following diagnosis. Furthermore, metastatic TNBC bears a dismal prognosis with an average survival of 12 months. Although the prevalence of genetic alterations among women with TNBC differs significantly by ethnicity, race and age, BRCA mutations (including both germline mutations and somatic genetic aberrations) are found in up to 20-25% of unselected patients and especially in those of the basal-like immunophenotype. Therefore, defects in the DNA repair pathway could represent a promising therapeutic target for this subgroup of TNBC patients. Poly(ADP-ribose) polymerase (PARP) inhibitors exploit this deficiency through synthetic lethality and have emerged as promising anticancer therapies, especially in BRCA1 or BRCA2 mutation carriers. Several PARP inhibitors are currently being evaluated in the adjuvant, neo-adjuvant, and metastatic setting for the treatment of breast cancer patients with a deficient homologous recombination pathway. In this article, we review the major molecular characteristics of TNBC, the mechanisms of homologous recombination, and the role of PARP inhibition as an emerging therapeutic strategy.
Collapse
Affiliation(s)
- Marios Papadimitriou
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| | - Giannis Mountzios
- Department of Medical Oncology, 251 Airforce General Hospital, Athens, Greece
| | - Christos A Papadimitriou
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| |
Collapse
|
329
|
Slosberg ED, Kang BP, Peguero J, Taylor M, Bauer TM, Berry DA, Braiteh F, Spira A, Meric-Bernstam F, Stein S, Piha-Paul SA, Salvado A. Signature program: a platform of basket trials. Oncotarget 2018; 9:21383-21395. [PMID: 29765547 PMCID: PMC5940401 DOI: 10.18632/oncotarget.25109] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 03/21/2018] [Indexed: 11/25/2022] Open
Abstract
Investigating targeted therapies can be challenging due to diverse tumor mutations and slow patient accrual for clinical studies. The Signature Program is a series of 8 phase 2, agent-specific basket protocols using a rapid study start-up approach involving no predetermined study sites. Each protocol evaluated 1 agent (buparlisib, dovitinib, binimetinib, encorafenib, sonidegib, BGJ398, ceritinib, or ribociclib) in patients with solid or hematologic malignancies and an actionable mutation. The primary endpoint of each study was the clinical benefit rate (ie, complete or partial response, or stable disease) at 16 weeks. A total of 192 individual sites were opened in the United States, with a median start-up time of 3.6 weeks. The most common tumor types among the 595 treated patients were colorectal (9.2%), non-small cell lung adenocarcinoma (9.1%), and ovarian (8.4%). Frequent genetic alterations were in PIK3CA, RAS, p16, and PTEN. Overall, 30 partial or complete responses were observed with 6 compounds in 16 tumor types. The Signature Program presents a unique and successful approach for rapid signal finding across multiple tumors and allowed various agents to be evaluated in patients with rare alterations. Incorporating these program features in conventional studies could lead to improved trial efficiencies and patient outcomes.
Collapse
Affiliation(s)
- Eric D. Slosberg
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
- Current affiliation: Daiichi Sankyo, Inc, Basking Ridge, NJ, USA
| | | | | | | | - Todd M. Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN, USA
| | - Donald A. Berry
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Berry Consultants, Austin, TX, USA
| | - Fadi Braiteh
- US Oncology Research and Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| | | | | | | | | | - August Salvado
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| |
Collapse
|
330
|
Turk AA, Wisinski KB. PARP inhibitors in breast cancer: Bringing synthetic lethality to the bedside. Cancer 2018; 124:2498-2506. [PMID: 29660759 DOI: 10.1002/cncr.31307] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/29/2022]
Abstract
Individuals with breast and ovarian cancer susceptibility gene 1 (BRCA1) or BRCA2 germline mutations have a significantly increased lifetime risk for breast and ovarian cancers. BRCA-mutant cancer cells have abnormal homologous recombination (HR) repair of DNA. In these tumors, the base excision repair (BER) pathway is important for cell survival. The poly(adenosine diphosphate-ribose) polymerase (PARP) enzymes play a key role in BER, and PARP inhibitors are effective in causing cell death in BRCA-mutant cells while sparing normal cells-a concept called synthetic lethality. PARP inhibitors are the first cancer therapeutics designed to exploit synthetic lethality. Recent clinical trials in BRCA-mutant, metastatic breast cancer demonstrated improved outcomes with single-agent PARP inhibitors (olaparib and talazoparib) over chemotherapy. However, resistance to PARP inhibitors remains a challenge. Primarily due to myelosuppression, the combination of PARP inhibitors with chemotherapy has been difficult. Novel combinations with chemotherapy, immunotherapy, and other targeted therapies are being pursued. In this review, the authors discuss current knowledge of PARP inhibitors in BRCA-mutant breast cancer and potential future directions for these agents. Cancer 2018;124:2498-506. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Anita A Turk
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Kari B Wisinski
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| |
Collapse
|
331
|
Abstract
OPINION STATEMENT Triple-negative breast cancer (TNBC) accounts for 15% of all breast cancers and is associated with poor long-term outcomes compared to other breast cancer subtypes. Currently, chemotherapy remains the main modality of treatment for early-stage TNBC, as there is no approved targeted therapy for this subtype. The biologic heterogeneity of TNBC has hindered the development and evaluation of novel agents, but recent advancements in subclassifying TNBC have paved the way for further investigation of more effective systemic therapies, including cytotoxic and targeted agents. TNBC is enriched for germline BRCA mutation and for somatic deficiencies in homologous recombination DNA repair, the so-called "BRCAness" phenotype. Together, germline BRCA mutations and BRCAness are promising biomarkers of susceptibility to DNA-damaging therapy. Various investigational approaches are consequently being investigated in early-stage TNBC, including immune checkpoint inhibitors, platinum compounds, PI3K pathway inhibitors, and androgen receptor inhibitors. Due to the biological diversity found within TNBC, patient selection based on molecular biomarkers could aid the design of early-phase clinical trials, ultimately accelerating the clinical application of effective new agents. TNBC is an aggressive breast cancer subtype, for which multiple targeted approaches will likely be required for patient outcomes to be substantially improved.
Collapse
|
332
|
Abstract
OPINION STATEMENT The single agent activity of PARP inhibitors (PARPi) in germline BRCA mutated (gBRCAm) breast and ovarian cancer suggests untapped potential for this new class of drug in breast cancer. The US Food and Drug Administration has approved three PARPi (olaparib, rucaparib, and niraparib) so far to treat certain ovarian cancers, including those with gBRCAm and olaparib for treatment of gBRCAm breast cancers. Several PARPi are now under clinical development for breast cancer in the various treatment settings. Recently, two phase III trials of olaparib (OlympiaD) and talazoparib (EMBRACA) demonstrated 3-month progression-free survival improvement with PARPi compared to physician's choice single agent chemotherapy in metastatic gBRCAm breast cancer. To date, PARPi seems less efficacious in metastatic breast cancer patients than those with BRCA mutated platinum-sensitive recurrent ovarian cancer, perhaps reflecting the biologic heterogeneity and low somatic BRCA mutation rate in breast cancer. The use of PARPi is gradually evolving, including combination strategies with chemotherapy, targeted agents, radiotherapy, or immunotherapy in women with and without gBRCAm. The role of predictive biomarkers, including molecular signatures and homologous recombination repair deficiency scores based on loss of heterozygosity and other structural genomic aberrations, will be crucial to identify a subgroup of patients who may have benefit from PARPi. An improved understanding of the mechanisms underlying PARPi clinical resistance will also be important to enable the development of new approaches to increase efficacy. This is a field rich in opportunity, and the coming years should see a better understanding of which breast cancer patients we should treat with PARPi and where these agents should come in over the course of treatment.
Collapse
Affiliation(s)
- Alexandra S Zimmer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Dr. MSC1906 Building 10, Room 4B54, Bethesda, MD, 20892-1906, USA.
| | - Mitchell Gillard
- School of Medicine, Stony Brook University School of Medicine, 101 Nicolls Road Stony Brook, Bethesda, NY, 11794-8434, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Dr. MSC1906 Building 10, Room 4B54, Bethesda, MD, 20892-1906, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, 10 Center Dr. MSC1906 Building 10, Room 4B54, Bethesda, MD, 20892-1906, USA
| |
Collapse
|
333
|
Loibl S, O'Shaughnessy J, Untch M, Sikov WM, Rugo HS, McKee MD, Huober J, Golshan M, von Minckwitz G, Maag D, Sullivan D, Wolmark N, McIntyre K, Ponce Lorenzo JJ, Metzger Filho O, Rastogi P, Symmans WF, Liu X, Geyer CE. Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): a randomised, phase 3 trial. Lancet Oncol 2018; 19:497-509. [DOI: 10.1016/s1470-2045(18)30111-6] [Citation(s) in RCA: 528] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/27/2022]
|
334
|
Kempf L, Goldsmith JC, Temple R. Challenges of developing and conducting clinical trials in rare disorders. Am J Med Genet A 2018; 176:773-783. [PMID: 28815894 DOI: 10.1002/ajmg.a.38413] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/13/2022]
Abstract
Rare disease drug development is a rapidly expanding field. Clinical researchers in rare diseases face many challenges when conducting trials in small populations. Disease natural history is often poorly understood and the ability to detect clinically meaningful outcomes requires understanding of their rate of occurrence and variability, both of which contribute to difficulties in powering a study. Standard trial designs are not optimized to obtain adequate safety and efficacy data from small numbers of patients, so alternative designs (enrichment, crossover, adaptive, N-of 1) need to be considered. The affected patients can be hard to identify, especially early in the course of their disease, are generally geographically dispersed, and are often children. Trials are frequently conducted on an international scale and may be subject to complex or multiple regulatory agency oversights and may be affected by local customs, cultures, and practices. A basic understanding of the FDA programs supporting development of drugs for rare diseases is provided by this review and the role of early consultation with the FDA is emphasized. Of recent FDA New Molecular Entities (NME) approvals, 41% (17 approvals) in 2014, 47% (21 approvals) in 2015, and 41% (9 approvals) in 2016 were for rare disease indications. Through effective interactions and collaborations with physicians, institutions, and patient groups, sponsors have been successful in bringing new treatments to market for individuals affected by rare diseases. Challenges to drug development have been overcome through the focused efforts of patients/families, non-profit patient advocacy groups, drug developers, and regulatory authorities.
Collapse
Affiliation(s)
- Lucas Kempf
- Rare Diseases Program, Silver Spring, Maryland
| | | | - Robert Temple
- Office of Drug Evaluation I/Office of New Drugs/CDER/FDA, Silver Spring, Maryland
| |
Collapse
|
335
|
Sizemore ST, Mohammad R, Sizemore GM, Nowsheen S, Yu H, Ostrowski MC, Chakravarti A, Xia F. Synthetic Lethality of PARP Inhibition and Ionizing Radiation is p53-dependent. Mol Cancer Res 2018; 16:1092-1102. [PMID: 29592899 DOI: 10.1158/1541-7786.mcr-18-0106] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
PARP inhibitors (PARPi) are potentially effective therapeutic agents capable of inducing synthetic lethality in tumors with deficiencies in homologous recombination (HR)-mediated DNA repair such as those carrying BRCA1 mutations. However, BRCA mutations are rare, the majority of tumors are proficient in HR repair, and thus most tumors are resistant to PARPi. Previously, we observed that ionizing radiation (IR) initiates cytoplasmic translocation of BRCA1 leading to suppression of HR-mediated DNA repair and induction of synthetic PARPi lethality in wild-type BRCA1 and HR-proficient tumor cells. The tumor suppressor p53 was identified as a key factor that regulates DNA damage-induced BRCA1 cytoplasmic sequestration following IR. However, the role of p53 in IR-induced PARPi sensitization remains unclear. This study elucidates the role of p53 in IR-induced PARPi cytotoxicity in HR-proficient cancer cells and suggests p53 status may help define a patient population that might benefit from this treatment strategy. Sensitization to PARPi following IR was determined in vitro and in vivo utilizing human breast and glioma tumor cells carrying wild-type BRCA1 and p53, and in associated cells in which p53 function was modified by knockdown or mutation. In breast and glioma cells with proficient HR repair, IR-induced BRCA1 cytoplasmic sequestration, HR repair inhibition, and subsequent PARPi sensitization in vitro and in vivo was dependent upon functional p53.Implications: Implications: p53 status determines PARP inhibitor sensitization by ionizing radiation in multiple BRCA1 and HR-proficient tumor types and may predict which patients are most likely to benefit from combination therapy. Mol Cancer Res; 16(7); 1092-102. ©2018 AACR.
Collapse
Affiliation(s)
- Steven T Sizemore
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Rahman Mohammad
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Gina M Sizemore
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Clinic, Rochester, Minnesota
| | - Hao Yu
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Fen Xia
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
336
|
Abstract
Adaptive clinical trials are an innovative trial design aimed at reducing resources, decreasing time to completion and number of patients exposed to inferior interventions, and improving the likelihood of detecting treatment effects. The last decade has seen an increasing use of adaptive designs, particularly in drug development. They frequently differ importantly from conventional clinical trials as they allow modifications to key trial design components during the trial, as data is being collected, using preplanned decision rules. Adaptive designs have increased likelihood of complexity and also potential bias, so it is important to understand the common types of adaptive designs. Many clinicians and investigators may be unfamiliar with the design considerations for adaptive designs. Given their complexities, adaptive trials require an understanding of design features and sources of bias. Herein, we introduce some common adaptive design elements and biases and specifically address response adaptive randomization, sample size reassessment, Bayesian methods for adaptive trials, seamless trials, and adaptive enrichment using real examples.
Collapse
Affiliation(s)
- Jay Jh Park
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kristian Thorlund
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, ON, Canada.,The Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - Edward J Mills
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, ON, Canada.,The Bill and Melinda Gates Foundation, Seattle, WA, USA
| |
Collapse
|
337
|
|
338
|
Janku F, Yap TA, Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol 2018; 15:273-291. [PMID: 29508857 DOI: 10.1038/nrclinonc.2018.28] [Citation(s) in RCA: 764] [Impact Index Per Article: 109.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The PI3K-AKT-mTOR pathway is one of the most frequently dysregulated pathways in cancer and, consequently, more than 40 compounds that target key components of this signalling network have been tested in clinical trials involving patients with a range of different cancers. The clinical development of many of these agents, however, has not advanced to late-phase randomized trials, and the antitumour activity of those that have been evaluated in comparative prospective studies has typically been limited, or toxicities were found to be prohibitive. Nevertheless, the mTOR inhibitors temsirolimus and everolimus and the PI3K inhibitors idelalisib and copanlisib have been approved by the FDA for clinical use in the treatment of a number of different cancers. Novel compounds with greater potency and selectivity, as well as improved therapeutic indices owing to reduced risks of toxicity, are clearly required. In addition, biomarkers that are predictive of a response, such as PIK3CA mutations for inhibitors of the PI3K catalytic subunit α isoform, must be identified and analytically and clinically validated. Finally, considering that oncogenic activation of the PI3K-AKT-mTOR pathway often occurs alongside pro-tumorigenic aberrations in other signalling networks, rational combinations are also needed to optimize the effectiveness of treatment. Herein, we review the current experience with anticancer therapies that target the PI3K-AKT-mTOR pathway.
Collapse
Affiliation(s)
- Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
339
|
Curigliano G. Addition of platinum salts to neoadjuvant chemotherapy in triple-negative breast cancer: a new standard of care? Lancet Oncol 2018; 19:434-436. [PMID: 29501365 DOI: 10.1016/s1470-2045(18)30129-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, 20141 Milan, Italy.
| |
Collapse
|
340
|
Adjuvant versus neoadjuvant chemotherapy in triple-negative breast cancer patients with BRCA mutations. Breast Cancer Res Treat 2018; 170:101-109. [PMID: 29470805 DOI: 10.1007/s10549-018-4727-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/19/2018] [Indexed: 11/25/2022]
Abstract
PURPOSE As triple-negative breast cancers are associated with earlier recurrences and poorer survival, optimal treatment of early-stage breast cancer is essential. Several retrospective studies in triple-negative breast cancer have reported conflicting results in overall survival in patients receiving neoadjuvant or adjuvant systemic therapy. This study aims to analyze outcomes of adjuvant versus neoadjuvant in patients with early-stage triple-negative breast cancer with and without BRCA germline mutations. METHODS Patients with stage I or II triple-negative breast cancer who had BRCA testing were identified from a prospective cohort study of 4027 patients. Clinical, demographic, genetic test results, chemotherapy, recurrence, and survival data were analyzed. Overall survival and disease-free survival were estimated using the Kaplan-Meier method. RESULTS 319 patients with stage I and II triple-negative breast cancer who met eligibility criteria were included in the analysis. 187 received adjuvant chemotherapy (58.6%) and 132 received neoadjuvant chemotherapy (41.4%). 135 were BRCA positive (42.3%) and 184 were BRCA negative (57.7%). There was no significant association between overall survival or disease-free survival and treatment with neoadjuvant versus adjuvant in the overall cohort. Furthermore, there were no significant differences between patient subgroups (neoadjuvant BRCA positive, neoadjuvant BRCA negative, adjuvant BRCA positive, and adjuvant BRCA negative) with respect to either overall survival or disease-free survival. CONCLUSIONS Neoadjuvant versus adjuvant with standard anthracycline- and taxane-containing regimens results in similar disease-free survival and overall survival among patients with stage I and II triple-negative breast cancer regardless of BRCA status. Further studies are needed to evaluate whether similar results are observed with newer agents.
Collapse
|
341
|
Alexander BM, Ba S, Berger MS, Berry DA, Cavenee WK, Chang SM, Cloughesy TF, Jiang T, Khasraw M, Li W, Mittman R, Poste GH, Wen PY, Yung WKA, Barker AD. Adaptive Global Innovative Learning Environment for Glioblastoma: GBM AGILE. Clin Cancer Res 2018; 24:737-743. [PMID: 28814435 DOI: 10.1158/1078-0432.ccr-17-0764] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/05/2017] [Accepted: 08/10/2017] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is a deadly disease with few effective therapies. Although much has been learned about the molecular characteristics of the disease, this knowledge has not been translated into clinical improvements for patients. At the same time, many new therapies are being developed. Many of these therapies have potential biomarkers to identify responders. The result is an enormous amount of testable clinical questions that must be answered efficiently. The GBM Adaptive Global Innovative Learning Environment (GBM AGILE) is a novel, multi-arm, platform trial designed to address these challenges. It is the result of the collective work of over 130 oncologists, statisticians, pathologists, neurosurgeons, imagers, and translational and basic scientists from around the world. GBM AGILE is composed of two stages. The first stage is a Bayesian adaptively randomized screening stage to identify effective therapies based on impact on overall survival compared with a common control. This stage also finds the population in which the therapy shows the most promise based on clinical indication and biomarker status. Highly effective therapies transition in an inferentially seamless manner in the identified population to a second confirmatory stage. The second stage uses fixed randomization to confirm the findings from the first stage to support registration. Therapeutic arms with biomarkers may be added to the trial over time, while others complete testing. The design of GBM AGILE enables rapid clinical testing of new therapies and biomarkers to speed highly effective therapies to clinical practice. Clin Cancer Res; 24(4); 737-43. ©2017 AACR.
Collapse
Affiliation(s)
- Brian M Alexander
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Sujuan Ba
- National Foundation for Cancer Research, Bethesda, Maryland
| | - Mitchel S Berger
- Department for Neurological Surgery, University of California-San Francisco, San Francisco, California
| | - Donald A Berry
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Berry Consultants, Austin, Texas
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California-San Diego, La Jolla, California
| | - Susan M Chang
- Department for Neurological Surgery, University of California-San Francisco, San Francisco, California
| | - Timothy F Cloughesy
- Neuro-Oncology Program, University of California-Los Angeles, Los Angeles, California
| | - Tao Jiang
- Department of Clinical Oncology, Capital Medical University, Beijing, China
| | - Mustafa Khasraw
- NHMRC Clinical Trials Centre, The University of Sydney Medical School, Australia
| | - Wenbin Li
- Glioma Department, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Robert Mittman
- School of Biological and Health Systems Engineering, School of Computing, Informatics, and Decision Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona
- National Biomarker Development Alliance, Arizona State University, Tempe, Arizona
| | - George H Poste
- National Biomarker Development Alliance, Arizona State University, Tempe, Arizona
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, Arizona
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - W K Alfred Yung
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anna D Barker
- National Biomarker Development Alliance, Arizona State University, Tempe, Arizona
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| |
Collapse
|
342
|
McAndrew N, DeMichele A. Neoadjuvant Chemotherapy Considerations in Triple-Negative Breast Cancer. THE JOURNAL OF TARGETED THERAPIES IN CANCER 2018; 7:52-69. [PMID: 29577076 PMCID: PMC5865448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The optimal neoadjuvant chemotherapy (NACT) regimen in triple-negative breast cancer (TNBC) has not been clearly defined. Achieving a pathologic complete response (pCR) provides important prognostic information, and, especially in TNBC, is considered a surrogate endpoint for event-free survival. Thus, many neoadjuvant studies in TNBC focus on this as a primary endpoint, and such information may be used for accelerated US Food and Drug Administration approval. Current controversies in the field include: (1) the role of platinum-based compounds; (2) the optimal chemotherapy backbone; and (3) the benefits of additional therapy after surgery. Conflicting results of 2 major studies adding carboplatin to NACT have highlighted the need to balance potential benefits to disease outcomes against increased toxicity. While the PROGECT study suggests efficacy of a nonanthracycline-containing regimen, this is observational data, and evidence in the form of a clinical trial remains to be seen. Data surrounding optimal taxane use support the use of nab-paclitaxel in place of paclitaxel in limited clinical situations. Although bevacizumab may increase pCR rates, this has not translated into survival benefit. Capecitabine shows promise in patients who have not achieved pCR after NACT. The neoadjuvant setting remains an important model for drug development. This review will focus on the most important and most current neoadjuvant trials in women with TNBC.
Collapse
Affiliation(s)
- Nicholas McAndrew
- Department of Medicine, Division of Hematology/Oncology at the University of Pennsylvania
| | - Angela DeMichele
- Department of Medicine, Division of Hematology/Oncology at the University of Pennsylvania; Also at the Abramson Cancer Center, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania
| |
Collapse
|
343
|
Amer AM, Zaid M, Chaudhury B, Elganainy D, Lee Y, Wilke CT, Cloyd J, Wang H, Maitra A, Wolff RA, Varadhachary G, Overman MJ, Lee JE, Fleming JB, Tzeng CW, Katz MH, Holliday EB, Krishnan S, Minsky BD, Herman JM, Taniguchi CM, Das P, Crane CH, Le O, Bhosale P, Tamm EP, Koay EJ. Imaging-based biomarkers: Changes in the tumor interface of pancreatic ductal adenocarcinoma on computed tomography scans indicate response to cytotoxic therapy. Cancer 2018; 124:1701-1709. [PMID: 29370450 PMCID: PMC5891375 DOI: 10.1002/cncr.31251] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/22/2017] [Accepted: 12/21/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The assessment of pancreatic ductal adenocarcinoma (PDAC) response to therapy remains challenging. The objective of this study was to investigate whether changes in the tumor/parenchyma interface are associated with response. METHODS Computed tomography (CT) scans before and after therapy were reviewed in 4 cohorts: cohort 1 (99 patients with stage I/II PDAC who received neoadjuvant chemoradiation and surgery); cohort 2 (86 patients with stage IV PDAC who received chemotherapy), cohort 3 (94 patients with stage I/II PDAC who received protocol‐based neoadjuvant gemcitabine chemoradiation), and cohort 4 (47 patients with stage I/II PDAC who received neoadjuvant chemoradiation and were prospectively followed in a registry). The tumor/parenchyma interface was visually classified as either a type I response (the interface remained or became well defined) or a type II response (the interface became poorly defined) after therapy. Consensus (cohorts 1‐3) and individual (cohort 4) visual scoring was performed. Changes in enhancement at the interface were quantified using a proprietary platform. RESULTS In cohort 1, type I responders had a greater probability of achieving a complete or near‐complete pathologic response (21% vs 0%; P = .01). For cohorts 1, 2, and 3, type I responders had significantly longer disease‐free and overall survival, independent of traditional covariates of outcomes and of baseline and normalized cancer antigen 19‐9 levels. In cohort 4, 2 senior radiologists achieved a κ value of 0.8, and the interface score was associated with overall survival. The quantitative method revealed high specificity and sensitivity in classifying patients as type I or type II responders (with an area under the receiver operating curve of 0.92 in cohort 1, 0.96 in cohort 2, and 0.89 in cohort 3). CONCLUSIONS Changes at the PDAC/parenchyma interface may serve as an early predictor of response to therapy. Cancer 2018;124:1701‐9. © 2018 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial‐NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made. An imaging feature of pancreatic cancer is identified that indicates a response to cytotoxic therapies. This may be helpful as an early predictor of response for clinical trials and for deciding whether to change therapy.
Collapse
Affiliation(s)
- Ahmed M Amer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mohamed Zaid
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Baishali Chaudhury
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dalia Elganainy
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yeonju Lee
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher T Wilke
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jordan Cloyd
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gauri Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffery E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa Bay, Florida
| | - Ching Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew H Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emma B Holliday
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bruce D Minsky
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph M Herman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cullen M Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher H Crane
- Department of Radiation Oncology, Memorial Sloan Cancer Center, New York, New York
| | - Ott Le
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Priya Bhosale
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric P Tamm
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
344
|
Peck RW. Precision Medicine Is Not Just Genomics: The Right Dose for Every Patient. Annu Rev Pharmacol Toxicol 2018; 58:105-122. [DOI: 10.1146/annurev-pharmtox-010617-052446] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Richard W. Peck
- Pharma Research and Exploratory Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| |
Collapse
|
345
|
Manem VSK, Salgado R, Aftimos P, Sotiriou C, Haibe-Kains B. Network science in clinical trials: A patient-centered approach. Semin Cancer Biol 2017; 52:135-150. [PMID: 29278737 DOI: 10.1016/j.semcancer.2017.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 02/08/2023]
Abstract
There has been a paradigm shift in translational oncology with the advent of novel molecular diagnostic tools in the clinic. However, several challenges are associated with the integration of these sophisticated tools into clinical oncology and daily practice. High-throughput profiling at the DNA, RNA and protein levels (omics) generate a massive amount of data. The analysis and interpretation of these is non-trivial but will allow a more thorough understanding of cancer. Linear modelling of the data as it is often used today is likely to limit our understanding of cancer as a complex disease, and at times under-performs to capture a phenotype of interest. Network science and systems biology-based approaches, using machine learning and network science principles, that integrate multiple data sources, can uncover complex changes in a biological system. This approach will integrate a large number of potential biomarkers in preclinical studies to better inform therapeutic decisions and ultimately make substantial progress towards precision medicine. It will however require development of a new generation of clinical trials. Beyond discussing the challenges of high-throughput technologies, this review will develop a framework on how to implement a network science approach in new clinical trial designs in order to advance cancer care.
Collapse
Affiliation(s)
- Venkata S K Manem
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Brussels, Belgium; Department of Pathology, GZA Hospitals Antwerp, Belgium
| | - Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Brussels, Belgium; Medical Oncology Clinic, Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Haibe-Kains
- Bioinformatics and Computational Genomics Laboratory, Princess Margaret Cancer Center, Toronto, ON, Canada; Department of Computer Science, University of Toronto, Toronto, ON, Canada; Ontario Institute of Cancer Research, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
346
|
Bergen ES, Bartsch R. My burning issues in the neoadjuvant treatment for breast cancer. MEMO 2017; 11:27-30. [PMID: 29606978 PMCID: PMC5862920 DOI: 10.1007/s12254-017-0378-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 06/08/2023]
Abstract
A combination of anthracyclines and taxanes remains the standard of care for neoadjuvant chemotherapy (NACT) resulting in increased breast conservation rate (BCR) and decreased recurrence rate [1]. Whether pathological complete response (pCR) could be an appropriate surrogate parameter for long-term survival is still a matter of debate. In patients with triple-negative breast cancer (TNBC) and HER2-positive breast cancer (BC), a six to nine times higher risk for relapse has been reported if no pCR was achieved [2, 3]. Within these aggressive subtypes the strongest association between pCR and long-term outcome could be observed [4]. However, a pooled analysis of recently conducted trials could only identify pCR as a surrogate endpoint for improved event-free survival (EFS) and overall survival (OS) on an individual patient level as opposed to the trial level [5]. Even in TNBC, demonstrating that an increased pCR converts into a significant survival benefit would require a study population markedly larger than calculated for previously conducted trials [6, 7].
Collapse
Affiliation(s)
- Elisabeth S. Bergen
- Comprehensive Cancer Center, Vienna, Austria
- Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria
| | - Rupert Bartsch
- Comprehensive Cancer Center, Vienna, Austria
- Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria
- German Breast Group, Neu-Isenburg, Germany
| |
Collapse
|
347
|
Effect of exosome biomarkers for diagnosis and prognosis of breast cancer patients. Clin Transl Oncol 2017; 20:906-911. [PMID: 29143228 DOI: 10.1007/s12094-017-1805-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Exosomes are gradually detected as an indicator for diagnosis and prognosis of breast cancer in clinic and a systematic review was conducted. METHODS A search for clinical studies published before July 1, 2017 was performed. Methods of exosome purification and identification from all studies were extracted. For diagnosis evaluation, the comparison of exosome biomarkers expression between breast cancer patients and healthy women was obtained; for prognosis prediction, the correlation between exosome biomarkers expression and chemotherapy resistance, overall survival (OS), disease-free survival (DFS), recurrence and metastasis of breast cancer was also extracted. RESULTS A total of 11 studies with 921 breast cancer patients were included. Ultracentrifugation is the most frequent method to purify exosomes and transmission electron microscopy is commonly used to identify exosomes. Exosome biomarkers (such as HER2, CD47, Del-1, miR-1246 and miR-21) in breast cancer patients are significantly higher than those in healthy controls, exosomal GSTP1 and TRPC5 are related to chemotherapy resistance, exosome-carrying TRPC5, NANOG, NEUROD1, HTR7, KISS1R and HOXC are correlated to PFS, DFS or OS, and some exosomal proteins (HER2, KDR, CD49d, CXCR4 and CD44) as well as miRNAs (miR-340-5p, miR-17-5p, miR-130a-3p, miR-93-5p) are associated with tumor recurrence or distant organ metastasis. CONCLUSIONS Exosome biomarkers can be used for early diagnosis and prognosis of breast cancer patients in clinic.
Collapse
|
348
|
Precision medicine for urothelial bladder cancer: update on tumour genomics and immunotherapy. Nat Rev Urol 2017; 15:92-111. [PMID: 29133939 DOI: 10.1038/nrurol.2017.179] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Effective management of advanced urothelial bladder cancer is challenging. New discoveries that improve our understanding of molecular bladder cancer subtypes have revealed numerous potentially targetable genomic alterations and demonstrated the efficacy of treatments that harness the immune system. These findings have begun to change paradigms of bladder cancer therapy. For example, DNA repair pathway mutations in genes such as ERCC2, FANCC, ATM, RB1, and others can predict responses to neoadjuvant platinum-based chemotherapies and to targeted therapies on the basis of mutation status. Furthermore, an increasing number of pan-cancer clinical trials (commonly referred to as basket or umbrella trials) are enrolling patients on the basis of molecular and genetic predictors of response. These studies promise to provide improved insight into the true utility of personalized medicine in the treatment of bladder cancer and many other cancer types. Finally, therapies that modulate immune responses have shown great benefit in many cancer types. Several immune checkpoint inhibitors that target programmed cell death protein 1 (PD1), its ligand PDL1, and cytotoxic T lymphocyte-associated protein 4 (CTLA4) have already been approved for use in bladder cancer, representing the most important change to the urological oncologist's tool-kit in over a decade. These advances also provide opportunities for personalization of bladder cancer therapy.
Collapse
|
349
|
Wang C, Kar S, Lai X, Cai W, Arfuso F, Sethi G, Lobie PE, Goh BC, Lim LHK, Hartman M, Chan CW, Lee SC, Tan SH, Kumar AP. Triple negative breast cancer in Asia: An insider's view. Cancer Treat Rev 2017; 62:29-38. [PMID: 29154023 DOI: 10.1016/j.ctrv.2017.10.014] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/29/2017] [Accepted: 10/31/2017] [Indexed: 12/31/2022]
Abstract
While tremendous improvement has been made for the treatment of breast cancers, the treatment of triple negative breast cancer (TNBC) still remains a challenge due to its aggressive characteristics and limited treatment options. Most of the studies on TNBC were conducted in Western population and TNBC is reported to be more frequent in the African women. This review encapsulates the studies conducted on TNBC patients in Asian population and elucidates the similarities and differences between these two regions. The current treatment of TNBC includes surgery, radiotherapy and chemotherapy. In addition to the current chemotherapies, which mainly include cytotoxic agents, such as taxanes and anthracyclines, many clinical trials are investigating the potential use of other chemotherapy drugs, targeted therapeutics and combinational therapies to treat TNBC. Moreover, this review also integrates the studies involving novel markers, which will help us to dissect the pathologic process of TNBC and in turn facilitate the development of better treatment strategies to combat TNBC.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shreya Kar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xianning Lai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wanpei Cai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Peter E Lobie
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School at Shenzhen, Shenzhen, China
| | - Boon C Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore
| | - Lina H K Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; NUS Immunology Program, National University of Singapore, Singapore
| | - Mikael Hartman
- Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; Department of Surgery, National University Cancer Institute, National University Health System, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Ching W Chan
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore
| | - Soo C Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore
| | - Sing H Tan
- Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore; OncoCare Cancer Centre, Gleneagles Medical Centre, Singapore.
| | - Alan P Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
350
|
Tao JJ, Schram AM, Hyman DM. Basket Studies: Redefining Clinical Trials in the Era of Genome-Driven Oncology. Annu Rev Med 2017; 69:319-331. [PMID: 29120700 DOI: 10.1146/annurev-med-062016-050343] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Understanding a tumor's detailed molecular profile has become increasingly necessary to deliver the standard of care for patients with advanced cancer. Innovations in both tumor genomic sequencing technology and the development of drugs that target molecular alterations have fueled recent gains in genome-driven oncology care. "Basket studies," or histology-agnostic clinical trials in genomically selected patients, represent one important research tool to continue making progress in this field. We review key aspects of genome-driven oncology care, including the purpose and utility of basket studies, biostatistical considerations in trial design, genomic knowledgebase development, and patient matching and enrollment models, which are critical for translating our genomic knowledge into clinically meaningful outcomes.
Collapse
Affiliation(s)
- Jessica J Tao
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; , ,
| | - Alison M Schram
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; , ,
| | - David M Hyman
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; , , .,Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|