201
|
Gyawali B, Niraula S. Lessons From Adaptive Randomization: Spying the I-SPY2 Trial in Breast Cancer. J Natl Compr Canc Netw 2020; 18:1441-1444. [PMID: 33152697 DOI: 10.6004/jnccn.2020.7648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
202
|
Coates JTT, Rodriguez-Berriguete G, Puliyadi R, Ashton T, Prevo R, Wing A, Granata G, Pirovano G, McKenna GW, Higgins GS. The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress. Cell Death Discov 2020; 6:110. [PMID: 33133645 PMCID: PMC7591508 DOI: 10.1038/s41420-020-00343-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Platinum chemotherapies are highly effective cytotoxic agents but often induce resistance when used as monotherapies. Combinatorial strategies limit this risk and provide effective treatment options for many cancers. Here, we repurpose atovaquone (ATQ), a well-tolerated & FDA-approved anti-malarial agent by demonstrating that it potentiates cancer cell death of a subset of platinums. We show that ATQ in combination with carboplatin or cisplatin induces striking and repeatable concentration- and time-dependent cell death sensitization in vitro across a variety of cancer cell lines. ATQ induces mitochondrial reactive oxygen species (mROS), depleting intracellular glutathione (GSH) pools in a concentration-dependent manner. The superoxide dismutase mimetic MnTBAP rescues ATQ-induced mROS production and pre-loading cells with the GSH prodrug N-acetyl cysteine (NAC) abrogates the sensitization. Together, these findings implicate ATQ-induced oxidative stress as key mediator of the sensitizing effect. At physiologically achievable concentrations, ATQ and carboplatin furthermore synergistically delay the growth of three-dimensional avascular spheroids. Clinically, ATQ is a safe and specific inhibitor of the electron transport chain (ETC) and is concurrently being repurposed as a candidate tumor hypoxia modifier. Together, these findings suggest that ATQ is deserving of further study as a candidate platinum sensitizing agent.
Collapse
Affiliation(s)
| | | | - Rathi Puliyadi
- Department of Oncology, University of Oxford, Oxford, UK
| | - Thomas Ashton
- Department of Oncology, University of Oxford, Oxford, UK
| | - Remko Prevo
- Department of Oncology, University of Oxford, Oxford, UK
| | - Archie Wing
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
203
|
Hewitt K, Son J, Glencer A, Borowsky AD, Cooperberg MR, Esserman LJ. The Evolution of Our Understanding of the Biology of Cancer Is the Key to Avoiding Overdiagnosis and Overtreatment. Cancer Epidemiol Biomarkers Prev 2020; 29:2463-2474. [PMID: 33033145 DOI: 10.1158/1055-9965.epi-20-0110] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/06/2020] [Accepted: 10/01/2020] [Indexed: 11/16/2022] Open
Abstract
There has been a tremendous evolution in our thinking about cancer since the 1880s. Breast cancer is a particularly good example to evaluate the progress that has been made and the new challenges that have arisen due to screening that inadvertently identifies indolent lesions. The degree to which overdiagnosis is a problem depends on the reservoir of indolent disease, the disease heterogeneity, and the fraction of the tumors that have aggressive biology. Cancers span the spectrum of biological behavior, and population-wide screening increases the detection of tumors that may not cause harm within the patient's lifetime or may never metastasize or result in death. Our approach to early detection will be vastly improved if we understand, address, and adjust to tumor heterogeneity. In this article, we use breast cancer as a case study to demonstrate how the approach to biological characterization, diagnostics, and therapeutics can inform our approach to screening, early detection, and prevention. Overdiagnosis can be mitigated by developing diagnostics to identify indolent disease, incorporating biology and risk assessment in screening strategies, changing the pathology rules for tumor classification, and refining the way we classify precancerous lesions. The more the patterns of cancers can be seen across other cancers, the more it is clear that our approach should transcend organ of origin. This will be particularly helpful in advancing the field by changing both our terminology for what is cancer and also by helping us to learn how best to mitigate the risk of the most aggressive cancers.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Kelly Hewitt
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Jennifer Son
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Alexa Glencer
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Alexander D Borowsky
- Department of Pathology, University of California, Davis, Davis, California.,Athena Breast Health Network
| | - Matthew R Cooperberg
- Department of Urology, University of California, San Francisco, San Francisco, California.,Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, California
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, California. .,Athena Breast Health Network
| |
Collapse
|
204
|
Mechanism of action biomarkers predicting response to AKT inhibition in the I-SPY 2 breast cancer trial. NPJ Breast Cancer 2020; 6:48. [PMID: 33083527 PMCID: PMC7532145 DOI: 10.1038/s41523-020-00189-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
The AKT inhibitor MK2206 (M) was evaluated in I-SPY 2 and graduated in
the HER2+, HR−, and HR− HER2+ signatures. We hypothesized that AKT signaling axis
proteins/genes may specifically predict response to M and tested 26 phospho-proteins
and 10 genes involved in AKT-mTOR-HER signaling; in addition, we tested 9 genes from
a previous study in the metastatic setting. One hundred and fifty patients had gene
expression data from pretreatment biopsies available for analysis (M: 94, control:
56) and 138 had protein data (M: 87, control: 51). Logistic modeling was used to
assess biomarker performance in pre-specified analysis. In general, phospho-protein
biomarkers of activity in the AKT-mTOR-HER pathway appeared more predictive of
response to M than gene expression or total protein biomarkers in the same pathway;
however, the nature of the predictive biomarkers differed in the HER2+ and TN
groups. In the HER2+ subset, patients achieving a pCR in M had higher levels of
multiple AKT kinase substrate phospho-proteins (e.g., pmTOR, pTSC2). In contrast, in
the TN subset responding patients had lower levels of AKT pathway phospho-proteins,
such as pAKT, pmTOR, and pTSC2. Pathway mutations did not appear to account for
these associations. Additional exploratory whole-transcriptome analysis revealed
immune signaling as strongly associated with response to M in the HER2+ subset.
While our sample size is small, these results suggest that the measurement of
particular AKT kinase substrate phospho-proteins could be predictive of MK2206
efficacy in both HER2+ and TN tumors and that immune signaling may play a role in
response in HER2+ patients.
Collapse
|
205
|
Robbe J, Moretta J, Vicier C, Sabatier R, Noguès C, Gonçalves A. Inhibiteurs de PARP dans les cancers du sein : développement clinique actuel et perspectives. Bull Cancer 2020; 107:1024-1041. [DOI: 10.1016/j.bulcan.2020.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/25/2020] [Accepted: 07/05/2020] [Indexed: 12/16/2022]
|
206
|
Zhao Y, Zhang LX, Jiang T, Long J, Ma ZY, Lu AP, Cheng Y, Cao DS. The ups and downs of Poly(ADP-ribose) Polymerase-1 inhibitors in cancer therapy–Current progress and future direction. Eur J Med Chem 2020; 203:112570. [DOI: 10.1016/j.ejmech.2020.112570] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
|
207
|
Poly (ADP-ribose) Polymerase Inhibition in Patients with Breast Cancer and BRCA 1 and 2 Mutations. Drugs 2020; 80:131-146. [PMID: 31823331 DOI: 10.1007/s40265-019-01235-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The poly-(ADP-ribose) polymerase (PARP) inhibitors olaparib and talazoparib, have recently been approved for use in patients with metastatic breast cancer (BC) and germline BRCA 1 or 2 mutations due to improved progression-free survival compared to chemotherapy. An increasing number of clinical trials are evaluating the role of PARP inhibitors (PARPi) in BC, alone and in combination with other therapies (including immunotherapy), as well as in earlier stages of the disease. This review describes the unique mechanism of action of these drugs and puts into clinical context the results of pivotal clinical trials. We also discuss the future development of PARPi in BC, their potential combination with other strategies, including chemotherapy and immune-checkpoint inhibitors, and the impact of these treatments in current genetic counselling.
Collapse
|
208
|
Huynh MM, Pambid MR, Jayanthan A, Dorr A, Los G, Dunn SE. The dawn of targeted therapies for triple negative breast cancer (TNBC): a snapshot of investigational drugs in phase I and II trials. Expert Opin Investig Drugs 2020; 29:1199-1208. [DOI: 10.1080/13543784.2020.1818067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- My-my Huynh
- Pre-clinical R&D, Phoenix Molecular Designs, Vancouver, BC, Canada
| | - Mary Rose Pambid
- Pre-clinical R&D, Phoenix Molecular Designs, Vancouver, BC, Canada
| | - Aarthi Jayanthan
- Pre-clinical R&D, Phoenix Molecular Designs, Vancouver, BC, Canada
| | - Andrew Dorr
- Clinical Operations, Phoenix Molecular Designs, San Diego, CA, USA
| | - Gerrit Los
- Clinical Operations, Phoenix Molecular Designs, San Diego, CA, USA
| | - Sandra E. Dunn
- Pre-clinical R&D, Phoenix Molecular Designs, Vancouver, BC, Canada
- Clinical Operations, Phoenix Molecular Designs, San Diego, CA, USA
| |
Collapse
|
209
|
t'Kint de Roodenbeke MD, Pondé N, Buisseret L, Piccart M. Management of early breast cancer in patients bearing germline BRCA mutations. Semin Oncol 2020; 47:243-248. [PMID: 32912765 DOI: 10.1053/j.seminoncol.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 11/11/2022]
Abstract
Women diagnosed with breast cancers (BCs) that harbor BRCA1/2 mutations have an increased lifetime risk of a second BC and ovarian cancer. They may benefit from risk-reducing surgical strategies such as mastectomy and salpingo-oophorectomy. In cases of triple negative BC with BRCA mutation, there is some evidence that adding platinum-agents in the neoadjuvant setting improves the pathologic complete response. Lastly, ongoing clinical trials testing the efficacy of PARP inhibitor therapy in tumors with BRCA1/2 mutations will be determinant for future guideline recommendations in selecting best adjuvant treatment options for this specific population. For pre-menopausal patients whose tumors have BRCA mutations and hormone-receptor positive BC, the option of combined bilateral annexectomy and hormonal therapy with aromatase inhibitor can be discussed with high-risk patients. This review summarizes the latest results from clinical trials evaluating treatment and prevention strategies for breast cancers harboring BRCA1/2 mutations and discusses the current management of this patient population.
Collapse
Affiliation(s)
| | - Noam Pondé
- Oncology Department AC Camargo Cancer Center, Sao Paulo, Brazil
| | - Laurence Buisseret
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Martine Piccart
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
210
|
Park JJH, Harari O, Dron L, Lester RT, Thorlund K, Mills EJ. An overview of platform trials with a checklist for clinical readers. J Clin Epidemiol 2020; 125:1-8. [PMID: 32416336 DOI: 10.1016/j.jclinepi.2020.04.025] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVES The objective of the study was to outline key considerations for general clinical readers when critically evaluating publications on platform trials and for researchers when designing these types of clinical trials. STUDY DESIGN AND SETTING In this review, we describe key concepts of platform trials with case study discussion of two hallmark platform trials in STAMPEDE and I-SPY2. We provide reader's guide to platform trials with a critical appraisal checklist. RESULTS Platform trials offer flexibilities of dropping ineffective arms early based on interim data and introducing new arms into the trial. For platform trials, it is important to consider how interventions are compared and evaluated throughout and how new interventions are introduced. For intervention comparisons, it is important to consider what the primary analysis is, what and how many interventions are active simultaneously, and allocation between different arms. Interim evaluation considerations should include the number and timing of interim evaluations and outcomes and statistical rules used to drop interventions. New interventions are usually introduced based on scientific merits, so consideration of these merits is important, together with the timing and mechanisms in which new interventions are added. CONCLUSION More efforts are needed to improve the scientific literacy of platform trials. Our review provides an overview of the important concepts of platform trials.
Collapse
Affiliation(s)
- Jay J H Park
- Department of Medicine, Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Cytel, Vancouver, British Columbia, Canada
| | - Ofir Harari
- Cytel, Vancouver, British Columbia, Canada; Department of Health Research Methodology, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Louis Dron
- Cytel, Vancouver, British Columbia, Canada; Department of Health Research Methodology, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Richard T Lester
- Department of Medicine, Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kristian Thorlund
- Cytel, Vancouver, British Columbia, Canada; Department of Health Research Methodology, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada
| | - Edward J Mills
- Cytel, Vancouver, British Columbia, Canada; Department of Health Research Methodology, Evidence, and Impact (HEI), McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
211
|
Yee D, DeMichele AM, Yau C, Isaacs C, Symmans WF, Albain KS, Chen YY, Krings G, Wei S, Harada S, Datnow B, Fadare O, Klein M, Pambuccian S, Chen B, Adamson K, Sams S, Mhawech-Fauceglia P, Magliocco A, Feldman M, Rendi M, Sattar H, Zeck J, Ocal IT, Tawfik O, LeBeau LG, Sahoo S, Vinh T, Chien AJ, Forero-Torres A, Stringer-Reasor E, Wallace AM, Pusztai L, Boughey JC, Ellis ED, Elias AD, Lu J, Lang JE, Han HS, Clark AS, Nanda R, Northfelt DW, Khan QJ, Viscusi RK, Euhus DM, Edmiston KK, Chui SY, Kemmer K, Park JW, Liu MC, Olopade O, Leyland-Jones B, Tripathy D, Moulder SL, Rugo HS, Schwab R, Lo S, Helsten T, Beckwith H, Haugen P, Hylton NM, Van't Veer LJ, Perlmutter J, Melisko ME, Wilson A, Peterson G, Asare AL, Buxton MB, Paoloni M, Clennell JL, Hirst GL, Singhrao R, Steeg K, Matthews JB, Asare SM, Sanil A, Berry SM, Esserman LJ, Berry DA. Association of Event-Free and Distant Recurrence-Free Survival With Individual-Level Pathologic Complete Response in Neoadjuvant Treatment of Stages 2 and 3 Breast Cancer: Three-Year Follow-up Analysis for the I-SPY2 Adaptively Randomized Clinical Trial. JAMA Oncol 2020; 6:1355-1362. [PMID: 32701140 PMCID: PMC7378873 DOI: 10.1001/jamaoncol.2020.2535] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/17/2020] [Indexed: 01/04/2023]
Abstract
Importance Pathologic complete response (pCR) is a known prognostic biomarker for long-term outcomes. The I-SPY2 trial evaluated if the strength of this clinical association persists in the context of a phase 2 neoadjuvant platform trial. Objective To evaluate the association of pCR with event-free survival (EFS) and pCR with distant recurrence-free survival (DRFS) in subpopulations of women with high-risk operable breast cancer treated with standard therapy or one of several novel agents. Design, Setting, and Participants Multicenter platform trial of women with operable clinical stage 2 or 3 breast cancer with no prior surgery or systemic therapy for breast cancer; primary tumors were 2.5 cm or larger. Women with tumors that were ERBB2 negative/hormone receptor (HR) positive with low 70-gene assay score were excluded. Participants were adaptively randomized to one of several different investigational regimens or control therapy within molecular subtypes from March 2010 through 2016. The analysis included participants with follow-up data available as of February 26, 2019. Interventions Standard-of-care neoadjuvant therapy consisting of taxane treatment with or without (as control) one of several investigational agents or combinations followed by doxorubicin and cyclophosphamide. Main Outcomes and Measures Pathologic complete response and 3-year EFS and DRFS. Results Of the 950 participants (median [range] age, 49 [23-77] years), 330 (34.7%) achieved pCR. Three-year EFS and DRFS for patients who achieved pCR were both 95%. Hazard ratios for pCR vs non-pCR were 0.19 for EFS (95% CI, 0.12-0.31) and 0.21 for DRFS (95% CI, 0.13-0.34) and were similar across molecular subtypes, varying from 0.14 to 0.18 for EFS and 0.10 to 0.20 for DRFS. Conclusions and Relevance The 3-year outcomes from the I-SPY2 trial show that, regardless of subtype and/or treatment regimen, including 9 novel therapeutic combinations, achieving pCR after neoadjuvant therapy implies approximately an 80% reduction in recurrence rate. The goal of the I-SPY2 trial is to rapidly identify investigational therapies that may improve pCR when validated in a phase 3 confirmatory trial. Whether pCR is a validated surrogate in the sense that a therapy that improves pCR rate can be assumed to also improve long-term outcome requires further study. Trial Registration ClinicalTrials.gov Identifier: NCT01042379.
Collapse
Affiliation(s)
- Douglas Yee
- Masonic Cancer Center, University of Minnesota, Minneapolis
| | | | - Christina Yau
- Department of Surgery, University of California, San Francisco
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Kathy S Albain
- Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco
| | - Gregor Krings
- Department of Pathology, University of California, San Francisco
| | - Shi Wei
- Department of Pathology, University of Alabama Birmingham
| | - Shuko Harada
- Department of Pathology, University of Alabama Birmingham
| | - Brian Datnow
- Department of Pathology, University of California, San Diego
| | - Oluwole Fadare
- Department of Pathology, University of California, San Diego
| | - Molly Klein
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Stefan Pambuccian
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Beiyun Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Kathi Adamson
- Department of Pathology, Swedish Cancer Institute, Seattle, Washington
| | - Sharon Sams
- Department of Pathology, University of Colorado, Denver
| | | | | | - Mike Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia
| | - Mara Rendi
- Department of Anatomic Pathology, University of Washington, Seattle
| | - Husain Sattar
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Jay Zeck
- Department of Pathology, Georgetown University, Washington, DC
| | - Idris T Ocal
- Laboratory Medicine and Pathology, Mayo Clinic Scottsdale, Scottsdale, Arizona
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas, Lawrence
| | | | - Sunati Sahoo
- Department of Pathology, University of Texas Southwestern, Dallas
| | - Tuyethoa Vinh
- Inova Pathology Institute, Inova Health System, Falls Church, Virginia
| | - A Jo Chien
- Division of Hematology and Oncology, University of California, San Francisco
| | | | | | - Anne M Wallace
- Department of Surgery, University of California, San Diego
| | - Lajos Pusztai
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic Rochester, Rochester, Minnesota
| | - Erin D Ellis
- Medical Oncology, Swedish Cancer Institute, Seattle, Washington
| | | | - Janice Lu
- Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles
| | - Julie E Lang
- Surgery, Keck School of Medicine, University of Southern California, Los Angeles
| | - Hyo S Han
- Medical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Amy S Clark
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Rita Nanda
- Hematology and Oncology, University of Chicago Medical Center, Chicago, Illinois
| | | | - Qamar J Khan
- Medical Oncology, University of Kansas Medical Center, Lawrence
| | | | - David M Euhus
- Department of Surgery, Johns Hopkins Medicine, Baltimore, Maryland
| | | | | | - Kathleen Kemmer
- Knight Cancer Institute, Oregon Health & Science University, Portland
| | - John W Park
- Division of Hematology and Oncology, University of California, San Francisco
| | - Minetta C Liu
- Department of Oncology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Olufunmilayo Olopade
- Hematology and Oncology, University of Chicago Medical Center, Chicago, Illinois
| | | | - Debasish Tripathy
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Stacy L Moulder
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Hope S Rugo
- Division of Hematology and Oncology, University of California, San Francisco
| | | | - Shelly Lo
- Medical Oncology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | | | | | | | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Laura J Van't Veer
- Department of Laboratory Medicine, University of California, San Francisco
| | | | - Michelle E Melisko
- Division of Hematology and Oncology, University of California, San Francisco
| | - Amy Wilson
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | - Garry Peterson
- Department of Surgery, University of California, San Francisco
| | - Adam L Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | | | | | - Gillian L Hirst
- Department of Surgery, University of California, San Francisco
| | - Ruby Singhrao
- Department of Surgery, University of California, San Francisco
| | - Katherine Steeg
- Department of Surgery, University of California, San Francisco
| | | | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | | | | | | |
Collapse
|
212
|
Zhang J, Yao L, Liu Y, Ouyang T, Li J, Wang T, Fan Z, Fan T, Lin B, Xie Y. Impact of the addition of carboplatin to anthracycline-taxane-based neoadjuvant chemotherapy on survival in BRCA1/2-mutated triple-negative breast cancer. Int J Cancer 2020; 148:941-949. [PMID: 32720318 DOI: 10.1002/ijc.33234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/25/2020] [Accepted: 07/17/2020] [Indexed: 11/11/2022]
Abstract
Whether adding carboplatin to standard neoadjuvant chemotherapy improves survival in BRCA1/2-mutated triple-negative breast cancer (TNBC) is unknown. In this retrospective study, we aimed to explore the efficacy of anthracycline-taxane (A-T)-based or anthracycline-taxane/carboplatin (A-TP)-based neoadjuvant chemotherapy in BRCA1/2-mutated TNBC. A total of 1585 operable primary breast cancer patients were treated with either neoadjuvant A-T (n = 886) or A-TP regimen (n = 699). BRCA1 and BRCA2 germline mutations were determined in all subjects. Pathological complete response (pCR), recurrence-free survival (RFS), distant recurrence-free survival (DRFS) and overall survival (OS) were estimated. Of the entire cohort, 102 patients (6.4%) carried a pathogenic BRCA1/2 germline mutation. After a median follow-up of 81 months, no significant differences in survival between the A-T and A-TP arms were found in the entire cohort. However, among 288 TNBC patients, BRCA1/2 mutation carriers had significantly better survival when treated with the A-TP regimen than with the A-T regimen (5-year RFS: 82.6% vs 47.9%; P = .024; 5-year DRFS: 88.5% vs 46.9%; P = .010; 5-year OS: 88.2% vs 49.9%; P = .036). Multivariate analyses revealed that the A-TP regimen was a significantly favourable factor for RFS and DRFS and showed a trend towards better OS when compared with the A-T regimen in BRCA1/2-mutated TNBC (RFS: adjusted hazard ratio [HR], 0.24; 95% confidence interval [CI], 0.06-0.91, P = .035; DRFS: HR, 0.17; 95% CI, 0.03-0.80; P = .025; OS: HR, 0.29; 95% CI, 0.06-1.49; P = .14). Our study suggested that BRCA1/2-mutated TNBC patients gain a survival benefit when carboplatin is added to standard A-T-based neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Juan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Yao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yiqiang Liu
- Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Tao Ouyang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jinfeng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Tianfeng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhaoqing Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Tie Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Benyao Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuntao Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
213
|
Wu SY, Wang H, Shao ZM, Jiang YZ. Triple-negative breast cancer: new treatment strategies in the era of precision medicine. SCIENCE CHINA-LIFE SCIENCES 2020; 64:372-388. [PMID: 32803712 DOI: 10.1007/s11427-020-1714-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancer (TNBC) remains the most aggressive cluster of all breast cancers, which is due to its rapid progression, high probabilities of early recurrence, and distant metastasis resistant to standard treatment. Following the advances in cancer genomics and transcriptomics that can illustrate the comprehensive profiling of this heterogeneous disease, it is now possible to identify different subclasses of TNBC according to both intrinsic signals and extrinsic microenvironment, which have a huge influence on predicting response to established therapies and picking up novel therapeutic targets for each cluster. In this review, we summarize basic characteristics and critical subtyping systems of TNBC, and particularly discuss newly found prospective targets and relevant medications, which were proved promising in clinical trials, thus shedding light on the future development of precision treatment strategies.
Collapse
Affiliation(s)
- Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hai Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
214
|
Shaikh SS, Emens LA. Current and emerging biologic therapies for triple negative breast cancer. Expert Opin Biol Ther 2020; 22:591-602. [PMID: 32713217 DOI: 10.1080/14712598.2020.1801627] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Triple negative breast cancer, defined by a lack of estrogen receptor, progesterone receptor, or human epidermal growth factor2, accounts for approximately 15% of breast cancer patients. Treatment options have historically been limited to chemotherapy, which has significant toxicity and a suboptimal impact on the five-year relapse rate and survival. AREAS COVERED Transcriptomic analyses reveal that TNBC is biologically heterogenous. Predictive biomarkers based on the distinct biology of the different subtypes of TNBC should identify patients that will derive the greatest benefit from a specifically targeted therapeutic agent. Two biomarker-driven treatments have recently been approved: poly-ADP ribose polymerase inhibitors for patients with germline BRCA mutations and atezolizumab in combination with nab-paclitaxel for patients expressing PD-L1 on tumor-infiltrating immune cells. EXPERT OPINION Identifying informative predictive biomarkers is critical for the optimal development of targeted drugs for TNBC. Some targeted agents, such as the antibody-drug conjugate sacituzumab govitecan-hziy and the precision medicines capivasertib and ipatisertib, have already shown promising results in early clinical trials, and the results of definitive phase 3 trials are eagerly awaited. Additionally, testing novel immunotherapies and other targeted agents in earlier stages of disease, particularly the neoadjuvant setting, is a high priority.
Collapse
Affiliation(s)
- Saba S Shaikh
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leisha A Emens
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
215
|
Arasu VA, Kim P, Li W, Strand F, McHargue C, Harnish R, Newitt DC, Jones EF, Glymour MM, Kornak J, Esserman LJ, Hylton NM, ISPY2 investigators. Predictive Value of Breast MRI Background Parenchymal Enhancement for Neoadjuvant Treatment Response among HER2- Patients. JOURNAL OF BREAST IMAGING 2020; 2:352-360. [PMID: 32803155 PMCID: PMC7418876 DOI: 10.1093/jbi/wbaa028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Women with advanced HER2- breast cancer have limited treatment options. Breast MRI functional tumor volume (FTV) is used to predict pathologic complete response (pCR) to improve treatment efficacy. In addition to FTV, background parenchymal enhancement (BPE) may predict response and was explored for HER2- patients in the I-SPY-2 TRIAL. METHODS Women with HER2- stage II or III breast cancer underwent prospective serial breast MRIs during four neoadjuvant chemotherapy timepoints. BPE was quantitatively calculated using whole-breast manual segmentation. Logistic regression models were systematically explored using pre-specified and optimized predictor selection based on BPE or combined with FTV. RESULTS A total of 352 MRI examinations in 88 patients (29 with pCR, 59 non-pCR) were evaluated. Women with hormone receptor (HR)+HER2- cancers who achieved pCR demonstrated a significantly greater decrease in BPE from baseline to pre-surgery compared to non-pCR patients (odds ratio 0.64, 95% confidence interval (CI): 0.39-0.92, P = 0.04). The associated BPE area under the curve (AUC) was 0.77 (95% CI: 0.56-0.98), comparable to the range of FTV AUC estimates. Among multi-predictor models, the highest cross-validated AUC of 0.81 (95% CI: 0.73-0.90) was achieved with combined FTV+HR predictors, while adding BPE to FTV+HR models had an estimated AUC of 0.82 (95% CI: 0.74-0.92). CONCLUSION Among women with HER2- cancer, BPE alone demonstrated association with pCR in women with HR+HER2- breast cancer, with similar diagnostic performance to FTV. BPE predictors remained significant in multivariate FTV models, but without added discrimination for pCR prediction. This may be due to small sample size limiting ability to create subtype-specific multivariate models.
Collapse
Affiliation(s)
- Vignesh A Arasu
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
- University of California San Francisco, Department of Epidemiology and Biostatistics, San Francisco, CA
- Kaiser Permanente Medical Center, Department of Radiology, Vallejo, CA
- Kaiser Permanente Northern California, Oakland, CA
| | - Paul Kim
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
| | - Wen Li
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
| | - Fredrik Strand
- Karolinska University Hospital, Breast Radiology, Stockholm, Sweden
| | - Cody McHargue
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
| | - Roy Harnish
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
| | - David C Newitt
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
| | - Ella F Jones
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
| | - M Maria Glymour
- University of California San Francisco, Department of Epidemiology and Biostatistics, San Francisco, CA
| | - John Kornak
- University of California San Francisco, Department of Epidemiology and Biostatistics, San Francisco, CA
| | - Laura J Esserman
- University of California San Francisco, Department of Surgery, San Francisco, CA
| | - Nola M Hylton
- University of California San Francisco, Department of Radiology and Biomedical Imaging, San Francisco, CA
| | | |
Collapse
|
216
|
D’Amico F, Danese S, Peyrin-Biroulet L. Adaptive Designs: Lessons for Inflammatory Bowel Disease Trials. J Clin Med 2020; 9:jcm9082350. [PMID: 32717997 PMCID: PMC7464489 DOI: 10.3390/jcm9082350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/10/2020] [Accepted: 07/22/2020] [Indexed: 11/16/2022] Open
Abstract
In recent decades, scientific research has considerably evolved in the field of inflammatory bowel diseases (IBD) and clinical studies have become increasingly complex, including new outcomes, different study populations, and additional techniques of re-randomization and centralized control. In this context, randomized clinical trials are the gold standard for new drugs’ development. However, traditional study designs are time-consuming, expensive, and only a small percentage of the tested therapies are approved. For this reason, a new study design called “adaptive design” has been introduced, allowing to accumulate data during the study and to make predefined adjustments based on the results of scheduled interim analysis. Our aim is to clarify the advantages and drawbacks of adaptive designs in order to properly interpret study results and to identify their role in upcoming IBD trials.
Collapse
Affiliation(s)
- Ferdinando D’Amico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (F.D.); (S.D.)
- Department of Gastroenterology and Inserm NGERE U1256, Nancy University Hospital, University of Lorraine, 54500 Vandoeuvre-lès-Nancy, France
| | - Silvio Danese
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (F.D.); (S.D.)
- Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology and Inserm NGERE U1256, Nancy University Hospital, University of Lorraine, 54500 Vandoeuvre-lès-Nancy, France
- Correspondence: ; Tel.: +33-3831-53661
| |
Collapse
|
217
|
Abstract
PARP (poly(ADP-ribose) polymerase) inhibitors represent a novel class of anti-cancer therapy; they take advantage of synthetic lethality and induce cell death by exploiting a defect in DNA repair. This class of medication was initially evaluated in patients with BRCA-associated tumors, but efficacy was also demonstrated in other populations. Since 2014, four PARP inhibitors have been approved in various indications: olaparib, niraparib, and rucaparib in high-grade serous ovarian cancer, and olaparib and talazoparib in metastatic breast cancer. The exact indications and study populations vary slightly between the different approvals in both disease states but there is significant overlap. PARP inhibitors continue to be investigated in ongoing clinical trials. In line with other targeted therapies, benefit appears to be strongest in a distinct population of patients with BRCA mutations or other defects in homologous recombination repair. Combination therapies, which include anti-angiogenesis agents and immunotherapy, show promise as a strategy to broaden efficacy for unselected patients. Initial studies of PARP inhibitors in combination with chemotherapy were limited by toxicity, but further studies are underway. To date, head-to-head trials comparing various PARP inhibitors have not been conducted, so questions remain in terms of choosing a PARP inhibitor to administer when indications overlap, as well as how to sequence these medications. Here we review both completed and ongoing clinical trials involving PARP inhibitors and mechanisms of resistance to this class of drugs.
Collapse
|
218
|
Resistance to Neoadjuvant Treatment in Breast Cancer: Clinicopathological and Molecular Predictors. Cancers (Basel) 2020; 12:cancers12082012. [PMID: 32708049 PMCID: PMC7463925 DOI: 10.3390/cancers12082012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/03/2020] [Accepted: 07/20/2020] [Indexed: 01/30/2023] Open
Abstract
Neoadjuvant Chemotherapy (NAC) in Breast Cancer (BC) has proved useful for the reduction in tumor burden prior to surgery, allowing for a more extensive breast preservation and the eradication of subjacent micrometastases. However, the impact on prognosis is highly dependent on the establishment of Pathological Complete Response (pCR), in particular for Triple Negative (TN) and Hormonal Receptor negative/Human Epidermal growth factor Receptor 2 positive (HR-/HER2+) subtypes. Several pCR predictors, such as PAM50, Integrative Cluster (IntClust), mutations in PI3KCA, or the Trastuzumab Risk model (TRAR), are useful molecular tools for estimating response to treatment and are prognostic. Major evolution events during BC NAC that feature the Residual Disease (RD) are the loss of HR and HER2, which are prognostic of bad outcome, and stemness and immune depletion-related gene expression aberrations. This dynamic nature of the determinants of response to BC NAC, together with the extensive heterogeneity of BC, raises the need to discern the individual and subtype-specific determinants of resistance. Moreover, refining the current approaches for a comprehensive monitoring of tumor evolution during treatment, RD, and eventual recurrences is essential for identifying new actionable alterations and the integral best management of the disease.
Collapse
|
219
|
Meyer EL, Mesenbrink P, Dunger-Baldauf C, Fülle HJ, Glimm E, Li Y, Posch M, König F. The Evolution of Master Protocol Clinical Trial Designs: A Systematic Literature Review. Clin Ther 2020; 42:1330-1360. [PMID: 32622783 DOI: 10.1016/j.clinthera.2020.05.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/10/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Recent years have seen a change in the way that clinical trials are being conducted. There has been a rise of designs more flexible than traditional adaptive and group sequential trials which allow the investigation of multiple substudies with possibly different objectives, interventions, and subgroups conducted within an overall trial structure, summarized by the term master protocol. This review aims to identify existing master protocol studies and summarize their characteristics. The review also identifies articles relevant to the design of master protocol trials, such as proposed trial designs and related methods. METHODS We conducted a comprehensive systematic search to review current literature on master protocol trials from a design and analysis perspective, focusing on platform trials and considering basket and umbrella trials. Articles were included regardless of statistical complexity and classified as reviews related to planned or conducted trials, trial designs, or statistical methods. The results of the literature search are reported, and some features of the identified articles are summarized. FINDINGS Most of the trials using master protocols were designed as single-arm (n = 29/50), Phase II trials (n = 32/50) in oncology (n = 42/50) using a binary endpoint (n = 26/50) and frequentist decision rules (n = 37/50). We observed an exponential increase in publications in this domain during the last few years in both planned and conducted trials, as well as relevant methods, which we assume has not yet reached its peak. Although many operational and statistical challenges associated with such trials remain, the general consensus seems to be that master protocols provide potentially enormous advantages in efficiency and flexibility of clinical drug development. IMPLICATIONS Master protocol trials and especially platform trials have the potential to revolutionize clinical drug development if the methodologic and operational challenges can be overcome.
Collapse
Affiliation(s)
- Elias Laurin Meyer
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | - Yuhan Li
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Martin Posch
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Franz König
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
220
|
Angus DC, Berry S, Lewis RJ, Al-Beidh F, Arabi Y, van Bentum-Puijk W, Bhimani Z, Bonten M, Broglio K, Brunkhorst F, Cheng AC, Chiche JD, De Jong M, Detry M, Goossens H, Gordon A, Green C, Higgins AM, Hullegie SJ, Kruger P, Lamontagne F, Litton E, Marshall J, McGlothlin A, McGuinness S, Mouncey P, Murthy S, Nichol A, O’Neill GK, Parke R, Parker J, Rohde G, Rowan K, Turner A, Young P, Derde L, McArthur C, Webb SA. The REMAP-CAP (Randomized Embedded Multifactorial Adaptive Platform for Community-acquired Pneumonia) Study. Rationale and Design. Ann Am Thorac Soc 2020; 17:879-891. [PMID: 32267771 PMCID: PMC7328186 DOI: 10.1513/annalsats.202003-192sd] [Citation(s) in RCA: 275] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/08/2020] [Indexed: 12/22/2022] Open
Abstract
There is broad interest in improved methods to generate robust evidence regarding best practice, especially in settings where patient conditions are heterogenous and require multiple concomitant therapies. Here, we present the rationale and design of a large, international trial that combines features of adaptive platform trials with pragmatic point-of-care trials to determine best treatment strategies for patients admitted to an intensive care unit with severe community-acquired pneumonia. The trial uses a novel design, entitled "a randomized embedded multifactorial adaptive platform." The design has five key features: 1) randomization, allowing robust causal inference; 2) embedding of study procedures into routine care processes, facilitating enrollment, trial efficiency, and generalizability; 3) a multifactorial statistical model comparing multiple interventions across multiple patient subgroups; 4) response-adaptive randomization with preferential assignment to those interventions that appear most favorable; and 5) a platform structured to permit continuous, potentially perpetual enrollment beyond the evaluation of the initial treatments. The trial randomizes patients to multiple interventions within four treatment domains: antibiotics, antiviral therapy for influenza, host immunomodulation with extended macrolide therapy, and alternative corticosteroid regimens, representing 240 treatment regimens. The trial generates estimates of superiority, inferiority, and equivalence between regimens on the primary outcome of 90-day mortality, stratified by presence or absence of concomitant shock and proven or suspected influenza infection. The trial will also compare ventilatory and oxygenation strategies, and has capacity to address additional questions rapidly during pandemic respiratory infections. As of January 2020, REMAP-CAP (Randomized Embedded Multifactorial Adaptive Platform for Community-acquired Pneumonia) was approved and enrolling patients in 52 intensive care units in 13 countries on 3 continents. In February, it transitioned into pandemic mode with several design adaptations for coronavirus disease 2019. Lessons learned from the design and conduct of this trial should aid in dissemination of similar platform initiatives in other disease areas.Clinical trial registered with www.clinicaltrials.gov (NCT02735707).
Collapse
Affiliation(s)
- Derek C. Angus
- The Clinical Research Investigation and Systems Modeling of Acute Illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Roger J. Lewis
- Berry Consultants, LLC, Austin, Texas
- Department of Emergency Medicine, Harbor–University of California Los Angeles (UCLA) Medical Center, Torrance, California
- Department of Emergency Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Farah Al-Beidh
- Division of Anaesthetics, Pain Medicine and Intensive Care Medicine, Department of Surgery and Cancer, Imperial College London and Imperial College Healthcare National Health Service Trust, London, United Kingdom
| | - Yaseen Arabi
- Intensive Care Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | | | - Zahra Bhimani
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Marc Bonten
- Julius Center for Health Sciences and Primary Care
- Department of Medical Microbiology, and
| | | | - Frank Brunkhorst
- Center for Clinical Studies and Center for Sepsis Control and Care, Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Allen C. Cheng
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, Melbourne, Victoria, Australia
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jean-Daniel Chiche
- Medical Intensive Care Unit, Hôpital Cochin, Paris Descartes University, Paris, France
| | - Menno De Jong
- Department of Medical Microbiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Herman Goossens
- Department of Microbiology, Antwerp University Hospital, Antwerp, Belgium
| | - Anthony Gordon
- Division of Anaesthetics, Pain Medicine and Intensive Care Medicine, Department of Surgery and Cancer, Imperial College London and Imperial College Healthcare National Health Service Trust, London, United Kingdom
| | - Cameron Green
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Alisa M. Higgins
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | | | - Peter Kruger
- Intensive Care Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | - Edward Litton
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - John Marshall
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | | | - Shay McGuinness
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Cardiothoracic and Vascular Intensive Care Unit and
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Paul Mouncey
- Clinical Trials Unit, Intensive Care National Audit & Research Centre, London, United Kingdom
| | - Srinivas Murthy
- University of British Columbia School of Medicine, Vancouver, British Columbia, Canada
| | - Alistair Nichol
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Anesthesia and Intensive Care, St Vincent’s University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Genevieve K. O’Neill
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rachael Parke
- Cardiothoracic and Vascular Intensive Care Unit and
- Medical Research Institute of New Zealand, Wellington, New Zealand
- School of Nursing, University of Auckland, Auckland, New Zealand
| | - Jane Parker
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Gernot Rohde
- Department of Respiratory Medicine, University Hospital Frankfurt, Frankfurt, Germany
- CAPNETZ Foundation, Hannover, Germany
| | - Kathryn Rowan
- Clinical Trials Unit, Intensive Care National Audit & Research Centre, London, United Kingdom
| | - Anne Turner
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Paul Young
- Medical Research Institute of New Zealand, Wellington, New Zealand
- Intensive Care Unit, Wellington Hospital, Wellington, New Zealand; and
| | - Lennie Derde
- Julius Center for Health Sciences and Primary Care
- Intensive Care Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Colin McArthur
- Department of Critical Care Medicine, Auckland City Hospital, Auckland, New Zealand
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Steven A. Webb
- Australian and New Zealand Intensive Care Research Centre, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
- St. John of God Hospital, Subiaco, Western Australia, Australia
| |
Collapse
|
221
|
Onishi N, Li W, Gibbs J, Wilmes LJ, Nguyen A, Jones EF, Arasu V, Kornak J, Joe BN, Esserman LJ, Newitt DC, Hylton NM. Impact of MRI Protocol Adherence on Prediction of Pathological Complete Response in the I-SPY 2 Neoadjuvant Breast Cancer Trial. Tomography 2020; 6:77-85. [PMID: 32548283 PMCID: PMC7289255 DOI: 10.18383/j.tom.2020.00006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We investigated the impact of magnetic resonance imaging (MRI) protocol adherence on the ability of functional tumor volume (FTV), a quantitative measure of tumor burden measured from dynamic contrast-enhanced MRI, to predict response to neoadjuvant chemotherapy. We retrospectively reviewed dynamic contrast-enhanced breast MRIs for 990 patients enrolled in the multicenter I-SPY 2 TRIAL. During neoadjuvant chemotherapy, each patient had 4 MRI visits (pretreatment [T0], early-treatment [T1], inter-regimen [T2], and presurgery [T3]). Protocol adherence was rated for 7 image quality factors at T0-T2. Image quality factors confirmed by DICOM header (acquisition duration, early phase timing, field of view, and spatial resolution) were adherent if the scan parameters followed the standardized imaging protocol, and changes from T0 for a single patient's visits were limited to defined ranges. Other image quality factors (contralateral image quality, patient motion, and contrast administration error) were considered adherent if imaging issues were absent or minimal. The area under the receiver operating characteristic curve (AUC) was used to measure the performance of FTV change (percent change of FTV from T0 to T1 and T2) in predicting pathological complete response. FTV changes with adherent image quality in all factors had higher estimated AUC than those with non-adherent image quality, although the differences did not reach statistical significance (T1, 0.71 vs. 0.66; T2, 0.72 vs. 0.68). These data highlight the importance of MRI protocol adherence to predefined scan parameters and the impact of data quality on the predictive performance of FTV in the breast cancer neoadjuvant setting.
Collapse
Affiliation(s)
| | - Wen Li
- Departments of Radiology & Biomedical Imaging
| | | | | | - Alex Nguyen
- Departments of Radiology & Biomedical Imaging
| | | | | | | | | | - Laura J Esserman
- Surgery, University of California, San Francisco, San Francisco, CA
| | | | | |
Collapse
|
222
|
Rational Cancer Treatment Combinations: An Urgent Clinical Need. Mol Cell 2020; 78:1002-1018. [DOI: 10.1016/j.molcel.2020.05.031] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
|
223
|
Furlanetto J, Loibl S. Optimal Systemic Treatment for Early Triple-Negative Breast Cancer. Breast Care (Basel) 2020; 15:217-226. [PMID: 32774215 PMCID: PMC7383279 DOI: 10.1159/000508759] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Approximately 10-15% of all breast tumors are triple-negative breast cancer (TNBC). TNBC have a higher risk of relapse and distant metastases compared to other subtypes. The optimal systemic management of TNBC according to national and international guidelines is discussed herein. SUMMARY Anthracycline/taxane-based chemotherapy for patients with TNBC either in the neoadjuvant (NACT) or the adjuvant setting is considered standard of care. Exceptions are small tumors and a low-risk histology, in which chemotherapy can be spared. Dose-dense therapy is more effective in preventing recurrence and increasing survival. The use of nab-paclitaxel instead of a solvent-based taxane can lead to higher pathological complete response (pCR) rates and better outcomes. Platinum agents are effective in increasing pCR when added to anthracycline/taxane-based chemotherapy at the cost of increased toxicity. Long-term outcome data are lacking. In patients without a pCR, capecitabine leads to improved outcomes. KEY MESSAGES The standard treatment approach of TNBC is anthracycline/taxane-based chemotherapy, preferably within the NACT setting. Dose-dense schedules as well as platinum should be considered in the NACT setting. For patients without a pCR, capecitabine is an option to improve the outcome. The role of nab-paclitaxel is under debate. In case of immunogenic tumors, checkpoint inhibitors are promising new agents that merit further investigation.
Collapse
|
224
|
Le Du F, Brunot A, de la Motte Rouge T, Diéras V. Implications of BRCA Germline Mutations on Breast Cancer Medical Treatment. CURRENT BREAST CANCER REPORTS 2020. [DOI: 10.1007/s12609-020-00362-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
225
|
Tsimberidou AM, Fountzilas E, Nikanjam M, Kurzrock R. Review of precision cancer medicine: Evolution of the treatment paradigm. Cancer Treat Rev 2020; 86:102019. [PMID: 32251926 PMCID: PMC7272286 DOI: 10.1016/j.ctrv.2020.102019] [Citation(s) in RCA: 399] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022]
Abstract
In recent years, biotechnological breakthroughs have led to identification of complex and unique biologic features associated with carcinogenesis. Tumor and cell-free DNA profiling, immune markers, and proteomic and RNA analyses are used to identify these characteristics for optimization of anticancer therapy in individual patients. Consequently, clinical trials have evolved, shifting from tumor type-centered to gene-directed, histology-agnostic, with innovative adaptive design tailored to biomarker profiling with the goal to improve treatment outcomes. A plethora of precision medicine trials have been conducted. The majority of these trials demonstrated that matched therapy is associated with superior outcomes compared to non-matched therapy across tumor types and in specific cancers. To improve the implementation of precision medicine, this approach should be used early in the course of the disease, and patients should have complete tumor profiling and access to effective matched therapy. To overcome the complexity of tumor biology, clinical trials with combinations of gene-targeted therapy with immune-targeted approaches (e.g., checkpoint blockade, personalized vaccines and/or chimeric antigen receptor T-cells), hormonal therapy, chemotherapy and/or novel agents should be considered. These studies should target dynamic changes in tumor biologic abnormalities, eliminating minimal residual disease, and eradicating significant subclones that confer resistance to treatment. Mining and expansion of real-world data, facilitated by the use of advanced computer data processing capabilities, may contribute to validation of information to predict new applications for medicines. In this review, we summarize the clinical trials and discuss challenges and opportunities to accelerate the implementation of precision oncology.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX.
| | - Elena Fountzilas
- Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece
| | - Mina Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| |
Collapse
|
226
|
Lee JS, Yost SE, Yuan Y. Neoadjuvant Treatment for Triple Negative Breast Cancer: Recent Progresses and Challenges. Cancers (Basel) 2020; 12:E1404. [PMID: 32486021 PMCID: PMC7352772 DOI: 10.3390/cancers12061404] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive breast cancer with historically poor outcomes, primarily due to the lack of effective targeted therapies. The tumor molecular heterogeneity of TNBC has been well recognized, yet molecular subtype driven therapy remains lacking. While neoadjuvant anthracycline and taxane-based chemotherapy remains the standard of care for early stage TNBC, the optimal chemotherapy regimen is debatable. The addition of carboplatin to anthracycline, cyclophosphamide, and taxane (ACT) regimen is associated with improved complete pathologic response (pCR). Immune checkpoint inhibitor (ICI) combinations significantly increase pCR in TNBC. Increased tumor infiltrating lymphocyte (TILs) or the presence of DNA repair deficiency (DRD) mutation is associated with increased pCR. Other targets, such as poly-ADP-ribosyl polymerase inhibitors (PARPi) and Phosphatidylinositol-3-kinase/Protein Kinase B/mammalian target of rapamycin (PI3K-AKT-mTOR) pathway inhibitors, are being evaluated in the neoadjuvant setting. This review examines recent progress in neoadjuvant therapy of TNBC, including platinum, ICI, PARPi, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) pathway targeted therapies, and novel tumor microenvironment (TME) targeted therapy, in addition to biomarkers for the prediction of pCR.
Collapse
Affiliation(s)
| | | | - Yuan Yuan
- Department of Medical Oncology & Molecular Therapeutics, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA 91010, USA; (J.S.L.); (S.E.Y.)
| |
Collapse
|
227
|
Gonçalves A, Bertucci A, Bertucci F. PARP Inhibitors in the Treatment of Early Breast Cancer: The Step Beyond? Cancers (Basel) 2020; 12:cancers12061378. [PMID: 32471249 PMCID: PMC7352970 DOI: 10.3390/cancers12061378] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
Exquisitely exploiting defects in homologous recombination process, poly(ADP-ribose) polymerase (PARP) inhibitors have recently emerged as a promising class of therapeutics in human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer with germline breast cancer 1 (BRCA1) or breast cancer 2 (BRCA2) mutations (gBRCA1/2m). In this setting, PARP inhibitors, either as single agent or in combination with platinum-based chemotherapy, significantly increased progression-free survival, as compared to conventional chemotherapy. Accordingly, further therapeutic advances are expected at an earlier stage of the disease. In the neoadjuvant setting, veliparib failed to increase the pathological complete response rate when added to a carboplatin-based regimen, in unselected triple-negative breast cancer patients. Similarly, when administered before anthracycline-cyclophosphamide, the neoadjuvant olaparib-paclitaxel combination was not superior to carboplatin–paclitaxel, in patients with HER2-negative breast cancer and BRCA1/2 mutation, or homologous recombination defect. Yet, neoadjuvant talazoparib, administered as a single-agent in patients with HER2-negative breast cancer and germline BRCA1/2 mutation, achieved an impressive pathological complete response rate of nearly 50%. In the adjuvant setting, the results from the OlympiA phase III study, evaluating adjuvant olaparib in HER2-negative early breast cancer and germline BRCA1/2 mutations, are eagerly awaited. Ongoing trials should clarify whether PARP inhibitors might improve outcome when administered in the adjuvant or neoadjuvant setting in early breast cancer patients with BRCA1/2 mutation or homologous recombination defect.
Collapse
Affiliation(s)
- Anthony Gonçalves
- Medical Oncology Department, Cancer Research Center of Marseille (CRCM), INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France; (A.B.); (F.B.)
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France
- Correspondence: ; Tel.: +33-4-91-22-37-89; Fax: +33-4-91-22-36-70
| | - Alexandre Bertucci
- Medical Oncology Department, Cancer Research Center of Marseille (CRCM), INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France; (A.B.); (F.B.)
| | - François Bertucci
- Medical Oncology Department, Cancer Research Center of Marseille (CRCM), INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France; (A.B.); (F.B.)
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France
| |
Collapse
|
228
|
Chaudhary LN. Early stage triple negative breast cancer: Management and future directions. Semin Oncol 2020; 47:201-208. [PMID: 32507668 DOI: 10.1053/j.seminoncol.2020.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 12/31/2022]
Abstract
Triple negative breast cancer is the most aggressive kind of breast cancer with high risk of recurrences and poor outcomes. Systemic chemotherapy has significantly improved long term outcomes in early stage patients; however, metastatic recurrences still develop in a significant number of patients. Anthracycline and taxane based chemotherapy regimens are standard of care for early stage patients. Neoadjuvant treatment is preferred due to the ability to assess pathologic responses providing important prognostic information and guidance in adjuvant therapy decisions. Carboplatin addition to the anthracycline and taxane backbone is associated with a significant improvement in pathologic complete response but is associated with more toxicity. Understanding the immune microenvironment of triple negative disease is an exciting field and immune checkpoint inhibitors have shown great promise in further improving response rates in early stage patients. Patients with residual disease after neoadjuvant chemotherapy have a significantly higher risk of recurrence compared to those with complete responses. Adjuvant capecitabine for these high-risk patients have shown significant improvement in long term outcomes and is routinely used in this setting. Given the heterogeneity within triple negative tumors, molecular subtypes with variable genomic makeup and chemo sensitivities have been identified and will likely aid in further clinical developmental therapeutics.
Collapse
Affiliation(s)
- Lubna N Chaudhary
- Division of Hematology/Oncology, Froedtert and Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
229
|
Smith BD, Goetz MP, Boughey JC. Multidisciplinary Management of Breast Cancer With Extensive Regional Nodal Involvement. J Clin Oncol 2020; 38:2290-2298. [PMID: 32442073 DOI: 10.1200/jco.19.03036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice.
Collapse
Affiliation(s)
| | - Matthew P Goetz
- Department of Oncology, Mayo Clinic, Rochester, MN.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
230
|
Nanda R, Liu MC, Yau C, Shatsky R, Pusztai L, Wallace A, Chien AJ, Forero-Torres A, Ellis E, Han H, Clark A, Albain K, Boughey JC, Jaskowiak NT, Elias A, Isaacs C, Kemmer K, Helsten T, Majure M, Stringer-Reasor E, Parker C, Lee MC, Haddad T, Cohen RN, Asare S, Wilson A, Hirst GL, Singhrao R, Steeg K, Asare A, Matthews JB, Berry S, Sanil A, Schwab R, Symmans WF, van ‘t Veer L, Yee D, DeMichele A, Hylton NM, Melisko M, Perlmutter J, Rugo HS, Berry DA, Esserman LJ. Effect of Pembrolizumab Plus Neoadjuvant Chemotherapy on Pathologic Complete Response in Women With Early-Stage Breast Cancer: An Analysis of the Ongoing Phase 2 Adaptively Randomized I-SPY2 Trial. JAMA Oncol 2020; 6:676-684. [PMID: 32053137 PMCID: PMC7058271 DOI: 10.1001/jamaoncol.2019.6650] [Citation(s) in RCA: 499] [Impact Index Per Article: 99.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/03/2019] [Indexed: 02/01/2023]
Abstract
Importance Approximately 25% of patients with early-stage breast cancer who receive (neo)adjuvant chemotherapy experience a recurrence within 5 years. Improvements in therapy are greatly needed. Objective To determine if pembrolizumab plus neoadjuvant chemotherapy (NACT) in early-stage breast cancer is likely to be successful in a 300-patient, confirmatory randomized phase 3 neoadjuvant clinical trial. Design, Setting, and Participants The I-SPY2 study is an ongoing open-label, multicenter, adaptively randomized phase 2 platform trial for high-risk, stage II/III breast cancer, evaluating multiple investigational arms in parallel. Standard NACT serves as the common control arm; investigational agent(s) are added to this backbone. Patients with ERBB2 (formerly HER2)-negative breast cancer were eligible for randomization to pembrolizumab between November 2015 and November 2016. Interventions Participants were randomized to receive taxane- and anthracycline-based NACT with or without pembrolizumab, followed by definitive surgery. Main Outcomes and Measures The primary end point was pathologic complete response (pCR). Secondary end points were residual cancer burden (RCB) and 3-year event-free and distant recurrence-free survival. Investigational arms graduated when demonstrating an 85% predictive probability of success in a hypothetical confirmatory phase 3 trial. Results Of the 250 women included in the final analysis, 181 were randomized to the standard NACT control group (median [range] age, 47 [24.77] years). Sixty-nine women (median [range] age, 50 [27-71] years) were randomized to 4 cycles of pembrolizumab in combination with weekly paclitaxel followed by AC; 40 hormone receptor (HR)-positive and 29 triple-negative. Pembrolizumab graduated in all 3 biomarker signatures studied. Final estimated pCR rates, evaluated in March 2017, were 44% vs 17%, 30% vs 13%, and 60% vs 22% for pembrolizumab vs control in the ERBB2-negative, HR-positive/ERBB2-negative, and triple-negative cohorts, respectively. Pembrolizumab shifted the RCB distribution to a lower disease burden for each cohort evaluated. Adverse events included immune-related endocrinopathies, notably thyroid abnormalities (13.0%) and adrenal insufficiency (8.7%). Achieving a pCR appeared predictive of long-term outcome, where patients with pCR following pembrolizumab plus chemotherapy had high event-free survival rates (93% at 3 years with 2.8 years' median follow-up). Conclusions and Relevance When added to standard neoadjuvant chemotherapy, pembrolizumab more than doubled the estimated pCR rates for both HR-positive/ERBB2-negative and triple-negative breast cancer, indicating that checkpoint blockade in women with early-stage, high-risk, ERBB2-negative breast cancer is highly likely to succeed in a phase 3 trial. Pembrolizumab was the first of 10 agents to graduate in the HR-positive/ERBB2-negative signature. Trial Registration ClinicalTrials.gov Identifier: NCT01042379.
Collapse
Affiliation(s)
- Rita Nanda
- The University of Chicago, Chicago, Illinois
| | | | | | | | | | | | | | | | | | | | - Amy Clark
- University of Pennsylvania, Philadelphia
| | - Kathy Albain
- Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | | | | | | | | | | | | | | | | | | | | | | | | | - Smita Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | - Amy Wilson
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | | | | | - Adam Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Vagia E, Mahalingam D, Cristofanilli M. The Landscape of Targeted Therapies in TNBC. Cancers (Basel) 2020; 12:E916. [PMID: 32276534 PMCID: PMC7226210 DOI: 10.3390/cancers12040916] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
Triple negative breast cancer (TNBC) constitutes the most aggressive molecular subtype among breast tumors. Despite progress on the underlying tumor biology, clinical outcomes for TNBC unfortunately remain poor. The median overall survival for patients with metastatic TNBC is approximately eighteen months. Chemotherapy is the mainstay of treatment while there is a growing body of evidence that targeted therapies may be on the horizon with poly-ADP-ribose polymerase (PARP) and immune check-point inhibitors already established in the treatment paradigm of TNBC. A large number of novel therapeutic agents are being evaluated for their efficacy in TNBC. As novel therapeutics are now incorporated into clinical practice, it is clear that tumor heterogeneity and clonal evolution can result to de novo or acquired treatment resistance. As precision medicine and next generation sequencing is part of cancer diagnostics, tailored treatment approaches based on the expression of molecular markers are currently being implemented in clinical practice and clinical trial design. The scope of this review is to highlight the most relevant current knowledge regarding underlying molecular profile of TNBC and its potential application in clinical practice.
Collapse
Affiliation(s)
- Elena Vagia
- Division of Hematology Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (D.M.); (M.C.)
| | | | | |
Collapse
|
232
|
Pan-cancer analysis reveals synergistic effects of CDK4/6i and PARPi combination treatment in RB-proficient and RB-deficient breast cancer cells. Cell Death Dis 2020; 11:219. [PMID: 32249776 PMCID: PMC7136254 DOI: 10.1038/s41419-020-2408-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022]
Abstract
DNA damage results in mutations and plays critical roles in cancer development, progression, and treatment. Targeting DNA damage response in cancers by inhibiting poly-(ADP-ribose) polymerases (PARPs) offers an important therapeutic strategy. However, the failure of PARP inhibitors to markedly benefit patients suggests the necessity for developing new strategies to improve their efficacy. Here, we show that the expression of cyclin-dependent kinase 4/6 (CDK4/6) complex members significantly correlates with mutations (as proxies of DNA damages), and that the combination of CDK4/6 and PARP inhibitors shows synergy in both RB-proficient and RB-deficient breast cancer cells. As PARPs constitute sensors of DNA damage and are broadly involved in multiple DNA repair pathways, we hypothesized that the combined inhibition of PARPs and DNA repair (or repair-related) pathways critical for cancer (DRPCC) should show synergy. To identify druggable candidate DRPCC(s), we analyzed the correlation between the genome-wide expression of individual genes and the mutations for 27 different cancer types, assessing 7146 exomes and over 1,500,000 somatic mutations. Pathway enrichment analyses of the top-ranked genes correlated with mutations indicated “cell cycle pathway” as the top candidate DRPCC. Additionally, among functional cell-cycle complexes, the CDK4/6 complex showed the most significant negative correlation with mutations, also suggesting that combined CDK4/6 and PARP inhibition might exhibit synergy. Furthermore, combination treatment showed synergy in not only RB-proficient but also RB-deficient breast cancer cells in a reactive oxygen species-dependent manner. These findings suggest a potential therapeutic strategy to improve the efficacy of PARP and CDK4/6 inhibitors in cancer treatment.
Collapse
|
233
|
Chien AJ, Tripathy D, Albain KS, Symmans WF, Rugo HS, Melisko ME, Wallace AM, Schwab R, Helsten T, Forero-Torres A, Stringer-Reasor E, Ellis ED, Kaplan HG, Nanda R, Jaskowiak N, Murthy R, Godellas C, Boughey JC, Elias AD, Haley BB, Kemmer K, Isaacs C, Clark AS, Lang JE, Lu J, Korde L, Edmiston KK, Northfelt DW, Viscusi RK, Yee D, Perlmutter J, Hylton NM, van’t Veer LJ, DeMichele A, Wilson A, Peterson G, Buxton MB, Paoloni M, Clennell J, Berry S, Matthews JB, Steeg K, Singhrao R, Hirst GL, Sanil A, Yau C, Asare SM, Berry DA, Esserman LJ, on behalf of I-SPY 2 Consortium. MK-2206 and Standard Neoadjuvant Chemotherapy Improves Response in Patients With Human Epidermal Growth Factor Receptor 2-Positive and/or Hormone Receptor-Negative Breast Cancers in the I-SPY 2 Trial. J Clin Oncol 2020; 38:1059-1069. [PMID: 32031889 PMCID: PMC7106976 DOI: 10.1200/jco.19.01027] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2019] [Indexed: 01/07/2023] Open
Abstract
PURPOSE The phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin is a key pathway of survival and therapeutic resistance in breast cancer. We evaluated the pan-Akt inhibitor MK-2206 in combination with standard therapy in patients with high-risk early-stage breast cancer. PATIENTS AND METHODS I-SPY 2 is a multicenter, phase II, open-label, adaptively randomized neoadjuvant platform trial that screens experimental therapies and efficiently identifies potential predictive biomarker signatures. Patients are categorized by human epidermal growth factor receptor 2 (HER2), hormone receptor (HR), and MammaPrint statuses in a 2 × 2 × 2 layout. Patients within each of these 8 biomarker subtypes are adaptively randomly assigned to one of several experimental therapies, including MK-2206, or control. Therapies are evaluated for 10 biomarker signatures, each of which is a combination of these subtypes. The primary end point is pathologic complete response (pCR). A therapy graduates with one or more of these signatures if and when it has an 85% Bayesian predictive probability of success in a hypothetical phase III trial, adjusting for biomarker covariates. Patients in the current report received standard taxane- and anthracycline-based neoadjuvant therapy without (control) or with oral MK-2206 135 mg/week. RESULTS MK-2206 graduated with 94 patients and 57 concurrently randomly assigned controls in 3 graduation signatures: HR-negative/HER2-positive, HR-negative, and HER2-positive. Respective Bayesian mean covariate-adjusted pCR rates and percentage probability that MK-2206 is superior to control were 0.48:0.29 (97%), 0.62:0.36 (99%), and 0.46:0.26 (94%). In exploratory analyses, MK-2206 evinced a numerical improvement in event-free survival in its graduating signatures. The most significant grade 3-4 toxicity was rash (14% maculopapular, 8.6% acneiform). CONCLUSION The Akt inhibitor MK-2206 combined with standard neoadjuvant therapy resulted in higher estimated pCR rates in HR-negative and HER2-positive breast cancer. Although MK-2206 is not being further developed at this time, this class of agents remains of clinical interest.
Collapse
Affiliation(s)
- A. Jo Chien
- University of California, San Francisco, San Francisco, CA
| | | | | | | | - Hope S. Rugo
- University of California, San Francisco, San Francisco, CA
| | | | | | | | | | | | | | | | | | - Rita Nanda
- The University of Chicago Medical Center, Chicago, IL
| | | | - Rashmi Murthy
- The University of Texas, MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | - Janice Lu
- University of Southern California, Los Angeles, CA
| | - Larissa Korde
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | | | | | | | - Douglas Yee
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | | | - Nola M. Hylton
- University of California, San Francisco, San Francisco, CA
| | | | | | - Amy Wilson
- Quantum Leap Healthcare Collaborative, San Francisco, CA
| | - Garry Peterson
- University of California, San Francisco, San Francisco, CA
| | | | | | | | | | | | | | - Ruby Singhrao
- University of California, San Francisco, San Francisco, CA
| | | | | | - Christina Yau
- University of California, San Francisco, San Francisco, CA
| | - Smita M. Asare
- Quantum Leap Healthcare Collaborative, San Francisco, CA
| | | | | | - on behalf of I-SPY 2 Consortium
- University of California, San Francisco, San Francisco, CA
- The University of Texas, MD Anderson Cancer Center, Houston, TX
- Loyola University, Chicago, IL
- University of California, San Diego, La Jolla, CA
- University of Alabama at Birmingham, Birmingham, AL
- Swedish Cancer Institute, Seattle, WA
- The University of Chicago Medical Center, Chicago, IL
- Mayo Clinic, Rochester, MN
- University of Colorado Denver, Denver, CO
- University of Texas Southwestern, Dallas, TX
- Oregon Health & Science University, Portland, OR
- Georgetown University, Washington, DC
- University of Pennsylvania, Philadelphia, PA
- University of Southern California, Los Angeles, CA
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
- Inova Health System, Falls Church, VA
- Mayo Clinic, Scottsdale, AZ
- University of Arizona, Tucson, AZ
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
- Gemini Group, Bad Axe, MI
- Quantum Leap Healthcare Collaborative, San Francisco, CA
- Berry Consultants, Austin, TX
| |
Collapse
|
234
|
Diana A, Carlino F, Franzese E, Oikonomidou O, Criscitiello C, De Vita F, Ciardiello F, Orditura M. Early Triple Negative Breast Cancer: Conventional Treatment and Emerging Therapeutic Landscapes. Cancers (Basel) 2020; 12:E819. [PMID: 32235297 PMCID: PMC7225917 DOI: 10.3390/cancers12040819] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/19/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
Triple negative breast cancers (TNBCs) are characterized by worse prognosis, higher propensity to earlier metastases, and shorter survival after recurrence compared with other breast cancer subtypes. Anthracycline- and taxane-based chemotherapy is still the mainstay of treatment in early stages, although several escalation approaches have been evaluated to improve survival outcomes. The addition of platinum salts to standard neoadjuvant chemotherapy (NACT) remains controversial due to the lack of clear survival advantage, and the use of adjuvant capecitabine represents a valid treatment option in TNBC patients with residual disease after NACT. Recently, several clinical trials showed promising results through the use of poly ADP-ribose polymerase (PARP) inhibitors and by incorporating immunotherapy with chemotherapy, enriching treatment options beyond conventional cytotoxic agents. In this review, we provided an overview on the current standard of care and a comprehensive update of the recent advances in the management of early stage TNBC and focused on the latest emerging biomarkers and their clinical application to select the best therapeutic strategy in this hard-to-treat population.
Collapse
Affiliation(s)
- Anna Diana
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Francesca Carlino
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Elisena Franzese
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Olga Oikonomidou
- Cancer Research UK, Edinburgh Centre, MRC Institute Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, UK;
| | | | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Fortunato Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Michele Orditura
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| |
Collapse
|
235
|
Golshan M, Loibl S, Wong SM, Houber JB, O'Shaughnessy J, Rugo HS, Wolmark N, McKee MD, Maag D, Sullivan DM, Metzger-Filho O, Von Minckwitz G, Geyer CE, Sikov WM, Untch M. Breast Conservation After Neoadjuvant Chemotherapy for Triple-Negative Breast Cancer: Surgical Results From the BrighTNess Randomized Clinical Trial. JAMA Surg 2020; 155:e195410. [PMID: 31913413 DOI: 10.1001/jamasurg.2019.5410] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Importance Neoadjuvant systemic therapy (NST) is often administered to enable breast-conserving therapy (BCT) in stages II to III breast cancer. Objectives To prospectively evaluate the role of NST in conversion from BCT ineligibility to BCT eligibility and to assess the association of response to NST, germline BRCA (gBRCA) status, and region of treatment with surgical choice in women with triple-negative breast cancer (TNBC). Design, Setting, and Participants This prespecified secondary analysis of a multicentered, phase 3, double-blind, randomized clinical trial (BrighTNess) enrolled 634 eligible women across 145 centers in 15 countries in North America, Europe, and Asia. Women with operable, clinical stages II to III TNBC who underwent gBRCA mutation testing before initiating NST were eligible to participate. Data were collected from April 1, 2014, to December 8, 2016. This preplanned analysis was performed from January 5, 2018, to October 28, 2019. Interventions Study participants were randomized to receive 12 weeks of weekly paclitaxel alone or with the addition of carboplatin and/or veliparib, followed by 4 cycles of doxorubicin hydrochloride and cyclophosphamide. Main Outcomes and Measures Surgeons assessed BCT candidacy by clinical and radiographic criteria before and after NST. Surgical choices and whether BCT eligibility was associated with the likelihood of pathologic complete response were then analyzed. Results Among the 634 randomized patients (median age, 51 [range, 22-78] years), pre- and post-NST assessments were available for 604 patients. Of 141 patients deemed BCT ineligible at baseline, 75 (53.2%) converted to BCT eligible. Overall, 342 (68.1%) of 502 patients deemed BCT eligible after NST underwent BCT, including 42 (56.0%) of the 75 who converted to BCT eligible. Patients treated in Europe and Asia were more likely to undergo BCT (odds ratio, 2.66; 95% CI, 1.84-3.84) compared with those treated in North America. Among patients without gBRCA mutation undergoing mastectomy, those treated in North America were more likely to undergo contralateral prophylactic mastectomy (57 of 81 [70.4%] vs 6 of 30 [20.0%]; P < .001). Rates of pathologic complete response were similar between patients deemed BCT eligible at baseline and those who were BCT ineligible but converted to BCT eligibility after NST (55.3 [235 of 425] vs 49.3% [37 of 75]; P = .38). Conclusions and Relevance This prospective analysis of NST and BCT eligibility in TNBC demonstrates a conversion from BCT ineligibility to BCT eligibility of 53.2%. Lower BCT rates among eligible patients and higher bilateral mastectomy rates among patients without gBRCA mutation in North America merit investigation. Trial Registration ClinicalTrials.gov identifier: NCT02032277.
Collapse
Affiliation(s)
- Mehra Golshan
- Department of Surgery, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sibylle Loibl
- Department of Medical Oncology, German Breast Group, Neu-Isenburg, Germany
| | - Stephanie M Wong
- Department of Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Joyce O'Shaughnessy
- Department of Medical Oncology, Texas Oncology-Baylor Sammons Cancer Center, US Oncology, Dallas
| | - Hope S Rugo
- Department of Medical Oncology, University of California, San Francisco
| | - Norman Wolmark
- Department of Surgery, Allegheny General Hospital, Pittsburgh, Pennsylvania
| | | | | | | | - Otto Metzger-Filho
- Department of Surgery, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Charles E Geyer
- Department of Medical Oncology, Virginia Commonwealth University Massey Cancer Center, Richmond
| | - William M Sikov
- Department of Medical Oncology, Women and Infants Hospital of Rhode Island, Providence
| | - Michael Untch
- Department of Breast Surgery, Helios Klinikum Berlin-Buch, Berlin, Germany
| |
Collapse
|
236
|
Iwase M, Ando M, Aogi K, Aruga T, Inoue K, Shimomura A, Tokunaga E, Masuda N, Yamauchi H, Yamashita T, Iwata H. Long-term survival analysis of addition of carboplatin to neoadjuvant chemotherapy in HER2-negative breast cancer. Breast Cancer Res Treat 2020; 180:687-694. [PMID: 32140811 DOI: 10.1007/s10549-020-05580-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/18/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE Addition of carboplatin (CBDCA) to neoadjuvant chemotherapy (NAC) in triple-negative breast cancer (TNBC) has improved pathological complete response (pCR) rates in previous studies. We present long-term survival outcomes (disease-free survival [DFS], pre-planned secondary endpoint; overall survival [OS], post hoc exploratory endpoint) of our randomized study of the addition of CBDCA to NAC for HER2-negative breast cancer. METHODS Patients with stage II/III, HER2-negative breast cancer (N = 179) were randomly assigned to receive CP-CEF (four 3-week cycles of CBDCA [area under the curve, 5 mg/mL/min, day 1] and weekly paclitaxel [wPTX, 80 mg/m2, day 1, 8, 15] followed by four 3-week cycles of cyclophosphamide, epirubicin, and 5-fluorouracil [CEF, 500/100/500 mg/m2]) or P-CEF (four cycles of wPTX followed by four cycles of CEF) as NAC. DFS and OS were analyzed at each population of pCR status and assigned treatment arm. RESULTS Of 179 patients, 154 were available for long-term follow-up. At a median follow-up of 6.6 years (range, 0.7-8.0 years), patients who achieved pCR [n = 42, 23.5% (CP-CEF: n = 28, P-CEF: n = 16)] had longer DFS and OS than non-pCR patients [DFS; HR 0.15 (0.04-0.61), P = 0.008, OS; log-rank P = 0.003]. Addition of carboplatin to NAC significantly improved DFS and OS in the subset of patients with TNBC [DFS: HR, 0.22 (0.06-0.82), P = 0.015; OS: HR, 0.12 (0.01-0.96), P = 0.046], but not in the subset of patients with hormone receptor-positive disease or among all patients. CONCLUSIONS Addition of carboplatin to neoadjuvant chemotherapy significantly improved DFS and OS in patients with TNBC but not in those with hormone receptor-positive, HER2-negative breast cancer.
Collapse
Affiliation(s)
- Madoka Iwase
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, 464-8681, Japan
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masashi Ando
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan.
| | - Kenjiro Aogi
- Department of Breast Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Tomoyuki Aruga
- Department of Breast Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Kenichiro Inoue
- Division of Breast Oncology, Saitama Cancer Center, Saitama, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Eriko Tokunaga
- Department of Breast Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Norikazu Masuda
- Division of Breast, Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Hideko Yamauchi
- Department of Breast Surgical Oncology, St. Luke's International Hospital, Tokyo, Japan
| | - Toshinari Yamashita
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, 464-8681, Japan
| |
Collapse
|
237
|
Ran D, Zhou J, Chai Z, Li J, Xie C, Mao J, Lu L, Zhang Y, Wu S, Zhan C, Lu W. All-stage precisional glioma targeted therapy enabled by a well-designed D-peptide. Theranostics 2020; 10:4073-4087. [PMID: 32226540 PMCID: PMC7086363 DOI: 10.7150/thno.41382] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/11/2020] [Indexed: 12/17/2022] Open
Abstract
Uncontrollable cell proliferation and irreversible neurological damage make glioma one of the most deadly diseases in clinic. Besides the multiple biological barriers, glioma stem cells (GSCs) that are responsible for the maintenance and recurrence of tumor tissues also hinder the therapeutic efficacy of chemotherapy. Therefore, all-stage precisional glioma targeted therapy regimens that could efficiently deliver drugs to glioma cells and GSCs after overcoming multiple barriers have received increasing scrutiny. Methods: A polymeric micelle-based drug delivery system was developed by modifying a "Y-shaped" well-designed ligand of both GRP78 protein and quorum sensing receptor to achieve all-stage precisional glioma targeting, then we evaluated the targeting ability and barrier penetration ability both in vitro and in vivo. In order to achieve all-stage precisional therapy, we need kill both GSCs and glioma related cells. Parthenolide (PTL) has been investigated for its selective toxicity to glioma stem cells while Paclitaxel (PTX) and Temozolomide (TMZ) are widely used in experimental and clinical therapy of glioma respectively. So the in vivo anti-glioma effect of combination therapy was evaluated by Kaplan-Meier survival analysis and immunohistochemical (IHC) examination of tumor tissues. Results: The "Y-shaped" well-designed peptide, termed DWVAP, exhibited excellent glioma (and GSCs) homing and barrier penetration ability. When modified on micelle surface, DWVAP peptide significantly enhanced accumulation of micelles in brain and glioma. In addition, DWVAP micelles showed no immunogenicity and cytotoxicity, which could guarantee their safety when used in vivo. Treatment of glioma-bearing mice with PTL loaded DWVAP modified PEG-PLA micelles plus PTX loaded DWVAP modified PEG-PLA micelles or PTL loaded DWVAP modified PEG-PLA micelles plus TMZ showed improved anti-tumor efficacy in comparison to PTL and PTX loaded unmodified micelles or PTL loaded unmodified micelles plus TMZ. Conclusion: Combination of all-stage targeting strategy and concomitant use of chemotherapeutics and stem cell inhibitors could achieve precise targeted therapy for glioma.
Collapse
|
238
|
Sharma P, Connolly RM, Roussos Torres ET, Thompson A. Best Foot Forward: Neoadjuvant Systemic Therapy as Standard of Care in Triple-Negative and HER2-Positive Breast Cancer. Am Soc Clin Oncol Educ Book 2020; 40:1-16. [PMID: 32315235 DOI: 10.1200/edbk_281381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Neoadjuvant systemic treatment of early-stage breast cancer has been used to improve resectability and reduce the extent of breast and axillary surgery. More recently, several other merits of neoadjuvant systemic treatment have emerged, including the ability to tailor clinically available adjuvant systemic therapy options based on pathologic response and to serve as a platform for early assessment of novel agents and response biomarkers and as an avenue for treatment optimization investigations (local and systemic therapy escalation and de-escalation trials guided by pathologic response). Attainment of a pathologic complete response (pCR) is associated with excellent long-term outcomes; conversely, the presence of residual disease is associated with a high risk of recurrence for patients with HER2-positive breast cancer and triple-negative breast cancer (TNBC). Treatment strategies in early-stage HER2-positive breast cancer include regimens incorporating trastuzumab, pertuzumab, ado-trastuzumab emtansine, and neratinib, resulting in high pCR rates and overall excellent long-term outcomes. Currently available cytotoxic regimens yield pCR for 35% to 55% of patients with TNBC, and immune checkpoint inhibition is showing early promise for this subtype. New drug and predictive biomarker evaluations in the neoadjuvant setting aim to develop optimal treatment strategies for the individual patient, with the ultimate goal of maximizing efficacy and minimizing toxicity. Research efforts involving novel agents are being undertaken to address the high risk of recurrence for patients with residual disease. Omission of breast surgery following neoadjuvant chemotherapy requires further development of imaging and biopsy techniques to accurately assess the extent of residual disease before clinical application.
Collapse
Affiliation(s)
- Priyanka Sharma
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Westwood, KS
| | | | | | - Alastair Thompson
- Department of Surgery, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
239
|
Seidman AD, Maues J, Tomlin T, Bhatnagar V, Beaver JA. The Evolution of Clinical Trials in Metastatic Breast Cancer: Design Features and Endpoints That Matter. Am Soc Clin Oncol Educ Book 2020; 40:1-11. [PMID: 32223668 DOI: 10.1200/edbk_280451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The evolution of thought in assessing benefit in clinical trials of systemic therapy for metastatic breast cancer (MBC) is well documented, with most agents garnering regulatory approval based either on an advantage in overall survival (OS), time to progression (TTP), or progression-free survival (PFS) over an existing standard of care or objective response rate (ORR). Previous guidance for industry on clinical trial endpoints for the approval of cancer drugs and biologics was provided by the U.S. Food and Drug Administration (FDA) in 2007 and recently updated in 2018. The more recent FDA guidance recognizes that advances in science are facilitating the development of oncology products, which "may also result in the identification of additional endpoints that may be used to support approval of oncology products." This article critically addressed the evolution of thought on the advancement of clinical trials in MBC, from various stakeholder perspectives. Despite the term "stakeholder," the objective of all co-authors and parties concerned is to promote and inform the optimal design, conduct, and reporting of clinical trials for women with advanced breast cancer toward improving and extending lives. This article provides an overview of the evolving perspectives on this issue from the physician, regulatory agency, and patient and/or advocate points of view.
Collapse
Affiliation(s)
| | | | | | - Vishal Bhatnagar
- Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, MD
| | | |
Collapse
|
240
|
Garufi G, Palazzo A, Paris I, Orlandi A, Cassano A, Tortora G, Scambia G, Bria E, Carbognin L. Neoadjuvant therapy for triple-negative breast cancer: potential predictive biomarkers of activity and efficacy of platinum chemotherapy, PARP- and immune-checkpoint-inhibitors. Expert Opin Pharmacother 2020; 21:687-699. [PMID: 32052646 DOI: 10.1080/14656566.2020.1724957] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Despite recent advances in the molecular characterization of triple-negative breast cancer (TNBC), the standard treatment for early-stage TNBC is represented by the historically used anthracycline and taxane-based chemotherapy. In this modern era of precision medicine, several new therapeutic strategies and novel agents have been investigated in the neoadjuvant setting of TNBC, in order to individualize treatment. AREAS COVERED This review provides a comprehensive overview of the currently available evidence regarding the activity and efficacy of platinum agents, PARP- and immune-checkpoint-inhibitors for the neoadjuvant treatment of TNBC, highlighting the available data on potential predictive biomarkers of response or resistance to such treatments. EXPERT OPINION The genomic and immune landscape of TNBC has encouraged the exploration of drugs that interfere with the DNA repair mechanism and that modulate immune response. Overall, these drugs seem to improve the pCR rate in TNBC, despite preliminary and heterogeneous results. Taking into account the economic issues and the side effects of these drugs, it is crucial to further explore the potential predictive role of BRCA mutational status and homologous recombination deficiency score, for platinum agents and PARP-inhibitors, and tumor infiltrating lymphocytes and other immune biomarkers for checkpoint inhibitors, respectively.
Collapse
Affiliation(s)
- Giovanna Garufi
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy.,Università Cattolica Del Sacro Cuore , Roma, Italy
| | - Antonella Palazzo
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy
| | - Ida Paris
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy
| | - Armando Orlandi
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy
| | - Alessandra Cassano
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy.,Università Cattolica Del Sacro Cuore , Roma, Italy
| | - Giampaolo Tortora
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy.,Università Cattolica Del Sacro Cuore , Roma, Italy
| | - Giovanni Scambia
- Università Cattolica Del Sacro Cuore , Roma, Italy.,Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy
| | - Emilio Bria
- Oncologia Medica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy.,Università Cattolica Del Sacro Cuore , Roma, Italy
| | - Luisa Carbognin
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Roma, Italy
| |
Collapse
|
241
|
Dediu M, Zielinski C. A Proposal to Redefine Pathologic Complete Remission as Endpoint following Neoadjuvant Chemotherapy in Early Breast Cancer. Breast Care (Basel) 2020; 15:67-71. [PMID: 32231500 PMCID: PMC7098275 DOI: 10.1159/000500620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/20/2018] [Indexed: 12/24/2022] Open
Abstract
Many analyses of the efficacy of neoadjuvant treatment (NAT) for early breast cancer including a meta-analysis derived from 10 randomized trials came to the conclusion that patients who would achieve pathologic complete response (pCR) following NAT would experience significant improvement in disease-free and overall survival (OS). Thus, pCR was proposed as a surrogate endpoint for OS, with pCR representing a robust prognostic marker for survival at an individual level. In the current analysis, we argue that OS following NAT-induced pCR might have reflected the initial prognosis of patients mainly defined - among other factors - by the initial pathological lymph node status while being largely independent on the type of administrated treatment, thus pleading against the pCR surrogacy hypothesis. We therefore propose to redefine pCR as a surrogate endpoint of NAT trials by the involvement of additional biologic parameters.
Collapse
Affiliation(s)
| | - Christoph Zielinski
- Comprehensive Cancer Center, General Hospital, Medical University Vienna, Vienna, Austria
| |
Collapse
|
242
|
Rummel SK, Lovejoy LA, Turner CE, Shriver CD, Ellsworth RE. Should Genetic Testing for Cancer Predisposition Be Standard-of-Care for Women with Invasive Breast Cancer? The Murtha Cancer Center Experience. Cancers (Basel) 2020; 12:E234. [PMID: 31963545 PMCID: PMC7016980 DOI: 10.3390/cancers12010234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/22/2022] Open
Abstract
Currently, genetic testing is offered only to women diagnosed with breast cancer who meet a defined set of criteria and is not included as standard-of-care treatment at the time of diagnosis. Thus, a significant number of women diagnosed with breast cancer may miss the opportunity for precision medical treatment and risk management. The effects of eligibility, timing, and uptake of genetic testing were evaluated in a cohort of women with invasive breast cancer diagnosed between 2001-2018. Risk status was estimated using NCCN BRCA1/2 testing criteria and panel testing was performed for all women who had genomic DNA available. Of the 1231 women, 57.8% were eligible for genetic testing. Uptake of testing within high-risk women was 42.7% of which 6.6% pursued clinical testing only after a second tumor event. Mutation frequencies were 15.8%, 5.5%, and 4.0% in high-risk women with clinical testing, high-risk women without clinical testing, and low-risk women, respectively. More than 4% of all patients harbored pathogenic or likely pathogenic mutations detected only in the research setting. Inclusion of panel testing at the time of diagnosis would allow for appropriate surveillance and treatment strategies to be employed to reduce the risk of secondary tumors and improve patient outcome.
Collapse
Affiliation(s)
- Seth K. Rummel
- Chan Soon-Shiong Institute of Molecular Medicine at Windber, 620 Seventh Street, Windber, PA 15963, USA; (S.K.R.); (L.A.L.)
| | - Leann A. Lovejoy
- Chan Soon-Shiong Institute of Molecular Medicine at Windber, 620 Seventh Street, Windber, PA 15963, USA; (S.K.R.); (L.A.L.)
| | - Clesson E. Turner
- Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (C.E.T.); (C.D.S.)
| | - Craig D. Shriver
- Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (C.E.T.); (C.D.S.)
- Clinical Breast Care Project, Murtha Cancer Center Research Program, 4494 North Palmer Road, Bethesda, MD 20889, USA
| | - Rachel E. Ellsworth
- Clinical Breast Care Project, Murtha Cancer Center Research Program, 4494 North Palmer Road, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| |
Collapse
|
243
|
DNA damage repair functions and targeted treatment in breast cancer. Breast Cancer 2020; 27:355-362. [PMID: 31898156 DOI: 10.1007/s12282-019-01038-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
Cell DNA is continuously attacked by endogenous and exogenous agents, which causes DNA damage. During long-term evolution, complex defense systems for DNA damage repair are formed by cells to maintain genome stability. Defects in the DNA damage repair process may lead to various diseases, including tumors. Therefore, DNA damage repair systems have become a new anti-tumor drug target. To date, a number of inhibitors related to DNA damage repair systems have been developed, particularly for tumors with BRCA1 and BRCA2 mutations. Poly (ADP-ribose) polymerase inhibitors developed by synthetic lethality are widely used in individualized tumor therapy. In this review, we briefly introduce the mechanisms underlying DNA damage repair, particularly in breast cancer, and mainly focus on new treatments targeting the DNA damage repair pathway in breast cancer.
Collapse
|
244
|
Pragmatic Trials and Approaches to Transforming Care. Clin Trials 2020. [DOI: 10.1007/978-3-030-35488-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
245
|
Boxer AL, Gold M, Feldman H, Boeve BF, Dickinson SLJ, Fillit H, Ho C, Paul R, Pearlman R, Sutherland M, Verma A, Arneric SP, Alexander BM, Dickerson BC, Dorsey ER, Grossman M, Huey ED, Irizarry MC, Marks WJ, Masellis M, McFarland F, Niehoff D, Onyike CU, Paganoni S, Panzara MA, Rockwood K, Rohrer JD, Rosen H, Schuck RN, Soares HD, Tatton N. New directions in clinical trials for frontotemporal lobar degeneration: Methods and outcome measures. Alzheimers Dement 2020; 16:131-143. [PMID: 31668596 PMCID: PMC6949386 DOI: 10.1016/j.jalz.2019.06.4956] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Frontotemporal lobar degeneration (FTLD) is the most common form of dementia for those under 60 years of age. Increasing numbers of therapeutics targeting FTLD syndromes are being developed. METHODS In March 2018, the Association for Frontotemporal Degeneration convened the Frontotemporal Degeneration Study Group meeting in Washington, DC, to discuss advances in the clinical science of FTLD. RESULTS Challenges exist for conducting clinical trials in FTLD. Two of the greatest challenges are (1) the heterogeneity of FTLD syndromes leading to difficulties in efficiently measuring treatment effects and (2) the rarity of FTLD disorders leading to recruitment challenges. DISCUSSION New personalized endpoints that are clinically meaningful to individuals and their families should be developed. Personalized approaches to analyzing MRI data, development of new fluid biomarkers and wearable technologies will help to improve the power to detect treatment effects in FTLD clinical trials and enable new, clinical trial designs, possibly leveraged from the experience of oncology trials. A computational visualization and analysis platform that can support novel analyses of combined clinical, genetic, imaging, biomarker data with other novel modalities will be critical to the success of these endeavors.
Collapse
Affiliation(s)
- Adam L. Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA
| | | | - Howard Feldman
- Department of Neurosciences, University of California San Diego, San Diego, CA
| | | | | | | | - Carole Ho
- Denali Therapeutics, San Francisco, CA
| | | | | | | | | | | | | | | | - Earl Ray Dorsey
- Center for Health and Technology, University of Rochester, Rochester, NY
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA
| | - Edward D. Huey
- Departments of Psychiatry and Neurology, Columbia University, NY
| | | | - William J. Marks
- Clinical Neurology, Verily Life Sciences, South San Francisco, CA
| | - Mario Masellis
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, ON, Canada; Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, ON, Canada
| | | | - Debra Niehoff
- Association for Frontotemporal Degeneration, Radnor, PA
| | - Chiadi U. Onyike
- Department Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University, Baltimore, MD
| | - Sabrina Paganoni
- Healey Center for ALS, Massachusetts General Hospital, Boston, MA
| | | | - Kenneth Rockwood
- Division of Geriatric Medicine, Dalhousie University, Halifax, NS
| | - Jonathan D. Rohrer
- Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - Howard Rosen
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA
| | - Robert N. Schuck
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, FDA, Silver Spring, MD
| | | | - Nadine Tatton
- Association for Frontotemporal Degeneration, Radnor, PA
| |
Collapse
|
246
|
Stegall MD, Smith B, Bentall A, Schinstock C. The need for novel trial designs, master protocols, and research consortia in transplantation. Clin Transplant 2020; 34:e13759. [PMID: 31889338 DOI: 10.1111/ctr.13759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/16/2019] [Indexed: 12/24/2022]
Abstract
Large multicenter, randomized controlled trials are the paradigm for determining the efficacy and safety of new therapies. However, applying this classical approach to many areas of transplantation is difficult. For most types of organ transplants, the number of transplants performed is too small for such a trial (lung, pancreas, or vascular composite transplantation are examples). In larger populations such as kidney transplantation, the major unmet needs commonly involve small subsets of patients (antibody-mediated rejection, recurrent renal disease, etc). This issue is not unique to transplantation and has been successfully overcome in other areas of medicine. In oncology, for example, novel trial designs such as adaptive trial design and master protocols are now relatively common. In addition, the existence of multicenter, ongoing clinical research consortia have greatly enhanced the successful implementation of these novel trial designs. In this manuscript, we examine how novel trial designs, master protocols, and research consortia might enhance studies in transplantation aimed at the regulatory approval of new agents. Our premise is that more efficient approaches to clinical trials already exist and, through a coordinated effort by researchers, the pharmaceutical industry, and regulatory bodies like the FDA, they can be implemented in transplantation.
Collapse
Affiliation(s)
- Mark D Stegall
- Departments of Surgery and Immunology, Mayo Clinic, Rochester, Minnesota
| | - Byron Smith
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Andrew Bentall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Carrie Schinstock
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
247
|
Tovey H, Cheang MCU. Identifying Biomarkers to Pair with Targeting Treatments within Triple Negative Breast Cancer for Improved Patient Stratification. Cancers (Basel) 2019; 11:E1864. [PMID: 31769425 PMCID: PMC6966447 DOI: 10.3390/cancers11121864] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/07/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022] Open
Abstract
The concept of precision medicine has been around for many years and recent advances in high-throughput sequencing techniques are enabling this to become reality. Within the field of breast cancer, a number of signatures have been developed to molecularly sub-classify tumours. Notable examples recently approved by National Institute for Health and Care Excellence in the UK to guide treatment decisions for oestrogen receptors (ER)+ human epidermal growth factor receptor 2 (HER2)- patients include Prosigna test, EndoPredict, and Oncotype DX. However, a population of still unmet need are those with triple negative breast cancer (TNBC). Accounting for 15-20% of patients, this population has comparatively poor prognosis and as yet no targeted treatment options. Studies have shown that some patients with TNBC respond favourably to DNA damaging drugs (carboplatin) or agents which inhibit DNA damage response (poly ADP ribose polymerase (PARP) inhibitors). Known to be a heterogeneous population, there is a need to identify further TNBC patients who may benefit from these treatments. A number of signatures have been identified based on association with treatment response or specific genetic features/pathways however many of these were not restricted to TNBC patients and as of yet are not common practice in the clinic.
Collapse
Affiliation(s)
- Holly Tovey
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| | - Maggie Chon U. Cheang
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
248
|
Abstract
OPINION STATEMENT Triple-negative breast cancer (TNBC) is a particularly aggressive subtype of breast cancer. TNBC is a heterogenous subtype of breast cancer that is beginning to be refined by its molecular characteristics and clinical response to a targeted therapeutic approach. Until recently the backbone of therapy against TNBC has been cytotoxic chemotherapy. However, the breast oncology community is now seeing encouraging clinical activity from molecularly targeted approaches to TNBC. Recently, we have seen 3 newly approved targeted therapies for TNBC, including the PARP inhibitors olaparib and talazoparib for germline BRCA mutation associated breast cancer (gBRCAm-BC) and most recently the checkpoint inhibitor, atezolizumab in combination with nab-paclitaxel for programmed death-ligand 1 (PD-L1+) advanced TNBC. Improved biomarkers are needed to inform better patient selection for treatment with checkpoint inhibition. Higher response rates are seen when checkpoint inhibitors are combined with chemotherapy in the first-line setting and the use of these agents at an earlier stage of the disease does show promise. Antibody-drug conjugates are generating much excitement and may allow re-examination of prior cytotoxics that failed in development due to toxicity. Tumor sequencing is identifying potential molecular targets and ongoing studies are evaluating novel small molecule agents in this field such as AKT inhibition and many others. The treatment paradigm of chemotherapy as "one size fits all" approach for management of TNBC is changing based on molecular subtyping. Soon, the term TNBC may no longer be appropriate, as this heterogenous subtype of breast cancer is further refined by its molecular characteristics and clinical response to a targeted therapeutic approach.
Collapse
Affiliation(s)
- Tomas G Lyons
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 E. 66th Street, New York, NY, 10065, USA.
| |
Collapse
|
249
|
Wang H, Yee D. I-SPY 2: a Neoadjuvant Adaptive Clinical Trial Designed to Improve Outcomes in High-Risk Breast Cancer. CURRENT BREAST CANCER REPORTS 2019; 11:303-310. [PMID: 33312344 DOI: 10.1007/s12609-019-00334-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Purpose of Review The I-SPY 2 trial is an adaptive clinical trial platform designed to improve outcomes in high-risk breast cancer patients by testing new drugs in the neoadjuvant setting. The intent of this review is to discuss background, study structure, innovation, and outcomes of the I-SPY 2 trial. Recent Findings I-SPY 2 evaluates new agents combined with standard therapy with pathologic complete response (pCR) as the primary endpoint. I-SPY-2 uses clinical biomarkers to classify breast cancer into 10 subtypes, with Bayesian adaptive randomization to allow individualized patient assignment to therapy arms to maximize treatment effects. A total of 7 drugs have graduated from I-SPY 2. Multiple new agents are currently in active enrollment in I-SPY 2. Summary I-SPY 2 uses an individualized approach in clinical trial design to improve high-risk breast cancer outcomes. The purpose of this review is to encourage further research and innovation in this area and bring more precise treatment options to breast cancer patients.
Collapse
Affiliation(s)
- Haiyun Wang
- Masonic Cancer Center, University of Minnesota, MMC 806, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Douglas Yee
- Masonic Cancer Center, University of Minnesota, MMC 806, 420 Delaware St SE, Minneapolis, MN 55455, USA
| |
Collapse
|
250
|
Yi M, Dong B, Qin S, Chu Q, Wu K, Luo S. Advances and perspectives of PARP inhibitors. Exp Hematol Oncol 2019; 8:29. [PMID: 31737426 PMCID: PMC6849303 DOI: 10.1186/s40164-019-0154-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022] Open
Abstract
DNA damage repair deficiency leads to the increased risk of genome instability and oncogenic transformation. In the meanwhile, this deficiency could be exploited for cancer treatment by inducing excessive genome instability and catastrophic DNA damage. Continuous DNA replication in cancer cells leads to higher demand of DNA repair components. Due to the oncogenic loss of some DNA repair effectors (e.g. BRCA) and incomplete DNA repair repertoire, some cancer cells are addicted to certain DNA repair pathways such as Poly (ADP-ribose) polymerase (PARP)-related single-strand break repair pathway. The interaction between BRCA and PARP is a form of synthetic lethal effect which means the simultaneously functional loss of two genes lead to cell death, while defect in any single gene has a slight effect on cell viability. Based on synthetic lethal theory, Poly (ADP-ribose) polymerase inhibitor (PARPi) was developed aiming to selectively target cancer cells harboring BRCA1/2 mutations. Recently, a growing body of evidence indicated that a broader population of patients could benefit from PARPi therapy far beyond those with germline BRCA1/2 mutated tumors. Numerous biomarkers including homologous recombination deficiency and high level of replication pressure also herald high sensitivity to PARPi treatment. Besides, a series of studies indicated that PARPi-involved combination therapy such as PARPi with additional chemotherapy therapy, immune checkpoint inhibitor, as well as targeted agent had a great advantage in overcoming PARPi resistance and enhancing PARPi efficacy. In this review, we summarized the advances of PARPi in clinical application. Besides, we highlighted multiple promising PARPi-based combination strategies in preclinical and clinical studies.
Collapse
Affiliation(s)
- Ming Yi
- 1Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Bing Dong
- 2Department of Molecular Pathology, The Affiliated Cancer Hospital, Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Shuang Qin
- 1Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Qian Chu
- 1Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Kongming Wu
- 1Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China.,3Department of Medical Oncology, The Affiliated Cancer Hospital, Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Suxia Luo
- 3Department of Medical Oncology, The Affiliated Cancer Hospital, Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| |
Collapse
|