101
|
Abstract
First-line CDK4/6 inhibitor ribociclib plus letrozole improves survival in the metastatic setting, but lack of accrual of African American women is a shortcoming. Predicting benefit in the early-stage setting and diverse enrollment in trials need to be priorities.1.
Collapse
|
102
|
Wolf DM, Yau C, Wulfkuhle J, Brown-Swigart L, Gallagher RI, Lee PRE, Zhu Z, Magbanua MJ, Sayaman R, O'Grady N, Basu A, Delson A, Coppé JP, Lu R, Braun J, Asare SM, Sit L, Matthews JB, Perlmutter J, Hylton N, Liu MC, Pohlmann P, Symmans WF, Rugo HS, Isaacs C, DeMichele AM, Yee D, Berry DA, Pusztai L, Petricoin EF, Hirst GL, Esserman LJ, van 't Veer LJ. Redefining breast cancer subtypes to guide treatment prioritization and maximize response: Predictive biomarkers across 10 cancer therapies. Cancer Cell 2022; 40:609-623.e6. [PMID: 35623341 PMCID: PMC9426306 DOI: 10.1016/j.ccell.2022.05.005] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/16/2022] [Accepted: 05/06/2022] [Indexed: 12/26/2022]
Abstract
Using pre-treatment gene expression, protein/phosphoprotein, and clinical data from the I-SPY2 neoadjuvant platform trial (NCT01042379), we create alternative breast cancer subtypes incorporating tumor biology beyond clinical hormone receptor (HR) and human epidermal growth factor receptor-2 (HER2) status to better predict drug responses. We assess the predictive performance of mechanism-of-action biomarkers from ∼990 patients treated with 10 regimens targeting diverse biology. We explore >11 subtyping schemas and identify treatment-subtype pairs maximizing the pathologic complete response (pCR) rate over the population. The best performing schemas incorporate Immune, DNA repair, and HER2/Luminal phenotypes. Subsequent treatment allocation increases the overall pCR rate to 63% from 51% using HR/HER2-based treatment selection. pCR gains from reclassification and improved patient selection are highest in HR+ subsets (>15%). As new treatments are introduced, the subtyping schema determines the minimum response needed to show efficacy. This data platform provides an unprecedented resource and supports the usage of response-based subtypes to guide future treatment prioritization.
Collapse
Affiliation(s)
- Denise M Wolf
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA.
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Lamorna Brown-Swigart
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Rosa I Gallagher
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Pei Rong Evelyn Lee
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Zelos Zhu
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark J Magbanua
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Rosalyn Sayaman
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Nicholas O'Grady
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amrita Basu
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amy Delson
- Breast Science Advocacy Core, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jean Philippe Coppé
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA
| | - Ruixiao Lu
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Jerome Braun
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, CA 94118, USA
| | - Laura Sit
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey B Matthews
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Nola Hylton
- Department of Radiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Minetta C Liu
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Paula Pohlmann
- MedStar Georgetown University Hospital, Georgetown University, Washington, DC 20057, USA
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hope S Rugo
- Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Angela M DeMichele
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas Yee
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Lajos Pusztai
- Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Gillian L Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94143, USA.
| |
Collapse
|
103
|
Semiglazov V, Tseluiko A, Kudaybergenova A, Artemyeva A, Krivorotko P, Donskih R. Immunology and immunotherapy in breast cancer. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0597. [PMID: 35676750 PMCID: PMC9196061 DOI: 10.20892/j.issn.2095-3941.2021.0597] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/07/2022] [Indexed: 12/31/2022] Open
Abstract
Immuno-oncology is a rapidly developing field in medicine. Drug combination therapies have already been studied in many clinical trials on various tumor types. In recent years, a checkpoint inhibition therapy with monoclonal antibodies targeting PD-1 and its ligand PD-L1 has been developed. Breast cancer had been examined in the field of immune-oncology relatively recently. This review focuses on clinical evidence regarding immune checkpoint inhibition for curative treatment of various breast cancer subtypes. In addition, we present the results of studies demonstrating the prognostic and predictive value of levels of tumorinfiltrating lymphocytes (CD4 and CD8), their quantitative ratios, and their correlation with regulatory genes (PD-1, PD-L1, and FOX-P3).
Collapse
Affiliation(s)
- Vladimir Semiglazov
- Petrov National Medicine Cancer-Research Center Ministry of Health, Saint-Petersburg 197758, Russia
| | - Andrey Tseluiko
- Petrov National Medicine Cancer-Research Center Ministry of Health, Saint-Petersburg 197758, Russia
| | - Asel Kudaybergenova
- Petrov National Medicine Cancer-Research Center Ministry of Health, Saint-Petersburg 197758, Russia
| | - Anna Artemyeva
- Petrov National Medicine Cancer-Research Center Ministry of Health, Saint-Petersburg 197758, Russia
| | - Petr Krivorotko
- Petrov National Medicine Cancer-Research Center Ministry of Health, Saint-Petersburg 197758, Russia
| | - Roman Donskih
- Petrov National Medicine Cancer-Research Center Ministry of Health, Saint-Petersburg 197758, Russia
| |
Collapse
|
104
|
Barker AD, Lee JS. Translating "Big Data" in Oncology for Clinical Benefit: Progress or Paralysis. Cancer Res 2022; 82:2072-2075. [PMID: 35416976 PMCID: PMC9306452 DOI: 10.1158/0008-5472.can-22-0100] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 01/07/2023]
Abstract
The molecular characterization of cancer through genomics, data from multiomics technologies, molecular-driven clinical trials, and internet-enabled devices capturing patient context and real-world data are creating an unprecedented big data revolution across the cancer research-care continuum. While big data has translated to benefit for some patients, it has also created new problems. Our intent in this brief communication is to explore some examples of progress and key challenges that remain. The problems are not intractable, but success will require rethinking and rebuilding an information and evidence-based learning system that moves beyond paralysis to shape a better future for patients with cancer.
Collapse
Affiliation(s)
- Anna D. Barker
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, California
- Complex Adaptive Systems Initiative and School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Jerry S.H. Lee
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, California
- Keck School of Medicine and Viterbi School of Engineering, University of Southern California, Los Angeles, California
| |
Collapse
|
105
|
A prospective trial of treatment de-escalation following neoadjuvant paclitaxel/trastuzumab/pertuzumab in HER2-positive breast cancer. NPJ Breast Cancer 2022; 8:63. [PMID: 35538105 PMCID: PMC9091255 DOI: 10.1038/s41523-022-00429-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 03/14/2022] [Indexed: 01/03/2023] Open
Abstract
De-escalating adjuvant therapy following pathologic complete response (pCR) to an abbreviated neoadjuvant regimen in human epidermal growth factor receptor 2-positive (HER2+) breast cancer is the focus of international research efforts. However, the feasibility of this approach and its appeal to patients and providers had not been formally investigated. We aimed to assess adherence to de-escalated adjuvant antibody doublet therapy (trastuzumab and pertuzumab [HP], without chemotherapy) among patients with pCR following neoadjuvant paclitaxel/HP (THP). In this single-arm prospective trial, patients with treatment-naïve stage II-III HER2+ breast cancer received neoadjuvant weekly paclitaxel ×12 and HP every 3 weeks ×4. The primary endpoint was receipt of adjuvant non-HER2-directed cytotoxic chemotherapy. Ninety-eight patients received ≥1 dose of THP on study. Patients had median age of 50 years, 86% had stage II tumors, and 34% were hormone receptor-negative. Five patients had incomplete clinical response following THP and received doxorubicin and cyclophosphamide before surgery; they were classified as non-pCR and censored from further analyses. The overall pCR rate was 56.7%. Among patients with pCR, the adherence rate to de-escalated antibody-only therapy (HP) was 98.2% (95% CI 90.3–100.0%), and the primary feasibility endpoint was reached. The majority of patients felt positive or neutral about their adjuvant treatment plans. With brief follow-up (median 19.1 months), there were no breast cancer recurrences. De-escalation of adjuvant chemotherapy among patients who experience pCR in early-stage HER2+ breast cancer is a practicable approach for both patients and physicians. Planned and ongoing prospective trials will determine the long-term efficacy of this approach. Trial registration clinicaltrials.gov, NCT03716180, https://clinicaltrials.gov/ct2/show/NCT03716180.
Collapse
|
106
|
Gandhi S, Brackstone M, Hong NJL, Grenier D, Donovan E, Lu FI, Skarpathiotakis M, Lee J, Boileau JF, Perera F, Simmons C, Joy AA, Tran WT, Tyono I, Van Massop A, Khalfan S. A Canadian national guideline on the neoadjuvant treatment of invasive breast cancer, including patient assessment, systemic therapy, and local management principles. Breast Cancer Res Treat 2022; 193:1-20. [PMID: 35224713 PMCID: PMC8993711 DOI: 10.1007/s10549-022-06522-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/16/2022] [Indexed: 12/11/2022]
Abstract
Purpose The neoadjuvant treatment of breast cancer (NABC) is a rapidly changing area that benefits from guidelines integrating evidence with expert consensus to help direct practice. This can optimize patient outcomes by ensuring the appropriate use of evolving neoadjuvant principles. Methods An expert panel formulated evidence-based practice recommendations spanning the entire neoadjuvant breast cancer treatment journey. These were sent for practice-based consensus across Canada using the modified Delphi methodology, through a secure online survey. Final recommendations were graded using the GRADE criteria for guidelines. The evidence was reviewed over the course of guideline development to ensure recommendations remained aligned with current relevant data. Results Response rate to the online survey was almost 30%; representation was achieved from various medical specialties from both community and academic centres in various Canadian provinces. Two rounds of consensus were required to achieve 80% or higher consensus on 59 final statements. Five additional statements were added to reflect updated evidence but not sent for consensus. Conclusions Key highlights of this comprehensive Canadian guideline on NABC include the use of neoadjuvant therapy for early stage triple negative and HER2 positive breast cancer, with subsequent adjuvant treatments for patients with residual disease. The use of molecular signatures, other targeted adjuvant therapies, and optimal response-based local regional management remain actively evolving areas. Many statements had evolving or limited data but still achieved high consensus, demonstrating the utility of such a guideline in helping to unify practice while further evidence evolves in this important area of breast cancer management.
Collapse
|
107
|
Chaft JE, Shyr Y, Sepesi B, Forde PM. Preoperative and Postoperative Systemic Therapy for Operable Non-Small-Cell Lung Cancer. J Clin Oncol 2022; 40:546-555. [PMID: 34985966 PMCID: PMC8853628 DOI: 10.1200/jco.21.01589] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/13/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
Cisplatin-based adjuvant chemotherapy remains the standard of care for patients with resected stage II or III non-small-cell lung cancer. However, biomarker-informed clinical trials are starting to push the management of early-stage lung cancer beyond cytotoxic chemotherapy. This review explores recent and ongoing studies focused on improving cytotoxic chemotherapy and incorporating targeted and immunotherapies in the management of early-stage, resectable lung cancer. Adjuvant osimertinib for patients with EGFR-mutant tumors, preoperative chemoimmunotherapy, and adjuvant immunotherapy could improve outcomes for selected patients with resectable lung cancer, and ongoing or planned studies leveraging biomarkers, immunotherapy, and targeted therapy may further improve survival. We also discuss the unique barriers associated with clinical trials of early-stage lung cancer and the need for innovative trial designs to overcome these challenges.
Collapse
Affiliation(s)
- Jamie E. Chaft
- Thoracic Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Patrick M. Forde
- Department of Medicine, Sidney Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
108
|
Second-Harmonic Generation Imaging Reveals Changes in Breast Tumor Collagen Induced by Neoadjuvant Chemotherapy. Cancers (Basel) 2022; 14:cancers14040857. [PMID: 35205605 PMCID: PMC8869853 DOI: 10.3390/cancers14040857] [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: 01/21/2022] [Accepted: 02/03/2022] [Indexed: 12/10/2022] Open
Abstract
Breast cancer is the most common invasive cancer in women, with most deaths attributed to metastases. Neoadjuvant chemotherapy (NACT) may be prescribed prior to surgical removal of the tumor for subsets of breast cancer patients but can have diverse undesired and off-target effects, including the increased appearance of the 'tumor microenvironment of metastasis', image-based multicellular signatures that are prognostic of breast tumor metastasis. To assess whether NACT can induce changes in two other image-based prognostic/predictive signatures derived from tumor collagen, we quantified second-harmonic generation (SHG) directionality and fiber alignment in formalin-fixed, paraffin-embedded sections of core needle biopsies and primary tumor excisions from 22 human epidermal growth factor receptor 2-overexpressing (HER2+) and 22 triple-negative breast cancers. In both subtypes, we found that SHG directionality (i.e., the forward-to-backward scattering ratio, or F/B) is increased by NACT in the bulk of the tumor, but not the adjacent tumor-stroma interface. Overall collagen fiber alignment is increased by NACT in triple-negative but not HER2+ breast tumors. These results suggest that NACT impacts the collagenous extracellular matrix in a complex and subtype-specific manner, with some prognostic features being unchanged while others are altered in a manner suggestive of a more metastatic phenotype.
Collapse
|
109
|
McAndrew NP. Updates on targeting human epidermal growth factor receptor 2-positive breast cancer: what's to know in 2021. Curr Opin Obstet Gynecol 2022; 34:41-45. [PMID: 34967814 DOI: 10.1097/gco.0000000000000762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW To highlight recent practice changing clinical trials, focusing on those leading to new drug approvals, in human epidermal growth factor receptor 2-positive (HER2+) breast cancer. RECENT FINDINGS The improved disease-free survival of adjuvant trastuzumab emtansine (T-DM1) over trastuzumab in patients with residual disease has made neoadjuvant sequencing of therapy standard for most patients with early stage disease. In patients with metastatic HER2+ breast cancer, trastuzumab deruxtecan has recently shown dramatically improved efficacy over T-DM1. Tucatinib is an oral tyrosine kinase inhibitor with best in class blood-brain barrier penetration. Margetuximab, a novel HER2-targeted chimeric monoclonal antibody with an engineered Fc receptor designed to activate local immune response, was recently approved in heavily pretreated patients based on modest but significant improvement in progression-free survival. SUMMARY Patients with HER2+ breast cancer have a variety of therapeutic options in the early stage and metastatic setting. Optimal sequencing of therapy will depend on patient-specific factors such as site of tumor progression and underlying comorbidities. De-escalation of the first-line metastatic regimen may be considered in select patients with hormone positive/HER2+ breast cancer, by using endocrine therapy instead of chemotherapy in combination with HER2-targeted therapy, which may improve side effects without sacrificing efficacy.
Collapse
|
110
|
Unveiling the Potential of Liquid Biopsy in HER2-Positive Breast Cancer Management. Cancers (Basel) 2022; 14:cancers14030587. [PMID: 35158855 PMCID: PMC8833720 DOI: 10.3390/cancers14030587] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Breast cancer (BC) is the most prevailing cancer in women worldwide. Amongst the different BC subtypes, human epidermal growth factor receptor 2 (HER2)-positive tumours are characterised by an overexpression of the HER2 membrane receptor. Nowadays, HER2-status assessment relies on immunohistochemical methodologies in the tumour tissue, which could be complemented by novel methodologies to improve the clinical management of these patients. In this regard, liquid biopsy is an easy, rapid, and minimally invasive tool to obtain circulating tumour components from body fluids. Herein, by reviewing the published studies, we aim to decipher the clinical validity of liquid biopsy in both early and metastatic HER2-positive BC. Abstract Invasive breast cancer (BC) is the most common cancer in women with a slightly increasing yearly incidence. BC immunohistochemical characterisation is a crucial tool to define the intrinsic nature of each tumour and personalise BC patients’ clinical management. In this regard, the characterisation of human epidermal growth factor receptor 2 (HER2) status guides physicians to treat with therapies tailored to this membrane receptor. Standardly, a tumour solid biopsy is therefore required, which is an invasive procedure and has difficulties to provide the complete molecular picture of the tumour. To complement these standard-of-care approaches, liquid biopsy is a validated methodology to obtain circulating tumour components such as circulating tumour DNA (ctDNA) and circulating tumour cells (CTCs) from body fluids in an easy-to-perform minimal-invasive manner. However, its clinical validity in cancer is still to be demonstrated. This review focusses on the utilisation of both ctDNA and CTCs in early and metastatic HER2-positive BC tumours. We discuss recently published studies deciphering the capacity of liquid biopsy to determine the response to neoadjuvant and adjuvant therapies as well as to predict patients’ outcomes.
Collapse
|
111
|
Gunasekara ADM, Anothaisintawee T, Youngkong S, Ha NT, McKay GJ, Attia J, Thakkinstian A. Neoadjuvant Treatment with HER2-Targeted Therapies in HER2-Positive Breast Cancer: A Systematic Review and Network Meta-Analysis. Cancers (Basel) 2022; 14:cancers14030523. [PMID: 35158791 PMCID: PMC8833584 DOI: 10.3390/cancers14030523] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/14/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Human epidermal growth factor receptor 2 (HER2)-positive breast cancer causes more aggressive progression of disease and poorer outcomes for patients. HER2-targeted medicines used as neoadjuvant systemic therapy could improve clinical outcomes in early-stage or locally advanced breast cancer patients. The purpose of this systematic review and network meta-analysis was to identify the neoadjuvant anti-HER2 therapy with the best balance between efficacy and safety. We found that trastuzumab emtansine + pertuzumab + chemotherapy had a high pathologic complete response with a low risk of adverse events compared to other neoadjuvant anti-HER2 regimens, while the pertuzumab + trastuzumab + chemotherapy regimen showed the highest disease-free survival. However, further trial data on neoadjuvant regimens with trastuzumab emtansine are needed to confirm these findings. Abstract This systematic review aimed to identify neoadjuvant anti-human epidermal growth factor receptor 2 (HER2) therapies with the best balance between efficacy and safety. Methods: A network meta-analysis was applied to estimate the risk ratios along with 95% confidence intervals (CIs) for pathological complete response (pCR) and serious adverse events (SAE). A mixed-effect parametric survival analysis was conducted to assess the disease-free survival (DFS) between treatments. Results: Twenty-one RCTs with eleven regimens of neoadjuvant anti-HER2 therapy (i.e., trastuzumab + chemotherapy (TC), lapatinib + chemotherapy (LC), pertuzumab + chemotherapy (PC), pertuzumab + trastuzumab (PT), trastuzumab emtansine + pertuzumab (T-DM1P), pertuzumab + trastuzumab + chemotherapy (PTC), lapatinib + trastuzumab + chemotherapy (LTC), trastuzumab emtansine + lapatinib + chemotherapy (T-DM1LC), trastuzumab emtansine + pertuzumab + chemotherapy(T-DM1PC), PTC followed by T-DM1P (PTC_T-DM1P), and trastuzumab emtansine (T-DM1)) and chemotherapy alone were included. When compared to TC, only PTC had a significantly higher DFS with a hazard ratio (95% CI) of 0.54 (0.32–0.91). The surface under the cumulative ranking curve (SUCRA) suggested that T-DM1LC (91.9%) was ranked first in achieving pCR, followed by the PTC_T-DM1P (90.5%), PTC (74.8%), and T-DM1PC (73.5%) regimens. For SAEs, LTC, LC, and T-DM1LC presented with the highest risks (SUCRA = 10.7%, 16.8%, and 20.8%), while PT (99.2%), T-DM1P (88%), and T-DM1 (83.9%) were the safest regimens. The T-DM1PC (73.5% vs. 71.6%), T-DM1 (70.5% vs. 83.9%), and PTC_T-DM1P (90.5% vs. 47.3%) regimens offered the optimal balance between pCR and SAE. Conclusions: The T-DM1PC, T-DM1, and PTC_T-DM1P regimens had the optimal balance between efficacy and safety, while DFS was highest for the PTC regimen. However, these results were based on a small number of studies, and additional RCTs assessing the efficacy of regimens with T-DM1 are still needed to confirm these findings.
Collapse
Affiliation(s)
- Agampodi Danushi M. Gunasekara
- Mahidol University Health Technology Assessment Graduate Program (MUHTA), Mahidol University, Bangkok 10400, Thailand; (A.D.M.G.); (N.T.H.); (A.T.)
- Department of Paraclinical Sciences, Faculty of Medicine, General Sir John Kotelawala Defence University, Ratmalana, Colombo 10390, Sri Lanka
| | - Thunyarat Anothaisintawee
- Mahidol University Health Technology Assessment Graduate Program (MUHTA), Mahidol University, Bangkok 10400, Thailand; (A.D.M.G.); (N.T.H.); (A.T.)
- Department of Family Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
- Correspondence: (T.A.); (S.Y.); Tel.: +66-2-201-1406 (T.A.)
| | - Sitaporn Youngkong
- Mahidol University Health Technology Assessment Graduate Program (MUHTA), Mahidol University, Bangkok 10400, Thailand; (A.D.M.G.); (N.T.H.); (A.T.)
- Social and Administrative Pharmacy Division, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Correspondence: (T.A.); (S.Y.); Tel.: +66-2-201-1406 (T.A.)
| | - Nguyen T. Ha
- Mahidol University Health Technology Assessment Graduate Program (MUHTA), Mahidol University, Bangkok 10400, Thailand; (A.D.M.G.); (N.T.H.); (A.T.)
- School of Medicine, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Gareth J. McKay
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University, Belfast BT12 6BA, UK;
| | - John Attia
- School of Medicine and Public Health, College of Health and Wellbeing, University of Newcastle, Newcastle, NSW 2308, Australia;
| | - Ammarin Thakkinstian
- Mahidol University Health Technology Assessment Graduate Program (MUHTA), Mahidol University, Bangkok 10400, Thailand; (A.D.M.G.); (N.T.H.); (A.T.)
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
112
|
Wu X, Zhang C, Zhang H. Immune Effective Score as a Predictor of Response to Neoadjuvant Trastuzumab Therapy and a Prognostic Indicator for HER2-Positive Breast Cancer. Curr Oncol 2022; 29:283-293. [PMID: 35049700 PMCID: PMC8775173 DOI: 10.3390/curroncol29010026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Background: HER2-positive breast cancer (BC) is a highly aggressive phenotype. The role of the host immune features in predictive response to anti-HER2 therapies and prognosis in BC has already been suggested. We aimed to develop a predictive and prognostic model and examine its relevance to the clinical outcomes of patients with HER2-positive BC. Methods: Immune effective score (IES) was constructed using principal component analysis algorithms. A bioinformatic analysis using four independent cohorts (GSE66305, n = 88; GSE130786, n = 110; TCGA, n = 123; METABRIC, n = 236) established associations between IES and clinical outcomes. Results: Genes associated with neoadjuvant trastuzumab therapy response were enriched in pathways related to antitumor immune activities. IES was demonstrated to be a predictive biomarker to neoadjuvant trastuzumab therapy benefits (GSE66305: area under the curve (AUC) = 0.804; GSE130786: AUC = 0.704). In addition, IES was identified as an independent prognostic factor for overall survival (OS) in the TCGA cohort (p = 0.036, hazard ratio (HR): 0.66, 95% confidence interval (CI): 0.449–0.97) and METABRIC cohort (p = 0.037, HR: 0.9, 95% CI: 0.81–0.99). Conclusion: IES has a predictive value for response to neoadjuvant trastuzumab therapy and independent prognostic value for HER2-positive breast cancer.
Collapse
Affiliation(s)
- Xueying Wu
- Institute of Molecular Medicine, Peking University, Beijing 100871, China; (X.W.); (C.Z.)
| | - Chenyang Zhang
- Institute of Molecular Medicine, Peking University, Beijing 100871, China; (X.W.); (C.Z.)
| | - Henghui Zhang
- Biomedical Innovation Center, Beijing Shijitan Hospital, School of Oncology, Capital Medical University, Beijing 100038, China
- Correspondence:
| |
Collapse
|
113
|
Yau C, Osdoit M, van der Noordaa M, Shad S, Wei J, de Croze D, Hamy AS, Laé M, Reyal F, Sonke GS, Steenbruggen TG, van Seijen M, Wesseling J, Martín M, Del Monte-Millán M, López-Tarruella S, Boughey JC, Goetz MP, Hoskin T, Gould R, Valero V, Edge SB, Abraham JE, Bartlett JMS, Caldas C, Dunn J, Earl H, Hayward L, Hiller L, Provenzano E, Sammut SJ, Thomas JS, Cameron D, Graham A, Hall P, Mackintosh L, Fan F, Godwin AK, Schwensen K, Sharma P, DeMichele AM, Cole K, Pusztai L, Kim MO, van 't Veer LJ, Esserman LJ, Symmans WF. Residual cancer burden after neoadjuvant chemotherapy and long-term survival outcomes in breast cancer: a multicentre pooled analysis of 5161 patients. Lancet Oncol 2022; 23:149-160. [PMID: 34902335 PMCID: PMC9455620 DOI: 10.1016/s1470-2045(21)00589-1] [Citation(s) in RCA: 262] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous studies have independently validated the prognostic relevance of residual cancer burden (RCB) after neoadjuvant chemotherapy. We used results from several independent cohorts in a pooled patient-level analysis to evaluate the relationship of RCB with long-term prognosis across different phenotypic subtypes of breast cancer, to assess generalisability in a broad range of practice settings. METHODS In this pooled analysis, 12 institutes and trials in Europe and the USA were identified by personal communications with site investigators. We obtained participant-level RCB results, and data on clinical and pathological stage, tumour subtype and grade, and treatment and follow-up in November, 2019, from patients (aged ≥18 years) with primary stage I-III breast cancer treated with neoadjuvant chemotherapy followed by surgery. We assessed the association between the continuous RCB score and the primary study outcome, event-free survival, using mixed-effects Cox models with the incorporation of random RCB and cohort effects to account for between-study heterogeneity, and stratification to account for differences in baseline hazard across cancer subtypes defined by hormone receptor status and HER2 status. The association was further evaluated within each breast cancer subtype in multivariable analyses incorporating random RCB and cohort effects and adjustments for age and pretreatment clinical T category, nodal status, and tumour grade. Kaplan-Meier estimates of event-free survival at 3, 5, and 10 years were computed for each RCB class within each subtype. FINDINGS We analysed participant-level data from 5161 patients treated with neoadjuvant chemotherapy between Sept 12, 1994, and Feb 11, 2019. Median age was 49 years (IQR 20-80). 1164 event-free survival events occurred during follow-up (median follow-up 56 months [IQR 0-186]). RCB score was prognostic within each breast cancer subtype, with higher RCB score significantly associated with worse event-free survival. The univariable hazard ratio (HR) associated with one unit increase in RCB ranged from 1·55 (95% CI 1·41-1·71) for hormone receptor-positive, HER2-negative patients to 2·16 (1·79-2·61) for the hormone receptor-negative, HER2-positive group (with or without HER2-targeted therapy; p<0·0001 for all subtypes). RCB score remained prognostic for event-free survival in multivariable models adjusted for age, grade, T category, and nodal status at baseline: the adjusted HR ranged from 1·52 (1·36-1·69) in the hormone receptor-positive, HER2-negative group to 2·09 (1·73-2·53) in the hormone receptor-negative, HER2-positive group (p<0·0001 for all subtypes). INTERPRETATION RCB score and class were independently prognostic in all subtypes of breast cancer, and generalisable to multiple practice settings. Although variability in hormone receptor subtype definitions and treatment across patients are likely to affect prognostic performance, the association we observed between RCB and a patient's residual risk suggests that prospective evaluation of RCB could be considered to become part of standard pathology reporting after neoadjuvant therapy. FUNDING National Cancer Institute at the US National Institutes of Health.
Collapse
Affiliation(s)
- Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
| | - Marie Osdoit
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Surgery, Institut Curie, Paris, France
| | | | - Sonal Shad
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jane Wei
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Diane de Croze
- Department of Tumor Biology, Institut Curie, Paris, France
| | | | - Marick Laé
- Department of Tumor Biology, Institut Curie, Paris, France; Department of Pathology, Université de Rouen Normandie, Rouen, France
| | - Fabien Reyal
- Department of Surgery, Institut Curie, Paris, France
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Tessa G Steenbruggen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maartje van Seijen
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jelle Wesseling
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Miguel Martín
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Maria Del Monte-Millán
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Sara López-Tarruella
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | | | - Tanya Hoskin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Rebekah Gould
- Department of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen B Edge
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jean E Abraham
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - John M S Bartlett
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, Canada; Deanery of Molecular, Genetic and Population Health Sciences, Edinburgh Cancer Research Centre, Edinburgh, UK; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Carlos Caldas
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Janet Dunn
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Helena Earl
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Larry Hayward
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Louise Hiller
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Elena Provenzano
- Department of Histopathology, University of Cambridge, Cambridge, UK
| | | | - Jeremy S Thomas
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - David Cameron
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Ashley Graham
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Peter Hall
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Lorna Mackintosh
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Fang Fan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kelsey Schwensen
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Priyanka Sharma
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Angela M DeMichele
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly Cole
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Lajos Pusztai
- Department of Medical Oncology, Yale University, New Haven, CT, USA
| | - Mi-Ok Kim
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - W Fraser Symmans
- Department of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
114
|
Liu H, Lv L, Gao H, Cheng M. Pathologic Complete Response and Its Impact on Breast Cancer Recurrence and Patient's Survival after Neoadjuvant Therapy: A Comprehensive Meta-Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:7545091. [PMID: 35003324 PMCID: PMC8741368 DOI: 10.1155/2021/7545091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Earlier research has illustrated prognostic significance of pathologic complete response (pCR) in neoadjuvant therapy (NAT) for breast cancer, whereas correlation between treatment after achieving pCR and survival improvement remains underexplored. We attempted to measure the relation between pCR achieved after NAT and breast cancer recurrence or patient's survival. METHODS We searched PubMed, EMBASE, Web of Science, and The Cochrane Library databases to find relevant articles from their inception to November 2020. According to eligibility criteria, studies were selected and basic data were extracted. The primary endpoint was the correlation between pCR achieved after NAT and event-free survival (EFS) or overall survival (OS). The results were obtained by directly extracting specific information from the literature or estimating individual data by survival curves on DigitizeIt software, presented with HR and 95% CI. All data were processed on Stata 14.0 software. RESULTS Among 4338 articles, there were 25 eligible articles involving 8767 patients. The EFS of patients achieved pCR after NAT improved obviously (HR = 0.27; 95% CI, 0.24-0.31), especially in triple negative (HR = 0.17; 95% CI, 0.12-0.24) and HER2 positive (HR = 0.24; 95% CI, 0.20-0.30) breast cancer patients. As such, pCR after NAT was implicated in significantly increased OS (HR = 0.32; 95% CI, 0.27-0.37). CONCLUSION Achieving pCR after NAT was notably related to the improvement of EFS and OS, especially for patients with triple-negative and HER2-positive breast cancer. pCR can be a surrogate indicator for outcome of breast cancer patients after NAT, as well as a predictor of treatment efficacy after NAT. Besides, well-designed studies are still warranted for confirmation.
Collapse
Affiliation(s)
- Hui Liu
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| | - Liqiong Lv
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| | - Hui Gao
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| | - Ming Cheng
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| |
Collapse
|
115
|
Neoadjuvant Therapy with Concurrent Docetaxel, Epirubicin, and Cyclophosphamide (TEC) in High-Risk HER2-Negative Breast Cancers. Adv Ther 2021; 38:5752-5762. [PMID: 34699004 DOI: 10.1007/s12325-021-01933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Concurrent anthracycline and taxane is an effective and efficient way to deliver neoadjuvant chemotherapy for HER2-negative breast cancers. Data on efficacy and tolerance to 6 cycles of concurrent docetaxel, epirubicin, and cyclophosphamide (TEC) is limited. METHOD All patients with HER2-negative breast cancers who received neoadjuvant TEC from January 2013 to December 2019 were reviewed. RESULTS A total of 71 patients [57 luminal B disease; 14 triple negative breast cancer (TNBC)] received neoadjuvant TEC with prophylactic granulocyte colony-stimulating factor (G-CSF). The pathological complete response (pCR) rate was 26.3% and 28.6% for luminal B and TNBC, respectively. With median follow-up of 48.9 months, 3 years disease-free survival was 85.9%, and 3 years overall survival was 89.6%. Non-hematological toxicities were common but the majority was grade 1 or 2. The most common grade 3 or 4 toxicity were hematological, including neutropenia (26.8%) and anemia (15.5%). There was no cardiotoxicity observed. Half of the patients had at least one dose reduction but all patients completed the planned 6 cycles and had breast surgery done. CONCLUSION Six cycles of TEC with prophylactic G-CSF is an effective and tolerable neoadjuvant regime for HER2-negative breast cancers. Hematological toxicities were the most common toxicities. Although many patients required dose reduction, all patients completed treatment and there was no observed cardiotoxicity.
Collapse
|
116
|
Gion M, Pérez-García JM, Llombart-Cussac A, Sampayo-Cordero M, Cortés J, Malfettone A. Surrogate endpoints for early-stage breast cancer: a review of the state of the art, controversies, and future prospects. Ther Adv Med Oncol 2021; 13:17588359211059587. [PMID: 34868353 PMCID: PMC8640314 DOI: 10.1177/17588359211059587] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/25/2021] [Indexed: 01/07/2023] Open
Abstract
Drug approval for early-stage breast cancer (EBC) has been historically granted in the context of registration trials based on adequate outcomes such as disease-free survival and overall survival. Improvements in long-term outcomes have made it more difficult to demonstrate the clinical benefit of a new cancer drug in large, randomized, comparative clinical trials. Therefore, the use of surrogate endpoints rather than traditional measures allows for cancer drug trials to proceed with smaller sample sizes and shorter follow-up periods, which reduces drug development time. Among surrogate endpoints for breast cancer, the increase in pathological complete response (pCR) rates was considered appropriate for accelerated drug approval. The association between pCR and long-term outcomes was strongest in patients with aggressive tumor subtypes, such as triple-negative and human epidermal growth factor receptor 2 (HER2)-positive/hormone receptor-negative breast cancers. Whereas in hormone receptor-positive/HER2-negative EBC, the most accepted surrogate markers for endocrine therapy-based trials include changes in Ki67 and the preoperative endocrine prognostic index. Beyond the classic endpoints, further prognostic tools are required to provide EBC patients with individualized and effective therapies, and the neoadjuvant setting provides an excellent platform for drug development and biomarker discovery. Nowadays, the availability of multigene signatures is offering a standardized quantitative and reproducible tool to potentiate the efficacy of standard treatment for high-risk patients and develop de-escalated treatments for patients at lower risk of relapse. In this article, we first evaluate the surrogacies used for long-term outcomes and the underlying evidence supporting the use of each surrogate endpoint for the accelerated or regular drug approval process in EBC. Next, we provide an overview of the most recent studies and innovative strategies in a (neo)adjuvant setting as a platform to accelerate new drug approval. Finally, we highlight some clinical trials aimed at tailoring systemic treatment of EBC using prognosis-related factors or early biomarkers of drug sensitivity or resistance.
Collapse
Affiliation(s)
- María Gion
- University Hospital Ramon y Cajal, Madrid, Spain
| | - José Manuel Pérez-García
- International Breast Cancer Center (IBCC), Quironsalud Group, Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Antonio Llombart-Cussac
- Hospital Arnau de Vilanova, Valencia, Spain
- Universidad Catolica de Valencia San Vicente Martir, Valencia, Spain
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Miguel Sampayo-Cordero
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Quironsalud Group, Carrer de Vilana, 12, 08022 Barcelona, SpainVall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Andrea Malfettone
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| |
Collapse
|
117
|
Kurz D, Sánchez CS, Axenie C. Data-Driven Discovery of Mathematical and Physical Relations in Oncology Data Using Human-Understandable Machine Learning. Front Artif Intell 2021; 4:713690. [PMID: 34901835 PMCID: PMC8655230 DOI: 10.3389/frai.2021.713690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/08/2021] [Indexed: 11/19/2022] Open
Abstract
For decades, researchers have used the concepts of rate of change and differential equations to model and forecast neoplastic processes. This expressive mathematical apparatus brought significant insights in oncology by describing the unregulated proliferation and host interactions of cancer cells, as well as their response to treatments. Now, these theories have been given a new life and found new applications. With the advent of routine cancer genome sequencing and the resulting abundance of data, oncology now builds an "arsenal" of new modeling and analysis tools. Models describing the governing physical laws of tumor-host-drug interactions can be now challenged with biological data to make predictions about cancer progression. Our study joins the efforts of the mathematical and computational oncology community by introducing a novel machine learning system for data-driven discovery of mathematical and physical relations in oncology. The system utilizes computational mechanisms such as competition, cooperation, and adaptation in neural networks to simultaneously learn the statistics and the governing relations between multiple clinical data covariates. Targeting an easy adoption in clinical oncology, the solutions of our system reveal human-understandable properties and features hidden in the data. As our experiments demonstrate, our system can describe nonlinear conservation laws in cancer kinetics and growth curves, symmetries in tumor's phenotypic staging transitions, the preoperative spatial tumor distribution, and up to the nonlinear intracellular and extracellular pharmacokinetics of neoadjuvant therapies. The primary goal of our work is to enhance or improve the mechanistic understanding of cancer dynamics by exploiting heterogeneous clinical data. We demonstrate through multiple instantiations that our system is extracting an accurate human-understandable representation of the underlying dynamics of physical interactions central to typical oncology problems. Our results and evaluation demonstrate that, using simple-yet powerful-computational mechanisms, such a machine learning system can support clinical decision-making. To this end, our system is a representative tool of the field of mathematical and computational oncology and offers a bridge between the data, the modeler, the data scientist, and the practicing clinician.
Collapse
Affiliation(s)
- Daria Kurz
- Interdisziplinäres Brustzentrum, Helios Klinikum München West, Akademisches Lehrkrankenhaus der Ludwig-Maximilians Universität München, Munich, Germany
| | - Carlos Salort Sánchez
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Cristian Axenie
- Audi Konfuzius-Institut Ingolstadt Laboratory, Technische Hochschule Ingolstadt, Ingolstadt, Germany
| |
Collapse
|
118
|
Sargent RE, Vazquez E, Kang I, Lu J, Manchandia T, Sheth P, Terando A, Nelson ME, Carr A, Hong DS, Sener SF. Factors associated with relapse-free survival after neoadjuvant chemotherapy for breast cancer at a safety net medical center. Am J Surg 2021; 223:539-542. [PMID: 34801227 DOI: 10.1016/j.amjsurg.2021.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/13/2021] [Accepted: 11/14/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND This study was designed to assess prognostic factors associated with relapse-free survival (RFS) after neoadjuvant chemotherapy (NAC) for breast cancer. METHODS A single-institution retrospective analysis was performed including clinical, radiographic, and pathologic parameters for all breast cancer patients treated with NAC from 2015 to 2018. All patients had pre-and post-NAC MRI. RESULTS For 102 patients, median follow-up was 47.4 months, and the five-year RFS was 74%. The 41 (40%) patients who achieved pathologic complete response (pCR) after NAC had a significantly higher five-year RFS than the 61 not achieving pCR. For 31 patients with triple-negative cancers, the five-year RFS was significantly higher in those achieving pCR vs. no pCR. The 44 (43%) patients who achieved radiographic complete response (rCR) after NAC had similar five-year RFS to the 58 (57%) not achieving rCR. CONCLUSION pCR, node-negativity after NAC, and triple-negative subtype were prognostic factors associated with relapse-free survival after NAC.
Collapse
Affiliation(s)
- Rachel E Sargent
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Surgery and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Vazquez
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA
| | - Irene Kang
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Medicine and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Janice Lu
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Medicine and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Tejas Manchandia
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Radiology and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Pulin Sheth
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Radiology and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Alicia Terando
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Surgery and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Maria E Nelson
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Surgery and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Azadeh Carr
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Surgery and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - David S Hong
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Radiation Oncology and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Stephen F Sener
- Los Angeles County + University of Southern California (LAC+USC) Medical Center, Los Angeles, CA, USA; Department of Surgery and Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
119
|
Symmans WF, Yau C, Chen YY, Balassanian R, Klein ME, Pusztai L, Nanda R, Parker BA, Datnow B, Krings G, Wei S, Feldman MD, Duan X, Chen B, Sattar H, Khazai L, Zeck JC, Sams S, Mhawech-Fauceglia P, Rendi M, Sahoo S, Ocal IT, Fan F, LeBeau LG, Vinh T, Troxell ML, Chien AJ, Wallace AM, Forero-Torres A, Ellis E, Albain KS, Murthy RK, Boughey JC, Liu MC, Haley BB, Elias AD, Clark AS, Kemmer K, Isaacs C, Lang JE, Han HS, Edmiston K, Viscusi RK, Northfelt DW, Khan QJ, Leyland-Jones B, Venters SJ, Shad S, Matthews JB, Asare SM, Buxton M, Asare AL, Rugo HS, Schwab RB, Helsten T, Hylton NM, van 't Veer L, Perlmutter J, DeMichele AM, Yee D, Berry DA, Esserman LJ. Assessment of Residual Cancer Burden and Event-Free Survival in Neoadjuvant Treatment for High-risk Breast Cancer: An Analysis of Data From the I-SPY2 Randomized Clinical Trial. JAMA Oncol 2021; 7:1654-1663. [PMID: 34529000 DOI: 10.1001/jamaoncol.2021.3690] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Importance Residual cancer burden (RCB) distributions may improve the interpretation of efficacy in neoadjuvant breast cancer trials. Objective To compare RCB distributions between randomized control and investigational treatments within subtypes of breast cancer and explore the relationship with survival. Design, Setting, and Participants The I-SPY2 is a multicenter, platform adaptive, randomized clinical trial in the US that compares, by subtype, investigational agents in combination with chemotherapy vs chemotherapy alone in adult women with stage 2/3 breast cancer at high risk of early recurrence. Investigational treatments graduated in a prespecified subtype if there was 85% or greater predicted probability of higher rate of pathologic complete response (pCR) in a confirmatory, 300-patient, 1:1 randomized, neoadjuvant trial in that subtype. Evaluation of a secondary end point was reported from the 10 investigational agents tested in the I-SPY2 trial from March 200 through 2016, and analyzed as of September 9, 2020. The analysis plan included modeling of RCB within subtypes defined by hormone receptor (HR) and ERBB2 status and compared control treatments with investigational treatments that graduated and those that did not graduate. Interventions Neoadjuvant paclitaxel plus/minus 1 of several investigational agents for 12 weeks, then 12 weeks of cyclophosphamide/doxorubicin chemotherapy followed by surgery. Main Outcomes and Measures Residual cancer burden (pathological measure of residual disease) and event-free survival (EFS). Results A total of 938 women (mean [SD] age, 49 [11] years; 66 [7%] Asian, 103 [11%] Black, and 750 [80%] White individuals) from the first 10 investigational agents were included, with a median follow-up of 52 months (IQR, 29 months). Event-free survival worsened significantly per unit of RCB in every subtype of breast cancer (HR-positive/ERBB2-negative: hazard ratio [HZR], 1.75; 95% CI, 1.45-2.16; HR-positive/ERBB2-positive: HZR, 1.55; 95% CI, 1.18-2.05; HR-negative/ERBB2-positive: HZR, 2.39; 95% CI, 1.64-3.49; HR-negative/ERBB2-negative: HZR, 1.99; 95% CI, 1.71-2.31). Prognostic information from RCB was similar from treatments that graduated (HZR, 2.00; 95% CI, 1.57-2.55; 254 [27%]), did not graduate (HZR, 1.87; 95% CI, 1.61-2.17; 486 [52%]), or were control (HZR, 1.79; 95% CI, 1.42-2.26; 198 [21%]). Investigational treatments significantly lowered RCB in HR-negative/ERBB2-negative (graduated and nongraduated treatments) and ERBB2-positive subtypes (graduated treatments), with improved EFS (HZR, 0.61; 95% CI, 0.41-0.93) in the exploratory analysis. Conclusions and Relevance In this randomized clinical trial, the prognostic significance of RCB was consistent regardless of subtype and treatment. Effective neoadjuvant treatments shifted the distribution of RCB in addition to increasing pCR rate and appeared to improve EFS. Using a standardized quantitative method to measure response advances the interpretation of efficacy. Trial Registration ClinicalTrials.gov Identifier: NCT01042379.
Collapse
Affiliation(s)
- W Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Christina Yau
- Department of Surgery, University of California, San Francisco
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco
| | - Ron Balassanian
- Department of Pathology, University of California, San Francisco
| | - Molly E Klein
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Lajos Pusztai
- Department of Medicine, Medical Oncology, Yale University, New Haven, Connecticut
| | - Rita Nanda
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Barbara A Parker
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Brian Datnow
- Department of Pathology, University of California, San Diego, La Jolla
| | - Gregor Krings
- Department of Pathology, University of California, San Francisco
| | - Shi Wei
- Department of Anatomic Pathology, University of Alabama at Birmingham
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia
| | - Xiuzhen Duan
- Department of Pathology, Loyola University, Chicago, Illinois
| | - Beiyun Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Husain Sattar
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Laila Khazai
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Jay C Zeck
- Department of Pathology, Georgetown University, Washington, DC
| | - Sharon Sams
- Department of Pathology, University of Colorado Anschutz Medical Center, Aurora
| | | | - Mara Rendi
- Department of Anatomic Pathology, University of Washington, Seattle
| | - Sunati Sahoo
- Department of Pathology, University of Texas Southwestern, Dallas
| | - Idris Tolgay Ocal
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona
| | - Fang Fan
- Department of Pathology, University of Kansas Medical Center, Kansas City
| | | | - Tuyethoa Vinh
- Department of Pathology, Inova Health System, Fairfax, Virginia
| | - Megan L Troxell
- Department of Pathology, Oregon Health and Science University, Portland
| | - A Jo Chien
- Division of Hematology-Oncology, Department of Medicine, University of California, San Francisco
| | - Anne M Wallace
- Department of Surgery, University of California, San Diego, La Jolla
| | - Andres Forero-Torres
- Division of Hematology-Oncology, Department of Medicine, University of Alabama at Birmingham
| | - Erin Ellis
- Medical Oncology, Swedish Cancer Institute, Seattle, Washington
| | - Kathy S Albain
- Division of Hematology-Oncology, Department of Medicine, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Rashmi K Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Minetta C Liu
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Barbara B Haley
- Division of Hematology-Oncology, Department of Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Anthony D Elias
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora
| | - Amy S Clark
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia
| | - Kathleen Kemmer
- Division of Hematology-Oncology, Department of Medicine, Oregon Health & Science University, Portland
| | - Claudine Isaacs
- Division of Hematology-Oncology, Department of Medicine, Georgetown University, Washington, DC
| | - Julie E Lang
- Department of Surgery, University of Southern California, Los Angeles
| | - Hyo S Han
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Kirsten Edmiston
- Department of Surgery, Inova Schar Cancer Institute, Fairfax, Virginia
| | - Rebecca K Viscusi
- Department of Surgery, University of Arizona Health Sciences, Tucson, Arizona
| | - Donald W Northfelt
- Department of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Qamar J Khan
- Division of Oncology, Department of Medicine, University of Kansas, Kansas City
| | | | - Sara J Venters
- Department of Laboratory Medicine, University of California, San Francisco
| | - Sonal Shad
- Department of Surgery, University of California, San Francisco
| | | | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | - Adam L Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | - Hope S Rugo
- Division of Hematology-Oncology, Department of Medicine, University of California, San Francisco
| | - Richard B Schwab
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Teresa Helsten
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Laura van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco
| | | | - Angela M DeMichele
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia
| | - Douglas Yee
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis
| | | | | |
Collapse
|
120
|
de Freitas AJA, Causin RL, Varuzza MB, Hidalgo Filho CMT, da Silva VD, Souza CDP, Marques MMC. Molecular Biomarkers Predict Pathological Complete Response of Neoadjuvant Chemotherapy in Breast Cancer Patients: Review. Cancers (Basel) 2021; 13:cancers13215477. [PMID: 34771640 PMCID: PMC8582511 DOI: 10.3390/cancers13215477] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Breast cancer is the most common cancer in women worldwide. Although many studies have aimed to understand the genetic basis of breast cancer, leading to increasingly accurate diagnoses, only a few molecular biomarkers are used in clinical practice to predict response to therapy. Current studies aim to develop more personalized therapies to decrease the adverse effects of chemotherapy. Personalized medicine not only requires clinical, but also molecular characterization of tumors, which allows the use of more effective drugs for each patient. The aim of this study was to identify potential molecular biomarkers that can predict the response to therapy after neoadjuvant chemotherapy in patients with breast cancer. In this review, we summarize genomic, transcriptomic, and proteomic biomarkers that can help predict the response to therapy. Abstract Neoadjuvant chemotherapy (NAC) is often used to treat locally advanced disease for tumor downstaging, thus improving the chances of breast-conserving surgery. From the NAC response, it is possible to obtain prognostic information as patients may reach a pathological complete response (pCR). Those who do might have significant advantages in terms of survival rates. Breast cancer (BC) is a heterogeneous disease that requires personalized treatment strategies. The development of targeted therapies depends on identifying biomarkers that can be used to assess treatment efficacy as well as the discovery of new and more accurate therapeutic agents. With the development of new “OMICS” technologies, i.e., genomics, transcriptomics, and proteomics, among others, the discovery of new biomarkers is increasingly being used in the context of clinical practice, bringing us closer to personalized management of BC treatment. The aim of this review is to compile the main biomarkers that predict pCR in BC after NAC.
Collapse
Affiliation(s)
- Ana Julia Aguiar de Freitas
- Molecular Oncology Research Center, Barretos Cancer Hospital, Teaching and Research Institute, Barretos 14784-400, SP, Brazil; (A.J.A.d.F.); (R.L.C.); (M.B.V.)
| | - Rhafaela Lima Causin
- Molecular Oncology Research Center, Barretos Cancer Hospital, Teaching and Research Institute, Barretos 14784-400, SP, Brazil; (A.J.A.d.F.); (R.L.C.); (M.B.V.)
| | - Muriele Bertagna Varuzza
- Molecular Oncology Research Center, Barretos Cancer Hospital, Teaching and Research Institute, Barretos 14784-400, SP, Brazil; (A.J.A.d.F.); (R.L.C.); (M.B.V.)
| | | | | | | | - Márcia Maria Chiquitelli Marques
- Molecular Oncology Research Center, Barretos Cancer Hospital, Teaching and Research Institute, Barretos 14784-400, SP, Brazil; (A.J.A.d.F.); (R.L.C.); (M.B.V.)
- Barretos School of Health Sciences, Dr. Paulo Prata–FACISB, Barretos 14785-002, SP, Brazil
- Correspondence: ; Tel.: +55-17-3321-6600 (ext. 7057)
| |
Collapse
|
121
|
Thompson BM, Chala LF, Shimizu C, Mano MS, Filassi JR, Geyer FC, Torres US, de Mello GGN, da Costa Leite C. Pre-treatment MRI tumor features and post-treatment mammographic findings: may they contribute to refining the prediction of pathologic complete response in post-neoadjuvant breast cancer patients with radiologic complete response on MRI? Eur Radiol 2021; 32:1663-1675. [PMID: 34716780 DOI: 10.1007/s00330-021-08290-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/05/2021] [Accepted: 08/20/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Radiologic complete response (rCR) in breast cancer patients after neoadjuvant chemotherapy (NAC) does not necessarily correlate with pathologic complete response (pCR), a marker traditionally associated with better outcomes. We sought to verify if data extracted from two important steps of the imaging workup (tumor features at pre-treatment MRI and post-treatment mammographic findings) might assist in refining the prediction of pCR in post-NAC patients showing rCR. METHODS A total of 115 post-NAC women with rCR on MRI (2010-2016) were retrospectively assessed. Pre-treatment MRI (lesion morphology, size, and distribution) and post-treatment mammographic findings (calcification, asymmetry, mass, architectural distortion) were assessed, as well as clinical and molecular variables. Bivariate and multivariate analyses evaluated correlation between such variables and pCR. Post-NAC mammographic findings and their correlation with ductal in situ carcinoma (DCIS) were evaluated using Pearson's correlation. RESULTS Tumor distribution at pre-treatment MRI was the only significant predictive imaging feature on multivariate analysis, with multicentric lesions having lower odds of pCR (p = 0.035). There was no significant association between tumor size and morphology with pCR. Mammographic residual calcifications were associated with DCIS (p = 0.009). The receptor subtype remained as a significant predictor, with HR-HER2 + and triple-negative status demonstrating higher odds of pCR on multivariate analyses. CONCLUSIONS Multicentric lesions on pre-NAC MRI were associated with a lower chance of pCR in post-NAC rCR patients. The receptor subtype remained a reliable predictor of pCR. Residual mammographic calcifications correlated with higher odds of malignancy, making the correlation between mammography and MRI essential for surgical planning. Key Points • The presence of a multicentric lesion on pre-NAC MRI, even though the patient reaches a radiologic complete response on MRI, is associated with a lower chance of pCR. • Molecular status of the tumor remained the only significant predictor of pathologic complete response in such patients in the present study. • Post-neoadjuvant residual calcifications found on mammography were related to higher odds of residual malignancy, making the correlation between mammography and MRI essential for surgical planning.
Collapse
Affiliation(s)
- Bruna M Thompson
- Institute of Radiology, Clinics Hospital, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Luciano F Chala
- Fleury Group, Rua Cincinato Braga, 282, Bela Vista, São Paulo, SP, 01333-010, Brazil
| | - Carlos Shimizu
- Institute of Radiology, Clinics Hospital, School of Medicine, University of São Paulo, São Paulo, Brazil.,Fleury Group, Rua Cincinato Braga, 282, Bela Vista, São Paulo, SP, 01333-010, Brazil
| | - Max S Mano
- Department of Oncology, Hospital Sírio Libanês, São Paulo, Brazil
| | - José R Filassi
- Department of Gynecology and Obstetrics, Mastology Section, Instituto Do Câncer Do Estado de São Paulo, São Paulo, Brazil
| | - Felipe C Geyer
- Department of Pathology, Instituto Do Câncer Do Estado de São Paulo, São Paulo, Brazil
| | - Ulysses S Torres
- Fleury Group, Rua Cincinato Braga, 282, Bela Vista, São Paulo, SP, 01333-010, Brazil.
| | | | - Cláudia da Costa Leite
- Institute of Radiology, Clinics Hospital, School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
122
|
Zhao F, Huo X, Wang M, Liu Z, Zhao Y, Ren D, Xie Q, Liu Z, Li Z, Du F, Shen G, Zhao J. Comparing Biomarkers for Predicting Pathological Responses to Neoadjuvant Therapy in HER2-Positive Breast Cancer: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:731148. [PMID: 34778044 PMCID: PMC8581664 DOI: 10.3389/fonc.2021.731148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/08/2021] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION The predictive strength and accuracy of some biomarkers for the pathological complete response (pCR) to neoadjuvant therapy for HER2-positive breast cancer remain unclear. This study aimed to compare the accuracy of the HER2-enriched subtype and the presence of PIK3CA mutations, namely, TILs, HRs, and Ki-67, in predicting the pCR to HER2-positive breast cancer therapy. METHODS We screened studies that included pCR predicted by one of the following biomarkers: the HER2-enriched subtype and the presence of PIK3CA mutations, TILs, HRs, or Ki-67. We then calculated the pooled sensitivity, specificity, positive and negative predictive values (PPVs and NPVs, respectively), and positive and negative likelihood ratios (LRs). Summary receiver operating characteristic (SROC) curves and areas under the curve (AUCs) were used to estimate the diagnostic accuracy. RESULTS The pooled estimates of sensitivity and specificity for the HER2-enriched subtype and the presence of PIK3CA mutations, namely, TILs, HRs, and Ki-67, were 0.66 and 0.62, 0.85 and 0.27, 0.49 and 0.61, 0.54 and 0.64, and 0.68 and 0.51, respectively. The AUC of the HER2-enriched subtype was significantly higher (0.71) than those for the presence of TILs (0.59, p = 0.003), HRs (0.65, p = 0.003), and Ki-67 (0.62, p = 0.005). The AUC of the HER2-enriched subtype had a tendency to be higher than that of the presence of PIK3CA mutations (0.58, p = 0.220). Moreover, it had relatively high PPV (0.58) and LR+ (1.77), similar NPV (0.73), and low LR- (0.54) compared with the other four biomarkers. CONCLUSIONS The HER2-enriched subtype has a moderate breast cancer diagnostic accuracy, which is better than those of the presence of PIK3CA mutations, TILs, HRs, and Ki-67.
Collapse
Affiliation(s)
- Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Xingfa Huo
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Miaozhou Wang
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Zhen Liu
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Yi Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Qiqi Xie
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Zhilin Liu
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Zitao Li
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Feng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The VIPII Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| |
Collapse
|
123
|
Esserman L, Eklund M, Veer LV, Shieh Y, Tice J, Ziv E, Blanco A, Kaplan C, Hiatt R, Fiscalini AS, Yau C, Scheuner M, Naeim A, Wenger N, Lee V, Heditsian D, Brain S, Parker BA, LaCroix AZ, Madlensky L, Hogarth M, Borowsky A, Anton-Culver H, Kaster A, Olopade OI, Sheth D, Garcia A, Lancaster R, Plaza M. The WISDOM study: a new approach to screening can and should be tested. Breast Cancer Res Treat 2021; 189:593-598. [PMID: 34529196 DOI: 10.1007/s10549-021-06346-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/28/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Laura Esserman
- University of California, San Francisco, CA, 94158, USA.
| | | | | | - Yiwey Shieh
- University of California, San Francisco, CA, 94158, USA
| | - Jeffrey Tice
- University of California, San Francisco, CA, 94158, USA
| | - Elad Ziv
- University of California, San Francisco, CA, 94158, USA
| | - Amie Blanco
- University of California, San Francisco, CA, 94158, USA
| | - Celia Kaplan
- University of California, San Francisco, CA, 94158, USA
| | - Robert Hiatt
- University of California, San Francisco, CA, 94158, USA
| | | | - Christina Yau
- University of California, San Francisco, CA, 94158, USA
| | | | - Arash Naeim
- University of California, Los Angeles, CA, 90095, USA
| | - Neil Wenger
- University of California, Los Angeles, CA, 90095, USA
| | - Vivian Lee
- University of California, San Francisco, CA, 94158, USA
| | | | - Susie Brain
- University of California, San Francisco, CA, 94158, USA
| | | | | | | | | | | | | | | | | | - Deepa Sheth
- University of Chicago, Chicago, IL, 60637, USA
| | | | | | | |
Collapse
|
124
|
Mayer IA, Zhao F, Arteaga CL, Symmans WF, Park BH, Burnette BL, Tevaarwerk AJ, Garcia SF, Smith KL, Makower DF, Block M, Morley KA, Jani CR, Mescher C, Dewani SJ, Tawfik B, Flaum LE, Mayer EL, Sikov WM, Rodler ET, Wagner LI, DeMichele AM, Sparano JA, Wolff AC, Miller KD. Randomized Phase III Postoperative Trial of Platinum-Based Chemotherapy Versus Capecitabine in Patients With Residual Triple-Negative Breast Cancer Following Neoadjuvant Chemotherapy: ECOG-ACRIN EA1131. J Clin Oncol 2021; 39:2539-2551. [PMID: 34092112 PMCID: PMC8577688 DOI: 10.1200/jco.21.00976] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/08/2021] [Accepted: 05/09/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Patients with triple-negative breast cancer (TNBC) and residual invasive disease (RD) after completion of neoadjuvant chemotherapy (NAC) have a high-risk for recurrence, which is reduced by adjuvant capecitabine. Preclinical models support the use of platinum agents in the TNBC basal subtype. The EA1131 trial hypothesized that invasive disease-free survival (iDFS) would not be inferior but improved in patients with basal subtype TNBC treated with adjuvant platinum compared with capecitabine. PATIENTS AND METHODS Patients with clinical stage II or III TNBC with ≥ 1 cm RD in the breast post-NAC were randomly assigned to receive platinum (carboplatin or cisplatin) once every 3 weeks for four cycles or capecitabine 14 out of 21 days every 3 weeks for six cycles. TNBC subtype (basal v nonbasal) was determined by PAM50 in the residual disease. A noninferiority design with superiority alternative was chosen, assuming a 4-year iDFS of 67% with capecitabine. RESULTS Four hundred ten of planned 775 participants were randomly assigned to platinum or capecitabine between 2015 and 2021. After median follow-up of 20 months and 120 iDFS events (61% of full information) in the 308 (78%) patients with basal subtype TNBC, the 3-year iDFS for platinum was 42% (95% CI, 30 to 53) versus 49% (95% CI, 39 to 59) for capecitabine. Grade 3 and 4 toxicities were more common with platinum agents. The Data and Safety Monitoring Committee recommended stopping the trial as it was unlikely that further follow-up would show noninferiority or superiority of platinum. CONCLUSION Platinum agents do not improve outcomes in patients with basal subtype TNBC RD post-NAC and are associated with more severe toxicity when compared with capecitabine. Participants had a lower than expected 3-year iDFS regardless of study treatment, highlighting the need for better therapies in this high-risk population.
Collapse
Affiliation(s)
- Ingrid A. Mayer
- Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Fengmin Zhao
- Dana-Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, MA
| | | | | | - Ben H. Park
- Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Brian L. Burnette
- Cancer Research of Wisconsin and Northern Michigan (CROWN) NCORP, Green Bay, WI
| | | | | | - Karen L. Smith
- Johns Hopkins University, Sidney Kimmel Cancer Center, Baltimore, MD
| | | | | | | | | | - Craig Mescher
- Metro-Minnesota Community Oncology Research Consortium, St Louis Park, MN
| | | | | | | | | | | | | | | | | | | | - Antonio C. Wolff
- Johns Hopkins University, Sidney Kimmel Cancer Center, Baltimore, MD
| | - Kathy D. Miller
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN
| |
Collapse
|
125
|
Foldi J, Rozenblit M, Park TS, Knowlton CA, Golshan M, Moran M, Pusztai L. Optimal Management for Residual Disease Following Neoadjuvant Systemic Therapy. Curr Treat Options Oncol 2021; 22:79. [PMID: 34213636 DOI: 10.1007/s11864-021-00879-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 11/26/2022]
Abstract
OPINION STATEMENT Treatment sequencing in early-stage breast cancer has significantly evolved in recent years, particularly in the triple negative (TNBC) and human epidermal growth factor receptor 2 (HER2)-positive subsets. Instead of surgery first followed by chemotherapy, several clinical trials showed benefits to administering systemic chemotherapy (and HER2-targeted therapies) prior to surgery. These benefits include more accurate prognostic estimates based on the extent of residual cancer that can also guide adjuvant treatment, and frequent tumor downstaging that can lead to smaller surgeries in patients with large tumors at diagnosis. Patients with extensive invasive residual cancer after neoadjuvant therapy are at high risk for disease recurrence, and two pivotal clinical trials, CREATE-X and KATHERINE, demonstrated improved recurrence free survival with adjuvant capecitabine and ado-trastuzumab-emtansine (T-DM1) in TNBC and HER2-positive residual cancers, respectively. Patients who achieve pathologic complete response (pCR) have excellent long-term disease-free survival regardless of what chemotherapy regimen induced this favorable response. This allows escalation or de-escalation of adjuvant therapy: patients who achieved pCR could be spared further chemotherapy, while those with residual cancer could receive additional chemotherapy postoperatively. Ongoing clinical trials are testing this strategy (CompassHER2-pCR: NCT04266249). pCR also provides an opportunity to assess de-escalation of locoregional therapies. Currently, for patients with residual disease in the lymph nodes (ypN+), radiation therapy entails coverage of the undissected axilla, and may include supra/infraclavicular/internal mammary nodes in addition to the whole breast or chest wall, depending on the type of surgery. Ongoing trials are testing the safety of omitting post-mastectomy breast and post-lumpectomy nodal irradiation (NCT01872975) as well as omitting axillary lymph node dissection (NCT01901094) in the setting of pCR. Additionally, evolving technologies such as minimal residual disease (MRD) monitoring in the blood during follow-up may allow early intervention with "second-line systemic adjuvant therapy" for patients with molecular relapse which might prevent impending clinical relapse.
Collapse
Affiliation(s)
- Julia Foldi
- Section of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Mariya Rozenblit
- Section of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Tristen S Park
- Department of Surgery, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Christin A Knowlton
- Department of Therapeutic Radiation, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Mehra Golshan
- Department of Surgery, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Meena Moran
- Department of Therapeutic Radiation, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Lajos Pusztai
- Section of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, 300 George St, Suite 120, Rm 133, New Haven, CT, 06520, USA.
| |
Collapse
|
126
|
Saini KS, Punie K, Twelves C, Bortini S, de Azambuja E, Anderson S, Criscitiello C, Awada A, Loi S. Antibody-drug conjugates, immune-checkpoint inhibitors, and their combination in breast cancer therapeutics. Expert Opin Biol Ther 2021; 21:945-962. [PMID: 34043927 DOI: 10.1080/14712598.2021.1936494] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Advanced breast cancer (aBC) remains incurable and the quest for more effective systemic anticancer agents continues. Promising results have led to the FDA approval of three antibody-drug conjugates (ADCs) and two immune checkpoint inhibitors (ICIs) to date for patients with aBC. AREAS COVERED With the anticipated emergence of newer ADCs and ICIs for patients with several subtypes of breast cancer, and given their potential synergy, their use in combination is of clinical interest. In this article, we review the use of ADCs and ICIs in patients with breast cancer, assess the scientific rationale for their combination, and provide an overview of ongoing trials and some early efficacy and safety results of such dual therapy. EXPERT OPINION Improvement in the medicinal chemistry of next-generation ADCs, their rational combination with ICIs and other agents, and the development of multiparametric immune biomarkers could help to significantly improve the outlook for patients with refractory aBC.
Collapse
Affiliation(s)
- Kamal S Saini
- Clinical Development Services, Covance Inc, Princeton, NJ, USA
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Chris Twelves
- Leeds Institute of Medical Research, University of Leeds and Leeds Teaching Hospitals Trust, Leeds, UK
| | | | - Evandro de Azambuja
- Medical Support Team (Academic Promoting Team), Institut Jules Bordet, Brussels, Belgium.,Faculté de Médecine, Université Libre De Bruxelles (U.L.B.), Brussels, Belgium
| | - Steven Anderson
- Clinical Development Services, Covance Inc, Princeton, NJ, USA
| | - Carmen Criscitiello
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Ahmad Awada
- Medical Support Team (Academic Promoting Team), Institut Jules Bordet, Brussels, Belgium
| | - Sherene Loi
- Division of Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
127
|
Potter DA, Thomas A, Rugo HS. A Neoadjuvant Chemotherapy Trial for Early Breast Cancer is Impacted by COVID-19: Addressing Vaccination and Cancer Trials Through Education, Equity, and Outcomes. Clin Cancer Res 2021; 27:4486-4490. [PMID: 34108186 DOI: 10.1158/1078-0432.ccr-21-1133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/14/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022]
Abstract
While COVID-19 vaccine distribution has addressed vulnerabilities related to age and comorbidities, there is a need to ensure vaccination of patients with cancer receiving experimental and routine treatment, where interruption of treatment by infection is likely to result in inferior outcomes. Among patients with cancer, those undergoing neoadjuvant chemotherapy (NAC) or adjuvant chemotherapy (Adj chemo) for early breast cancer (EBC) are at particularly high risk for inferior outcomes, in part, because optimal timing of chemotherapy is essential for promoting distant disease-free survival. COVID-19 data from the ongoing multicenter I-SPY 2 trial of NAC for EBC provides a window into the magnitude of the problem of treatment interruption, not only for the trial itself but also for routine Adj chemo. In the I-SPY 2 trial, 4.5% of patients had disruption of therapy by COVID-19, prior to wide vaccine availability, suggesting that nationally up to 5,700 patients with EBC were at risk for adverse outcomes from COVID-19 infection in 2020. To address this problem, vaccine education and public engagement are essential to overcome hesitancy, while equity of distribution is needed to address access. To accomplish these goals, healthcare organizations (HCO) need to not only call out disinformation but also engage the public with vaccine education and find common ground for vaccine acceptance, while partnering with state/local governments to improve efficiency of vaccine distribution. These approaches are important to improve trial access and to reduce susceptibility to COVID-19, as the pandemic could continue to impact access to clinical trials and routine cancer treatment.
Collapse
Affiliation(s)
- David A Potter
- University of Minnesota Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, Minneapolis, Minnesota.
| | - Alexandra Thomas
- Wake Forest Baptist Health Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, California
| |
Collapse
|
128
|
Kikano EG, Avril S, Marshall H, Jones RS, Montero AJ, Avril N. PET/CT Variants and Pitfalls in Breast Cancers. Semin Nucl Med 2021; 51:474-484. [PMID: 34088473 DOI: 10.1053/j.semnuclmed.2021.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There are a number of normal variants and pitfalls which are important to consider when evaluating F-18 Fluorodeoxyglucose (FDG) with Positron Emission Tomography (PET) in breast cancer patients. Although FDG-PET is not indicated for the initial diagnosis of breast cancer, focally increased glucose metabolism within breast tissue represents a high likelihood for a neoplastic process and requires further evaluation. Focally increased glucose metabolism is not unique to breast cancer. Other malignancies such as lymphoma, metastases from solid tumors as well as inflammatory changes also may demonstrate increased glucose metabolism either within the breast or at other sites throughout the body. Importantly, benign breast disease may also exhibit increased glucose metabolism, limiting the specificity of FDG-PET. Breast cancer has a wide range of metabolic activity attributed to tumor heterogeneity and breast cancer subtype. Intracellular signaling pathways regulating tumor glucose utilization contribute to these pitfalls of PET/CT in breast cancer. The evaluation of axillary lymph nodes by FDG-PET is less accurate than sentinel lymph node procedure, however is very accurate in identifying level II and III axillary lymph node metastases or retropectoral metastases. It is important to note that non-malignant inflammation in lymph nodes are often detected by modern PET/CT technology. Therefore, particular consideration should be given to recent vaccinations, particularly to COVID-19, which can commonly result in increased metabolic activity of axillary nodes. Whole body FDG-PET for staging of breast cancer requires specific attention to physiologic variants of FDG distribution and a careful comparison with co-registered anatomical imaging. The most important pitfalls are related to inflammatory changes including sarcoidosis, sarcoid like reactions, and other granulomatous diseases as well as secondary neoplastic processes.
Collapse
Affiliation(s)
- Elias George Kikano
- Department of Radiology, Division of Nuclear Medicine, Cleveland, Ohio; University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, Ohio
| | - Stefanie Avril
- Department of Pathology, Cleveland, Ohio; University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, Ohio
| | - Holly Marshall
- Department of Radiology, Division of Breast Imaging, Cleveland, Ohio; University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, Ohio
| | - Robert Stanley Jones
- Department of Radiology, Division of Nuclear Medicine, Cleveland, Ohio; University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, Ohio
| | - Alberto J Montero
- Department of Medicine, Solid Tumor Oncology, Cleveland, Ohio; University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, Ohio
| | - Norbert Avril
- Department of Radiology, Division of Nuclear Medicine, Cleveland, Ohio; University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
129
|
Magbanua MJM, Li W, Wolf DM, Yau C, Hirst GL, Swigart LB, Newitt DC, Gibbs J, Delson AL, Kalashnikova E, Aleshin A, Zimmermann B, Chien AJ, Tripathy D, Esserman L, Hylton N, van 't Veer L. Circulating tumor DNA and magnetic resonance imaging to predict neoadjuvant chemotherapy response and recurrence risk. NPJ Breast Cancer 2021; 7:32. [PMID: 33767190 PMCID: PMC7994408 DOI: 10.1038/s41523-021-00239-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
We investigated whether serial measurements of circulating tumor DNA (ctDNA) and functional tumor volume (FTV) by magnetic resonance imaging (MRI) can be combined to improve prediction of pathologic complete response (pCR) and estimation of recurrence risk in early breast cancer patients treated with neoadjuvant chemotherapy (NAC). We examined correlations between ctDNA and FTV, evaluated the additive value of ctDNA to FTV-based predictors of pCR using area under the curve (AUC) analysis, and analyzed the impact of FTV and ctDNA on distant recurrence-free survival (DRFS) using Cox regressions. The levels of ctDNA (mean tumor molecules/mL plasma) were significantly correlated with FTV at all time points (p < 0.05). Median FTV in ctDNA-positive patients was significantly higher compared to those who were ctDNA-negative (p < 0.05). FTV and ctDNA trajectories in individual patients showed a general decrease during NAC. Exploratory analysis showed that adding ctDNA information early during treatment to FTV-based predictors resulted in numerical but not statistically significant improvements in performance for pCR prediction (e.g., AUC 0.59 vs. 0.69, p = 0.25). In contrast, ctDNA-positivity after NAC provided significant additive value to FTV in identifying patients with increased risk of metastatic recurrence and death (p = 0.004). In this pilot study, we demonstrate that ctDNA and FTV were correlated measures of tumor burden. Our preliminary findings based on a limited cohort suggest that ctDNA at surgery improves FTV as a predictor of metastatic recurrence and death. Validation in larger studies is warranted.
Collapse
Affiliation(s)
- Mark Jesus M Magbanua
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Wen Li
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Denise M Wolf
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christina Yau
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gillian L Hirst
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lamorna Brown Swigart
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David C Newitt
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Jessica Gibbs
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Amy L Delson
- Breast Science Advocacy Core, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - A Jo Chien
- Division of Hematology Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura Esserman
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Nola Hylton
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Laura van 't Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
130
|
Abstract
PURPOSE OF REVIEW Platinum compounds are used in the treatment of various types of cancer. Here, we review the current role of cisplatin and carboplatin in the treatment of early stage and advanced triple-negative breast cancer (TNBC), and the use of biomarkers in predicting response to platinum therapy. RECENT FINDINGS Addition of carboplatin to a neoadjuvant chemotherapy regimen can result in improvement in the pathological complete response rates. The long-term benefit of the addition of carboplatin to standard chemotherapy regimens remains unproven. Single-agent platinum is an option in the treatment of advanced breast cancer. BRCA1/2 mutations predicted benefit from platinums in advanced, but not early stage breast cancer. There are yet no biomarkers to predict response to platinum in sporadic TNBC. Platinum compounds are an option in the treatment of TNBC. Identification of biomarkers to select tumors most likely to derive benefit from these agents is still needed. Ongoing trials are exploring the role of platinum in the adjuvant setting and in combination with other agents, including immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Filipa Lynce
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave YC-1275, Boston, MA, 02215, USA.
| | - Raquel Nunes
- Medical Oncology, Johns Hopkins Sidney Kimmel Cancer Center at Sibley Memorial Hospital, Building B, First Floor, 5255 Loughboro Rd, NW, Washington, DC, 20016, USA
| |
Collapse
|
131
|
Artzy-Randrup Y, Epstein T, Brown JS, Costa RLB, Czerniecki BJ, Gatenby RA. Novel evolutionary dynamics of small populations in breast cancer adjuvant and neoadjuvant therapy. NPJ Breast Cancer 2021; 7:26. [PMID: 33707440 PMCID: PMC7952601 DOI: 10.1038/s41523-021-00230-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/15/2021] [Indexed: 12/30/2022] Open
Abstract
Disseminated cancer cells (DCCs) are detected in the circulation and bone marrow of up to 40% of breast cancer (BC) patients with clinically localized disease. The formation of metastases is governed by eco-evolutionary interactions of DCCs with the tissue during the transition from microscopic populations to macroscopic disease. Here, we view BC adjuvant and neoadjuvant treatments in the context of small population extinction dynamics observed in the Anthropocene era. Specifically, the unique eco-evolutionary dynamics of small asexually reproducing cancer populations render them highly vulnerable to: (1) environmental and demographic fluctuations, (2) Allee effects, (3) genetic drift and (4) population fragmentation. Furthermore, these typically interact, producing self-reinforcing, destructive dynamics—termed the Extinction Vortex—eradicating the population even when none of the perturbations is individually capable of causing extinction. We propose that developing BC adjuvant and neoadjuvant protocols may exploit these dynamics to prevent recovery and proliferation of small cancer populations during and after treatment—termed “Eco-evolutionary rescue” in natural extinctions. We hypothesize more strategic application of currently available agents based on the extinction vulnerabilities of small populations could improve clinical outcomes.
Collapse
Affiliation(s)
- Yael Artzy-Randrup
- Department of Theoretical and Computational Ecology, IBED, University of Amsterdam, Amsterdam, The Netherlands.,Institute of Advanced Study, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamir Epstein
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Joel S Brown
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ricardo L B Costa
- Breast Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Brian J Czerniecki
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Breast Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert A Gatenby
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA. .,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA. .,Diagnostic Imaging Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
132
|
Du L, Yau C, Brown-Swigart L, Gould R, Krings G, Hirst GL, Bedrosian I, Layman RM, Carter JM, Klein M, Venters S, Shad S, van der Noordaa M, Chien AJ, Haddad T, Isaacs C, Pusztai L, Albain K, Nanda R, Tripathy D, Liu MC, Boughey J, Schwab R, Hylton N, DeMichele A, Perlmutter J, Yee D, Berry D, Van't Veer L, Valero V, Esserman LJ, Symmans WF. Predicted sensitivity to endocrine therapy for stage II-III hormone receptor-positive and HER2-negative (HR+/HER2-) breast cancer before chemo-endocrine therapy. Ann Oncol 2021; 32:642-651. [PMID: 33617937 DOI: 10.1016/j.annonc.2021.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/07/2021] [Accepted: 02/13/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND We proposed that a test for sensitivity to the adjuvant endocrine therapy component of treatment for patients with stage II-III breast cancer (SET2,3) should measure transcription related to estrogen and progesterone receptors (SETER/PR index) adjusted for a baseline prognostic index (BPI) combining clinical tumor and nodal stage with molecular subtype by RNA4 (ESR1, PGR, ERBB2, and AURKA). PATIENTS AND METHODS Patients with clinically high-risk, hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)-negative (HR+/HER2-) breast cancer received neoadjuvant taxane-anthracycline chemotherapy, surgery with measurement of residual cancer burden (RCB), and then adjuvant endocrine therapy. SET2,3 was measured from pre-treatment tumor biopsies, evaluated first in an MD Anderson Cancer Center (MDACC) cohort (n = 307, 11 years' follow-up, U133A microarrays), cut point was determined, and then independent, blinded evaluation was carried out in the I-SPY2 trial (n = 268, high-risk MammaPrint result, 3.8 years' follow-up, Agilent-44K microarrays, NCI Clinical Trials ID: NCT01042379). Primary outcome measure was distant relapse-free survival. Multivariate Cox regression models tested prognostic independence of SET2,3 relative to RCB and other molecular prognostic signatures, and whether other prognostic signatures could substitute for SETER/PR or RNA4 components of SET2,3. RESULTS SET2,3 added independent prognostic information to RCB in the MDACC cohort: SET2,3 [hazard ratio (HR) 0.23, P = 0.004] and RCB (HR 1.77, P < 0.001); and the I-SPY2 trial: SET2,3 (HR 0.27, P = 0.031) and RCB (HR 1.68, P = 0.008). SET2,3 provided similar prognostic information irrespective of whether RCB-II or RCB-III after chemotherapy, and in both luminal subtypes. Conversely, RCB was most strongly prognostic in cancers with low SET2,3 status (MDACC P < 0.001, I-SPY2 P < 0.001). Other molecular signatures were not independently prognostic; they could effectively substitute for RNA4 subtype within the BPI component of SET2,3, but they could not effectively substitute for SETER/PR index. CONCLUSIONS SET2,3 added independent prognostic information to chemotherapy response (RCB) and baseline prognostic score or subtype. Approximately 40% of patients with clinically high-risk HR+/HER2- disease had high SET2,3 and could be considered for clinical trials of neoadjuvant endocrine-based treatment.
Collapse
Affiliation(s)
- L Du
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C Yau
- Department of Surgery, University of California, San Francisco, USA
| | - L Brown-Swigart
- Department of Pathology, University of California, San Francisco, USA
| | - R Gould
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - G Krings
- Department of Pathology, University of California, San Francisco, USA
| | - G L Hirst
- Department of Surgery, University of California, San Francisco, USA
| | - I Bedrosian
- Department of Breast Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - R M Layman
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J M Carter
- Department of Pathology, Mayo Clinic, Rochester, USA
| | - M Klein
- Department of Pathology, University of Minnesota, Minneapolis, USA
| | - S Venters
- Department of Surgery, University of California, San Francisco, USA
| | - S Shad
- Department of Surgery, University of California, San Francisco, USA
| | | | - A J Chien
- Department of Medicine, University of California, San Francisco, USA
| | - T Haddad
- Department of Medicine, Mayo Clinic, Rochester, USA
| | - C Isaacs
- Department of Medicine, Georgetown University, Washington, USA
| | - L Pusztai
- Department of Medicine, Yale University School of Medicine, New Haven, USA
| | - K Albain
- Department of Medicine, Loyola University, Chicago, USA
| | - R Nanda
- Department of Medicine, University of Chicago, Chicago, USA
| | - D Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - M C Liu
- Department of Medicine, Mayo Clinic, Rochester, USA
| | - J Boughey
- Department of Surgery, Mayo Clinic, Rochester, USA
| | - R Schwab
- Department of Medicine, University of California, San Diego, USA
| | - N Hylton
- Department of Radiology, University of California, San Francisco, USA
| | - A DeMichele
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, San Philadelphia, USA
| | | | - D Yee
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - D Berry
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L Van't Veer
- Department of Pathology, University of California, San Francisco, USA
| | - V Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L J Esserman
- Department of Surgery, University of California, San Francisco, USA
| | - W F Symmans
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Pathology, The University of Texas MD Anderson Cancer Center, San Francisco, USA.
| |
Collapse
|
133
|
Leon-Ferre RA, Hieken TJ, Boughey JC. The Landmark Series: Neoadjuvant Chemotherapy for Triple-Negative and HER2-Positive Breast Cancer. Ann Surg Oncol 2021; 28:2111-2119. [PMID: 33486641 DOI: 10.1245/s10434-020-09480-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/04/2020] [Indexed: 01/12/2023]
Abstract
While historically breast cancer has been treated with primary surgery followed by adjuvant therapy, the delivery of systemic therapy in the neoadjuvant setting has become increasingly common, especially for triple-negative and HER2-positive breast cancer. The initial motivations for pursuing neoadjuvant chemotherapy (NAC) were decreasing the tumor burden in the breast and axilla to enable de-escalation of surgery, and use the strategy to advance drug development. While these remain of interest, recent trials have additionally demonstrated survival advantages from escalation of systemic treatment in patients with residual disease, and new studies are testing de-escalation of systemic therapy based on pathologic response. Thus, response information to NAC has become pivotal to guide adjuvant treatment recommendations, and has resulted in NAC being the preferred approach for most HER2-positive and triple-negative breast cancers. Herein, we review select landmark trials that have paved the way for the use of chemotherapy in the neoadjuvant setting for breast cancer.
Collapse
Affiliation(s)
| | - Tina J Hieken
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
134
|
Potter DA, Herrera-Ponzanelli CA, Hinojosa D, Castillo R, Hernandez-Cruz I, Arrieta VA, Franklin MJ, Yee D. Recent advances in neoadjuvant therapy for breast cancer. Fac Rev 2021; 10:2. [PMID: 33659921 PMCID: PMC7894264 DOI: 10.12703/r/10-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neoadjuvant trials for early breast cancer have accelerated the identification of novel active agents, enabling streamlined conduct of registration trials with fewer subjects. Measurement of neoadjuvant drug effects has also enabled the identification of patients with high risk of distant recurrence and has justified development of additional adjuvant approaches to improve outcomes. Neoadjuvant evaluation of new drugs was significantly improved by the introduction of pathologic complete response (pCR) rate as a quantitative surrogate endpoint for distant disease-free survival (DDFS) and event free survival (EFS). The neoadjuvant phase 2 platform trial I-SPY 2 simultaneously tests multiple drugs across multiple breast cancer subtypes using Bayesian methods of adaptive randomization for assessment of drug efficacy. In addition to the pCR endpoint, the I-SPY 2 trial has demonstrated that the residual cancer burden (RCB) score measures gradations of tumor response that correlate with DDFS and EFS across treatments and subtypes. For HER2-positive and triple-negative breast cancers that have failed to attain pCR with neoadjuvant chemotherapy (NAC), effective modifications of adjuvant treatment have improved outcomes and changed the standard of care for these subtypes. Neoadjuvant therapy is therefore preferred for stage II and III, as well as some stage I, HER2-positive and triple-negative tumors. Neoadjuvant endocrine therapy (NET) strategies have also emerged from innovative trials for stage II and III estrogen receptor (ER)-positive/HER2-negative tumors, as in the ALTERNATE trial. From neoadjuvant trials, opportunities have emerged to de-escalate therapy on the basis of metrics of response to chemotherapy or hormonal therapy. Neoadjuvant therapy for early breast cancer is therefore emerging as a promising approach to accelerate new drug development, optimize treatment strategies, and (where appropriate) de-escalate neoadjuvant therapy.
Collapse
Affiliation(s)
| | - César A Herrera-Ponzanelli
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Diego Hinojosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Rafael Castillo
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Irwin Hernandez-Cruz
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Victor A Arrieta
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
| | | | - Douglas Yee
- University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
135
|
Gogia A, Choudhary P, Deo SVS, Sharma D, Mathur S, Batra A, Raju Sagiraju H. Correlation of pathological complete response with outcomes in locally advanced breast cancer treated with neoadjuvant chemotherapy: An ambispective study. CANCER RESEARCH, STATISTICS, AND TREATMENT 2021. [DOI: 10.4103/crst.crst_197_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
136
|
Asaoka M, Gandhi S, Ishikawa T, Takabe K. Neoadjuvant Chemotherapy for Breast Cancer: Past, Present, and Future. Breast Cancer (Auckl) 2020; 14:1178223420980377. [PMID: 33402827 PMCID: PMC7747102 DOI: 10.1177/1178223420980377] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) had been developed as a systematic approach before definitive surgery for the treatment of locally advanced or inoperable breast cancer such as inflammatory breast cancer in the past. In addition to its impact on surgery, the neoadjuvant setting has a benefit of providing the opportunity to monitor the individual drug response. Currently, the subject of NAC has expanded to include patients with early-stage, operable breast cancer because it is revealed that the achievement of a pathologic complete response (pCR) is associated with excellent long-term outcomes, especially in patients with aggressive phenotype breast cancer. In addition, this approach provides the unique opportunity to escalate adjuvant therapy in those with residual disease after NAC. Neoadjuvant chemotherapy in breast cancer is a rapidly evolving topic with tremendous interest in ongoing clinical trials. Here, we review the improvements and further challenges in the NAC setting in translational breast cancer research.
Collapse
Affiliation(s)
- Mariko Asaoka
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Shipra Gandhi
- Breast Medicine, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
137
|
Immunotherapy for early breast cancer: too soon, too superficial, or just right? Ann Oncol 2020; 32:323-336. [PMID: 33307202 DOI: 10.1016/j.annonc.2020.11.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/24/2020] [Accepted: 11/29/2020] [Indexed: 01/01/2023] Open
Abstract
Immunotherapy emerged as a new treatment modality for breast cancer, and its use is approved in combination with chemotherapy for first-line therapy in metastatic triple-negative breast cancer overexpressing PD-L1. As immune checkpoint inhibitors alone have modest clinical activity in advanced breast cancer, there is a growing interest in combinatorial modalities, and particularly for their rapid development in the early disease setting. The plethora of ongoing immunotherapy trials in early breast cancer comes at a time when solid data in advanced disease are still imperfect. This review offers a perspective on the efforts to establish the efficacy and safety of immunotherapeutic agents in early breast cancer.
Collapse
|
138
|
Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann Oncol 2020; 32:229-239. [PMID: 33232761 PMCID: PMC9348585 DOI: 10.1016/j.annonc.2020.11.007] [Citation(s) in RCA: 268] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/29/2020] [Accepted: 11/08/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) is strongly associated with favorable outcome. We examined the utility of serial circulating tumor DNA (ctDNA) testing for predicting pCR and risk of metastatic recurrence. PATIENTS AND METHODS Cell-free DNA (cfDNA) was isolated from 291 plasma samples of 84 high-risk early breast cancer patients treated in the neoadjuvant I-SPY 2 TRIAL with standard NAC alone or combined with MK-2206 (AKT inhibitor) treatment. Blood was collected at pretreatment (T0), 3 weeks after initiation of paclitaxel (T1), between paclitaxel and anthracycline regimens (T2), or prior to surgery (T3). A personalized ctDNA test was designed to detect up to 16 patient-specific mutations (from whole-exome sequencing of pretreatment tumor) in cfDNA by ultra-deep sequencing. The median follow-up time for survival analysis was 4.8 years. RESULTS At T0, 61 of 84 (73%) patients were ctDNA positive, which decreased over time (T1: 35%; T2: 14%; and T3: 9%). Patients who remained ctDNA positive at T1 were significantly more likely to have residual disease after NAC (83% non-pCR) compared with those who cleared ctDNA (52% non-pCR; odds ratio 4.33, P = 0.012). After NAC, all patients who achieved pCR were ctDNA negative (n = 17, 100%). For those who did not achieve pCR (n = 43), ctDNA-positive patients (14%) had a significantly increased risk of metastatic recurrence [hazard ratio (HR) 10.4; 95% confidence interval (CI) 2.3-46.6]; interestingly, patients who did not achieve pCR but were ctDNA negative (86%) had excellent outcome, similar to those who achieved pCR (HR 1.4; 95% CI 0.15-13.5). CONCLUSIONS Lack of ctDNA clearance was a significant predictor of poor response and metastatic recurrence, while clearance was associated with improved survival even in patients who did not achieve pCR. Personalized monitoring of ctDNA during NAC of high-risk early breast cancer may aid in real-time assessment of treatment response and help fine-tune pCR as a surrogate endpoint of survival.
Collapse
|
139
|
Sharma P, Kimler BF, O'Dea A, Nye L, Wang YY, Yoder R, Staley JM, Prochaska L, Wagner J, Amin AL, Larson K, Balanoff C, Elia M, Crane G, Madhusudhana S, Hoffmann M, Sheehan M, Rodriguez R, Finke K, Shah R, Satelli D, Shrestha A, Beck L, McKittrick R, Pluenneke R, Raja V, Beeki V, Corum L, Heldstab J, LaFaver S, Prager M, Phadnis M, Mudaranthakam DP, Jensen RA, Godwin AK, Salgado R, Mehta K, Khan Q. Randomized Phase II Trial of Anthracycline-free and Anthracycline-containing Neoadjuvant Carboplatin Chemotherapy Regimens in Stage I-III Triple-negative Breast Cancer (NeoSTOP). Clin Cancer Res 2020; 27:975-982. [PMID: 33208340 DOI: 10.1158/1078-0432.ccr-20-3646] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/29/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Addition of carboplatin (Cb) to anthracycline chemotherapy improves pathologic complete response (pCR), and carboplatin plus taxane regimens also yield encouraging pCR rates in triple-negative breast cancer (TNBC). Aim of the NeoSTOP multisite randomized phase II trial was to assess efficacy of anthracycline-free and anthracycline-containing neoadjuvant carboplatin regimens. PATIENTS AND METHODS Patients aged ≥18 years with stage I-III TNBC were randomized (1:1) to receive either paclitaxel (P) weekly × 12 plus carboplatin AUC6 every 21 days × 4 followed by doxorubicin/cyclophosphamide (AC) every 14 days × 4 (CbP → AC, arm A), or carboplatin AUC6 + docetaxel (D) every 21 days × 6 (CbD, arm B). Stromal tumor-infiltrating lymphocytes (sTIL) were assessed. Primary endpoint was pCR in breast and axilla. Other endpoints included residual cancer burden (RCB), toxicity, cost, and event-free (EFS) and overall survival (OS). RESULTS One hundred patients were randomized; arm A (n = 48) or arm B (n = 52). pCR was 54% [95% confidence interval (CI), 40%-69%] in arm A and 54% (95% CI, 40%-68%) in arm B. RCB 0+I rate was 67% in both arms. Median sTIL density was numerically higher in those with pCR compared with those with residual disease (20% vs. 5%; P = 0.25). At median follow-up of 38 months, EFS and OS were similar in the two arms. Grade 3/4 adverse events were more common in arm A compared with arm B, with the most notable differences in neutropenia (60% vs. 8%; P < 0.001) and febrile neutropenia (19% vs. 0%; P < 0.001). There was one treatment-related death (arm A) due to acute leukemia. Mean treatment cost was lower for arm B compared with arm A (P = 0.02). CONCLUSIONS The two-drug CbD regimen yielded pCR, RCB 0+I, and survival rates similar to the four-drug regimen of CbP → AC, but with a more favorable toxicity profile and lower treatment-associated cost.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas.
| | - Bruce F Kimler
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas
| | - Anne O'Dea
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Lauren Nye
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Yen Y Wang
- University of Kansas Cancer Center, Kansas City, Kansas
| | - Rachel Yoder
- University of Kansas Cancer Center, Kansas City, Kansas
| | | | - Lindsey Prochaska
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Jamie Wagner
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Amanda L Amin
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Kelsey Larson
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Christa Balanoff
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Manana Elia
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Gregory Crane
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Sheshadri Madhusudhana
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Marc Hoffmann
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Maureen Sheehan
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | | | - Karissa Finke
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Rajvi Shah
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Deepti Satelli
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Anuj Shrestha
- Richard & Annette Bloch Cancer Center, Truman Medical Center, Kansas City, Missouri
| | - Larry Beck
- Tammy Walker Cancer Center, Salina Regional Health Center, Salina, Kansas
| | - Richard McKittrick
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Robert Pluenneke
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Vinay Raja
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Venkatadri Beeki
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Larry Corum
- Olathe Cancer Care, Olathe Medical Center, Olathe, Kansas
| | | | | | - Micki Prager
- University of Kansas Cancer Center, Kansas City, Kansas
| | - Milind Phadnis
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Dinesh Pal Mudaranthakam
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Roy A Jensen
- University of Kansas Cancer Center, Kansas City, Kansas
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew K Godwin
- University of Kansas Cancer Center, Kansas City, Kansas
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Roberto Salgado
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
| | - Kathan Mehta
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| | - Qamar Khan
- Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas
| |
Collapse
|
140
|
Affiliation(s)
- Yu Shyr
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee
- Associate Editor for Statistics, JAMA Oncology
| | - Derek Shyr
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
141
|
Nietz S, O'Neil DS, Ayeni O, Chen WC, Joffe M, Jacobson JS, Neugut AI, Ruff P, Mapanga W, Buccimazza I, Singh U, Čačala S, Stopforth L, Phakathi B, Chirwa T, Cubasch H. A comparison of complete pathologic response rates following neoadjuvant chemotherapy among South African breast cancer patients with and without concurrent HIV infection. Breast Cancer Res Treat 2020; 184:861-872. [PMID: 32875480 DOI: 10.1007/s10549-020-05889-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Among patients diagnosed with breast cancer (BC), women also living with HIV (WLWH) have worse survival than women without HIV. Chronic HIV infection may interfere with the effectiveness of BC treatment, contributing to this disparity. We attempted to determine the impact of HIV infection on response to neoadjuvant chemotherapy (NACT) among South African women with BC. METHODS We evaluated women from the South African Breast Cancer and HIV Outcomes cohort study who had stage I-III disease, initiated NACT, underwent definitive breast surgery, and had available surgical pathology reports. We compared pathologic complete response (pCR) rates among women with and without HIV infection, using multivariable logistic regression to control for differences in tumor characteristics. We also evaluated the impact of HIV infection on pCR within subgroups based on patient and tumor factors. RESULTS Of 715 women, the 173 (24.2%) WLWH were less likely to achieve pCR than women without HIV (8.7% vs 16.4%, [odds ratio (OR) 0.48, 95% confidence interval (95% CI) 0.27-0.86]). WLWH continued to have lower likelihood of achieving pCR on multivariable analysis (OR 0.52, 95% CI 0.28-0.98). A similar pattern was seen within subgroups, although HIV infection appeared to affect pCR more in ER/PR-positive BC (OR 0.24, 95% CI 0.08-0.71) than in ER/PR-negative BC (OR 0.94, 95% CI 0.39-2.29). CONCLUSION WLWH were less like to achieve pCR following NACT for BC than women without HIV. This reduced response to systemic therapy may contribute to the poorer BC outcomes seen in WLWH.
Collapse
Affiliation(s)
- Sarah Nietz
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, 7 York Rd, Parktown, Johannesburg, 2193, Gauteng, South Africa
| | - Daniel S O'Neil
- Sylvester Comprehensive Cancer Center, University of Miami Health System, 1121 NW 14th Street, SMOB, Rm 245B, Miami, FL, 33150, USA. .,Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, USA.
| | - Oluwatosin Ayeni
- SAMRC/Wits Developmental Pathways for Health Research Unit, Department of Pediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, 31 Princess of Wales Terrace, Parktown, Johannesburg, 2193, South Africa
| | - Wenlong Carl Chen
- Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, 31 Princess of Wales Terrace, Parktown, Johannesburg, 2193, South Africa.,National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa.,Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, 9 Jubilee Road, Parktown, Johannesburg, 2193, South Africa
| | - Maureen Joffe
- SAMRC/Wits Developmental Pathways for Health Research Unit, Department of Pediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, 31 Princess of Wales Terrace, Parktown, Johannesburg, 2193, South Africa.,South Africa Medical Research Council Common Epithelial Cancers Research Centre, University of Witwatersrand, Johannesburg, South Africa
| | - Judith S Jacobson
- Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 W 168th Street, Room 732, New York, NY, 10032, USA
| | - Alfred I Neugut
- Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 W 168th Street, Room 732, New York, NY, 10032, USA.,Division of Medical Oncology, Columbia University Medical Center, 722 W 168th Street, Room 725, New York, NY, 10032, USA
| | - Paul Ruff
- Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, 31 Princess of Wales Terrace, Parktown, Johannesburg, 2193, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193, South Africa
| | - Witness Mapanga
- Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, 31 Princess of Wales Terrace, Parktown, Johannesburg, 2193, South Africa
| | - Ines Buccimazza
- Departments of Surgery and Oncology, Inkosi Albert Luthuli Central Hospital, Private Bag X03, Mayville, Durban, 4058, South Africa
| | - Urishka Singh
- Departments of Surgery and Oncology, Inkosi Albert Luthuli Central Hospital, Private Bag X03, Mayville, Durban, 4058, South Africa
| | - Sharon Čačala
- Departments of Surgery and Oncology, Grey's Hospital, University of KwaZulu Natal, Townbush Road, Pietermaritzburg, 3100, KZN, South Africa.,Department of Surgery, Ngwelezana Hospital, Thanduyise Road, Empangeni, 3880, KZN, South Africa
| | - Laura Stopforth
- Departments of Surgery and Oncology, Grey's Hospital, University of KwaZulu Natal, Townbush Road, Pietermaritzburg, 3100, KZN, South Africa
| | - Boitumelo Phakathi
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, 7 York Rd, Parktown, Johannesburg, 2193, Gauteng, South Africa
| | - Tobias Chirwa
- Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, 31 Princess of Wales Terrace, Parktown, Johannesburg, 2193, South Africa.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, 27 St Andrews Road, Parktown, Johannesburg, 2193, South Africa
| | - Herbert Cubasch
- Department of Surgery, Faculty of Health Sciences, University of Witwatersrand, 7 York Rd, Parktown, Johannesburg, 2193, Gauteng, South Africa.,Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, 31 Princess of Wales Terrace, Parktown, Johannesburg, 2193, South Africa
| |
Collapse
|