1
|
Wang X, Zhang Y, Yang M, Wu N, Wang S, Chen H, Zhou T, Zhang Y, Wang X, Jin Z, Zheng A, Yao F, Zhang D, Jin F, Qin P, Wang J. Dynamic ultrasound-based modeling predictive of response to neoadjuvant chemotherapy in patients with early breast cancer. Sci Rep 2024; 14:31644. [PMID: 39738182 PMCID: PMC11685924 DOI: 10.1038/s41598-024-80409-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Early prediction of patient responses to neoadjuvant chemotherapy (NACT) is essential for the precision treatment of early breast cancer (EBC). Therefore, this study aims to noninvasively and early predict pathological complete response (pCR). We used dynamic ultrasound (US) imaging changes acquired during NACT, along with clinicopathological features, to create a nomogram and construct a machine learning model. This retrospective study included 304 EBC patients recruited from multiple centers. All enrollees had completed NACT regimens, and underwent US examinations at baseline and at each NACT cycle. We subsequently determined that percentage reduction of tumor maximum diameter from baseline to third cycle of NACT serves to independent predictor for pCR, enabling creation of a nomogram ([Formula: see text]). Our predictive accuracy further improved ([Formula: see text]) by combining dynamic US data and clinicopathological features in a machine learning model. Such models may offer a means of accurately predicting NACT responses in this setting, helping to individualize patient therapy. Our study may provide additional insights into the US-based response prediction by focusing on the dynamic changes of the tumor in the early and full NACT cycle.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China
| | - Yuting Zhang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China
| | - Mengting Yang
- Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Nan Wu
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China
| | - Shan Wang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China
| | - Hong Chen
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China
| | - Tianyang Zhou
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China
| | - Ying Zhang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China
| | - Xiaolan Wang
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zining Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Ang Zheng
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Fan Yao
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Dianlong Zhang
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Pan Qin
- Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Jia Wang
- Department of Breast Surgery, Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, China.
| |
Collapse
|
2
|
Afzal MZ, Vahdat LT. Evolving Management of Breast Cancer in the Era of Predictive Biomarkers and Precision Medicine. J Pers Med 2024; 14:719. [PMID: 39063972 PMCID: PMC11278458 DOI: 10.3390/jpm14070719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/17/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is the most common cancer among women in the world as well as in the United States. Molecular and histological differentiation have helped clinicians optimize treatments with various therapeutics, including hormonal therapy, chemotherapy, immunotherapy, and radiation therapy. Recently, immunotherapy has become the standard of care in locally advanced triple-negative breast cancer and an option across molecular subtypes for tumors with a high tumor mutation burden. Despite the advancements in personalized medicine directing the management of localized and advanced breast cancers, the emergence of resistance to these therapies is the leading cause of death among breast cancer patients. Therefore, there is a critical need to identify and validate predictive biomarkers to direct treatment selection, identify potential responders, and detect emerging resistance to standard therapies. Areas of active scientific and clinical research include novel personalized and predictive biomarkers incorporating tumor microenvironment, tumor immune profiling, molecular characterization, and histopathological differentiation to predict response and the potential emergence of resistance.
Collapse
Affiliation(s)
- Muhammad Zubair Afzal
- Medical Oncology, Comprehensive Breast Program, Dartmouth Cancer Center, Lebanon, NH 03755, USA
| | - Linda T. Vahdat
- Medical Oncology and Hematology (Interim), Dartmouth Cancer Center, Lebanon, NH 03755, USA;
| |
Collapse
|
3
|
McGale J, Khurana S, Huang A, Roa T, Yeh R, Shirini D, Doshi P, Nakhla A, Bebawy M, Khalil D, Lotfalla A, Higgins H, Gulati A, Girard A, Bidard FC, Champion L, Duong P, Dercle L, Seban RD. PET/CT and SPECT/CT Imaging of HER2-Positive Breast Cancer. J Clin Med 2023; 12:4882. [PMID: 37568284 PMCID: PMC10419459 DOI: 10.3390/jcm12154882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
HER2 (Human Epidermal Growth Factor Receptor 2)-positive breast cancer is characterized by amplification of the HER2 gene and is associated with more aggressive tumor growth, increased risk of metastasis, and poorer prognosis when compared to other subtypes of breast cancer. HER2 expression is therefore a critical tumor feature that can be used to diagnose and treat breast cancer. Moving forward, advances in HER2 in vivo imaging, involving the use of techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT), may allow for a greater role for HER2 status in guiding the management of breast cancer patients. This will apply both to patients who are HER2-positive and those who have limited-to-minimal immunohistochemical HER2 expression (HER2-low), with imaging ultimately helping clinicians determine the size and location of tumors. Additionally, PET and SPECT could help evaluate effectiveness of HER2-targeted therapies, such as trastuzumab or pertuzumab for HER2-positive cancers, and specially modified antibody drug conjugates (ADC), such as trastuzumab-deruxtecan, for HER2-low variants. This review will explore the current and future role of HER2 imaging in personalizing the care of patients diagnosed with breast cancer.
Collapse
Affiliation(s)
- Jeremy McGale
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Sakshi Khurana
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Alice Huang
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Tina Roa
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Randy Yeh
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dorsa Shirini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Parth Doshi
- Campbell University School of Osteopathic Medicine, Lillington, NC 27546, USA
| | - Abanoub Nakhla
- American University of the Caribbean School of Medicine, Cupecoy, Sint Maarten
| | - Maria Bebawy
- Touro College of Osteopathic Medicine, Middletown, NY 10940, USA
| | - David Khalil
- Campbell University School of Osteopathic Medicine, Lillington, NC 27546, USA
| | - Andrew Lotfalla
- Touro College of Osteopathic Medicine, Middletown, NY 10940, USA
| | - Hayley Higgins
- Touro College of Osteopathic Medicine, Middletown, NY 10940, USA
| | - Amit Gulati
- Department of Internal Medicine, Maimonides Medical Center, New York, NY 11219, USA
| | - Antoine Girard
- Department of Nuclear Medicine, CHU Amiens-Picardie, 80054 Amiens, France
| | - Francois-Clement Bidard
- Department of Medical Oncology, Inserm CIC-BT 1428, Curie Institute, Paris Saclay University, UVSQ, 78035 Paris, France
| | - Laurence Champion
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210 Saint-Cloud, France
- Laboratory of Translational Imaging in Oncology, Paris Sciences et Lettres (PSL) Research University, Institut Curie, 91401 Orsay, France
| | - Phuong Duong
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Laurent Dercle
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Romain-David Seban
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210 Saint-Cloud, France
- Laboratory of Translational Imaging in Oncology, Paris Sciences et Lettres (PSL) Research University, Institut Curie, 91401 Orsay, France
| |
Collapse
|
4
|
Dowling GP, Keelan S, Toomey S, Daly GR, Hennessy BT, Hill ADK. Review of the status of neoadjuvant therapy in HER2-positive breast cancer. Front Oncol 2023; 13:1066007. [PMID: 36793602 PMCID: PMC9923093 DOI: 10.3389/fonc.2023.1066007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Purpose The development of human epidermal growth factor receptor 2 (HER2)-directed therapies has revolutionized the treatment of HER2-positive breast cancer. The aim of this article is to review the continually evolving treatment strategies in the neoadjuvant setting of HER2-positive breast cancer, as well as the current challenges and future perspectives. Methods Searches were undertaken on PubMed and Clinicaltrials.gov for relevant publications and trials. Findings The current standard of care in high-risk HER2-positive breast cancer is to combine chemotherapy with dual anti-HER2 therapy, for a synergistic anti-tumor effect. We discuss the pivotal trials which led to the adoption of this approach, as well as the benefit of these neoadjuvant strategies for guiding appropriate adjuvant therapy. De-escalation strategies are currently being investigated to avoid over treatment, and aim to safely reduce chemotherapy, while optimizing HER2-targeted therapies. The development and validation of a reliable biomarker is essential to enable these de-escalation strategies and personalization of treatment. In addition, promising novel therapies are currently being explored to further improve outcomes in HER2-positive breast cancer.
Collapse
Affiliation(s)
- Gavin P Dowling
- Medical Oncology Lab, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.,The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Stephen Keelan
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Lab, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gordon R Daly
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Bryan T Hennessy
- Medical Oncology Lab, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Arnold D K Hill
- The Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland.,The Department of Surgery, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
5
|
Portnow LH, Kochkodan-Self JM, Maduram A, Barrios M, Onken AM, Hong X, Mittendorf EA, Giess CS, Chikarmane SA. Multimodality Imaging Review of HER2-positive Breast Cancer and Response to Neoadjuvant Chemotherapy. Radiographics 2023; 43:e220103. [PMID: 36633970 DOI: 10.1148/rg.220103] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2/neu or ErbB2)-positive breast cancers comprise 15%-20% of all breast cancers. The most common manifestation of HER2-positive breast cancer at mammography or US is an irregular mass with spiculated margins that often contains calcifications; at MRI, HER2-positive breast cancer may appear as a mass or as nonmass enhancement. HER2-positive breast cancers are often of intermediate to high nuclear grade at histopathologic analysis, with increased risk of local recurrence and metastases and poorer overall prognosis. However, treatment with targeted monoclonal antibody therapies such as trastuzumab and pertuzumab provides better local-regional control and leads to improved survival outcome. With neoadjuvant treatments, including monoclonal antibodies, taxanes, and anthracyclines, women are now potentially able to undergo breast conservation therapy and sentinel lymph node biopsy versus mastectomy and axillary lymph node dissection. Thus, the radiologist's role in assessing the extent of local-regional disease and response to neoadjuvant treatment at imaging is important to inform surgical planning and adjuvant treatment. However, assessment of treatment response remains difficult, with the potential for different imaging modalities to result in underestimation or overestimation of disease to varying degrees when compared with surgical pathologic analysis. In particular, the presence of calcifications at mammography is especially difficult to correlate with the results of pathologic analysis after chemotherapy. Breast MRI findings remain the best predictor of pathologic response. The authors review the initial manifestations of HER2-positive tumors, the varied responses to neoadjuvant chemotherapy, and the challenges in assessing residual cancer burden through a multimodality imaging review with pathologic correlation. © RSNA, 2023 Quiz questions for this article are available through the Online Learning Center.
Collapse
Affiliation(s)
- Leah H Portnow
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Jeanne M Kochkodan-Self
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Amy Maduram
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Mirelys Barrios
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Allison M Onken
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Xuefei Hong
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Elizabeth A Mittendorf
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Catherine S Giess
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| | - Sona A Chikarmane
- From the Departments of Radiology (L.H.P., J.M.K.S., A.M., M.B., C.S.G., S.A.C.), Pathology (A.M.O., X.H.), and Surgery (E.A.M.), Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115
| |
Collapse
|
6
|
Liu Y, Wang Y, Wang Y, Xie Y, Cui Y, Feng S, Yao M, Qiu B, Shen W, Chen D, Du G, Chen X, Liu Z, Li Z, Yang X, Liang C, Wu L. Early prediction of treatment response to neoadjuvant chemotherapy based on longitudinal ultrasound images of HER2-positive breast cancer patients by Siamese multi-task network: A multicentre, retrospective cohort study. EClinicalMedicine 2022; 52:101562. [PMID: 35928032 PMCID: PMC9343415 DOI: 10.1016/j.eclinm.2022.101562] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Early prediction of treatment response to neoadjuvant chemotherapy (NACT) in patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer can facilitate timely adjustment of treatment regimens. We aimed to develop and validate a Siamese multi-task network (SMTN) for predicting pathological complete response (pCR) based on longitudinal ultrasound images at the early stage of NACT. METHODS In this multicentre, retrospective cohort study, a total of 393 patients with biopsy-proven HER2-positive breast cancer were retrospectively enrolled from three hospitals in china between December 16, 2013 and March 05, 2021, and allocated into a training cohort and two external validation cohorts. Patients receiving full cycles of NACT and with surgical pathological results available were eligible for inclusion. The key exclusion criteria were missing ultrasound images and/or clinicopathological characteristics. The proposed SMTN consists of two subnetworks that could be joined at multiple layers, which allowed for the integration of multi-scale features and extraction of dynamic information from longitudinal ultrasound images before and after the first /second cycles of NACT. We constructed the clinical model as a baseline using multivariable logistic regression analysis. Then the performance of SMTN was evaluated and compared with the clinical model. FINDINGS The training cohort, comprising 215 patients, were selected from Yunnan Cancer Hospital. The two independent external validation cohorts, comprising 95 and 83 patients, were selected from Guangdong Provincial People's Hospital, and Shanxi Cancer Hospital, respectively. The SMTN yielded an area under the receiver operating characteristic curve (AUC) values of 0.986 (95% CI: 0.977-0.995), 0.902 (95%CI: 0.856-0.948), and 0.957 (95%CI: 0.924-0.990) in the training cohort and two external validation cohorts, respectively, which were significantly higher than that those of the clinical model (AUC: 0.524-0.588, P all < 0.05). The AUCs values of the SMTN within the anti-HER2 therapy subgroups were 0.833-0.972 in the two external validation cohorts. Moreover, 272 of 279 (97.5%) non-pCR patients (159 of 160 (99.4%), 53 of 54 (98.1%), and 60 of 65 (92.3%) in the training and two external validation cohorts, respectively) were successfully identified by the SMTN, suggesting that they could benefit from regime adjustment at the early-stage of NACT. INTERPRETATION The SMTN was able to predict pCR in the early-stage of NACT for HER2-positive breast cancer patients, which could guide clinicians in adjusting treatment regimes. FUNDING Key-Area Research and Development Program of Guangdong Province (No.2021B0101420006); National Natural Science Foundation of China (No.82071892, 82171920); Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application (No.2022B1212010011); the National Science Foundation for Young Scientists of China (No.82102019, 82001986); Project Funded by China Postdoctoral Science Foundation (No.2020M682643); the Outstanding Youth Science Foundation of Yunnan Basic Research Project (202101AW070001); Scientific research fund project of Department of Education of Yunnan Province(2022J0249). Science and technology Projects in Guangzhou (202201020001;202201010513); High-level Hospital Construction Project (DFJH201805, DFJHBF202105).
Collapse
Affiliation(s)
- Yu Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Ying Wang
- Department of Medical Ultrasonics, the First Affiliated Hospital of Guangzhou medical University, 151 Yanjiang West Road, 510120, China
| | - Yuxiang Wang
- Department of Ultrasound, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, 030013, China
| | - Yu Xie
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Yanfen Cui
- Department of Radiology, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, 030013, China
| | - Senwen Feng
- Department of General Surgery, Shenzhen YanTian district people's hospital (group), Shenzhen, 518081, China
| | - Mengxia Yao
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
| | - Bingjiang Qiu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Wenqian Shen
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
| | - Dong Chen
- Department of Medical Ultrasound, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Guoqing Du
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou, 510180, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhenhui Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
- Corresponding author at: Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China.
| | - Xiaotang Yang
- Department of Radiology, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, 030013, China
- Corresponding author at: Department of Radiology, Shanxi Cancer Hospital, Shanxi Medical University, Taiyuan, 030013, China.
| | - Changhong Liang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Corresponding author at: Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China.
| | - Lei Wu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Corresponding author at: Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhangshan Er Road, Guangzhou 510080, China.
| |
Collapse
|
7
|
Wang J, Wang X, Chen R, Liang M, Li M, Ma G, Xia T, Wang S. Circulating tumor cells may serve as a supplement to RECIST in neoadjuvant chemotherapy of patients with locally advanced breast cancer. Int J Clin Oncol 2022; 27:889-898. [PMID: 35122586 DOI: 10.1007/s10147-022-02125-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 01/20/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Circulating tumor cells (CTCs) have been shown to be associated with the response to neoadjuvant chemotherapy (NCT) and the prognosis of locally advanced breast cancer (LABC) patients. Our study aimed to investigate whether the change of CTC status during NCT could serve as a supplement to the Response Evaluation Criteria in Solid Tumors (RECIST) in the treatment and evaluation of LABC patients. METHODS 6 ml of blood samples were collected before NCT, after the first cycle of NCT and after the completion of NCT, respectively. According to the change of CTC number during NCT, the patients were divided into "CTC low-response (low-R)" group and "CTC high-response (high-R)" group. Survival data of each group of patients were obtained through long-term follow-up. RESULTS A total of 35 patients diagnosed with LABC were enrolled. The median follow-up for distant metastasis was 27 months (range 7-36 months). There was no significant difference in distant metastasis-free survival (DMFS) between PR/CR group and PD/SD group (P = 0.0914), while CTC low-R group had a worse DMFS than CTC high-R group (P = 0.0199). In PR/CR subgroup, patients with CTC low-R showed a lower DMFS compared with those with CTC high-R (P = 0.0159). However, in PD/SD subgroup, there was no significant difference in DMFS between CTC low-R and CTC high-R group (P = 0.7521). In terms of assessing response to NCT, CTC change or RECIST classification alone had an AUC of 0.533 (95% CI 0.277-0.790) and 0.700 (95% CI 0.611-0.789), respectively. When combining the two, the AUC slightly increased to 0.713 (95% CI 0.532-0.895). CONCLUSION The change of CTC number during NCT has a potential to serve as a supplement to RECIST in the assessment of NCT efficacy and the prognosis of LABC patients.
Collapse
Affiliation(s)
- Ji Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xinyang Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Rui Chen
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Mengdi Liang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Minghui Li
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Ge Ma
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
8
|
Early Changes in [ 18F]FDG Uptake as a Readout for PI3K/Akt/mTOR Targeted Drugs in HER-2-Positive Cancer Xenografts. Mol Imaging 2021; 2021:5594514. [PMID: 34113218 PMCID: PMC8169268 DOI: 10.1155/2021/5594514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/12/2021] [Accepted: 04/29/2021] [Indexed: 11/24/2022] Open
Abstract
We investigated the potential use of [18F]FDG PET as a response biomarker for PI3K pathway targeting therapies in two HER-2-overexpressing cancer models. Methods. CD-1 nude mice were inoculated with HER-2-overexpressing JIMT1 (trastuzumab-resistant) or SKOV3 (trastuzumab-sensitive) human cancer cells. Animals were treated with trastuzumab, everolimus (mTOR inhibitor), PIK90 (PI3K inhibitor), saline, or combination therapy. [18F]FDG scans were performed at baseline, two, and seven days after the start of the therapy. Tumors were delineated on CT images and relative tumor volumes (RTV) and maximum standardized uptake value (SUVmax) were calculated. Levels of pS6 and pAkt on protein tumor lysates were determined with ELISA. Results. In the SKOV3 xenografts, all treatment schedules resulted in a gradual decrease in RTV and delta SUVmax (ΔSUVmax). For all treatments combined, ΔSUVmax after 2 days was predictive for RTV after 7 days (r = 0.69, p = 0.030). In JIMT1 tumors, monotherapy with everolimus or PIK90 resulted in a decrease in RTV (−30% ± 10% and −20% ± 20%, respectively) and ΔSUVmax (−39% ± 36% and −42% ± 8%, respectively) after 7 days of treatment, but not earlier, while trastuzumab resulted in nonsignificant increases compared to control. Combination therapies resulted in RTV and ΔSUVmax decrease already at day 2, except for trastuzumab+everolimus, where an early flare was observed. For all treatments combined, ΔSUVmax after 2 days was predictive for RTV after 7 days (r = 0.48, p = 0.028), but the correlation could be improved when combination with everolimus (r = 0.59, p = 0.023) or trastuzumab (r = 0.69, p = 0.015) was excluded. Conclusion. Reduction in [18F]FDG after 2 days correlated with tumor volume changes after 7 days of treatment and confirms the use of [18F]FDG PET as an early response biomarker. Treatment response can however be underestimated in schedules containing trastuzumab or everolimus due to temporary increased [18F]FDG uptake secondary to negative feedback loop and crosstalk between different pathways.
Collapse
|
9
|
Li H, Wang J, Yi Z, Li C, Wang H, Zhang J, Wang T, Nan P, Lin F, Xu D, Qian H, Ma F. CDK12 inhibition enhances sensitivity of HER2+ breast cancers to HER2-tyrosine kinase inhibitor via suppressing PI3K/AKT. Eur J Cancer 2021; 145:92-108. [PMID: 33429148 DOI: 10.1016/j.ejca.2020.11.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Alhtough anti-HER2 tyrosine kinase inhibitors (TKIs) have radically prolonged survival and improved prognosis in HER2-positive breast cancer patients, resistance to these therapies is a constant obstacle leading to TKIs treatment failure and tumour progression. METHODS To develop new strategies to enhance TKIs efficiency by combining synergistic gene targets, we performed panel library screening using the CRISPR/Cas9 knockout technique based on data mining across TCGA data sets and verified the candidate target in preclinical models and breast cancer high-throughput sequencing data sets. RESULTS We identified that CDK12, co-amplified with HER2 in a high frequency, is powerful to sensitise or resensitise HER2-positive breast cancer to anti-HER2 TKIs lapatinib, evidenced by patient-derived organoids in vitro and cell-derived xenograft or patient-derived xenograft in vivo. Exploring mechanisms, we found that inhibition of CDK12 attenuated PI3K/AKT signal, which usually serves as an oncogenic driver and is reactivated when HER2-positive breast cancers develop resistance to lapatinib. Combining CDK12 inhibition exerted additional suppression on p-AKT activation induced by anti-HER2 TKIs lapatinib treatment. Clinically, via DNA sequencing data for tumour tissue and peripheral blood ctDNA, we found that HER2-positive breast cancer patients with CDK12 amplification responded more insensitively to anti-HER2 treatment than those without accompanying CDK12 amplification by harbouring a markedly shortened progression-free survival (PFS) (median PFS: 4.3 months versus 6.9 months; hazards ratio [HR] = 2.26 [95% confidence interval [CI] = 1.32-3.86]; P = 0.0028). CONCLUSIONS Dual inhibition of HER2/CDK12 will prominently benefit the outcomes of patients with HER2-positive breast cancer by sensitising or resensitising the tumours to anti-HER2 TKIs treatment.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cyclic N-Oxides/pharmacology
- Cyclin-Dependent Kinases/antagonists & inhibitors
- Cyclin-Dependent Kinases/genetics
- Cyclin-Dependent Kinases/metabolism
- Databases, Genetic
- Drug Resistance, Neoplasm
- Female
- Gene Amplification
- Gene Expression Regulation, Neoplastic
- Humans
- Indolizines/pharmacology
- Lapatinib/pharmacology
- Mice, Inbred BALB C
- Mice, Nude
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/metabolism
- Pyridinium Compounds/pharmacology
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Hui Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jinsong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zongbi Yi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiao Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haijuan Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingyao Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ting Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Nan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Feng Lin
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dongkui Xu
- Department of VIP, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Fei Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
10
|
Skarping I, Förnvik D, Heide-Jørgensen U, Rydén L, Zackrisson S, Borgquist S. Neoadjuvant breast cancer treatment response; tumor size evaluation through different conventional imaging modalities in the NeoDense study. Acta Oncol 2020; 59:1528-1537. [PMID: 33063567 DOI: 10.1080/0284186x.2020.1830167] [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] [Indexed: 01/10/2023]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) is offered to an increasing number of breast cancer (BC) patients, and comprehensive monitoring of treatment response is of utmost importance. Several imaging modalities are available to follow tumor response, although likely to provide different clinical information. We aimed to examine the association between early radiological response by three conventional imaging modalities and pathological complete response (pCR). Further, we investigated the agreement between these modalities pre-, during, and post-NACT, and the accuracy of predicting pathological residual tumor burden by these imaging modalities post-NACT. MATERIAL AND METHODS This prospective Swedish cohort study included 202 BC patients assigned to NACT (2014-2019). Breast imaging with clinically used modalities: mammography, ultrasound, and tomosynthesis was performed pre-, during, and post-NACT. We investigated the agreement of tumor size by the different imaging modalities, and their accuracy of tumor size estimation. Patients with a radiological complete response or radiological partial response (≥30% decrease in tumor diameter) during NACT were classified as radiological early responders. RESULTS Patients with an early radiological response by ultrasound had 2.9 times higher chance of pCR than early radiological non-responders; the corresponding relative chance for mammography and tomosynthesis tumor size measures was 1.8 and 2.8, respectively. Post-NACT, each modality, separately, could accurately estimate tumor size (within 5 mm margin compared to pathological evaluation) in 43-46% of all tumors. The diagnostic precision in predicting pCR post-NACT was similar between the three imaging modalities; however, tomosynthesis had slightly higher specificity and positive predictive values. CONCLUSION Breast imaging modalities correctly estimated pathological tumor size in less than half of the tumors. Based on this finding, predicting residual tumor size post-NACT is challenging using conventional imaging. Patients with early radiological non-response might need improved monitoring during NACT and be considered for changed treatment plans.
Collapse
Affiliation(s)
- Ida Skarping
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Daniel Förnvik
- Department of Translational Medicine, Medical Radiation Physics, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Uffe Heide-Jørgensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Lisa Rydén
- Department of Surgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Sophia Zackrisson
- Department of Translational Medicine, Diagnostic Radiology, Lund University, Skåne University Hospital, Lund, Sweden
- Department of Translational Medicine, Diagnostic Radiology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Skåne University Hospital, Lund, Sweden
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
11
|
Hong J, Wu J, Huang O, He J, Zhu L, Chen W, Li Y, Chen X, Shen K. Early response and pathological complete remission in Breast Cancer with different molecular subtypes: a retrospective single center analysis. J Cancer 2020; 11:6916-6924. [PMID: 33123282 PMCID: PMC7591996 DOI: 10.7150/jca.46805] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/06/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose: To evaluate the association of clinical early response and pathological complete remission (pCR) in breast cancer patients with different molecular subtypes. Materials and methods: Breast cancer patients who received neoadjuvant treatment (NAT) with clinical early response assessment from October 2008 to October 2018 were retrospectively analyzed. Clinical early response was defined as tumor size decreasing ≥30% evaluated by ultrasound after two cycles of NAT. Chi-square test was used to compare the pCR rates between the responder and non-responder groups with different molecular subtypes. Multivariate logistic regression was used to identify independent factors associated with the pCR. Results: A total of 328 patients were included: 100 responders and 228 non-responders. The progesterone receptor (PR) expression was an independent factor associated with clinical early response (OR=2.39, 95%CI=1.41-4.05, P=0.001). The pCR rate of breast was 50.0% for responders and 18.0% for non-responders (P<0.001). Regarding different molecular subtypes, responders had higher pCR rates than non-responders for patients with HER2 overexpression (OR=10.66, 95%CI=2.18-52.15, P=0.001), triple negative (OR=3.29, 95%CI=1.23-8.84, P=0.016) and Luminal (HER2-) subtypes (OR=8.58, 95%CI=3.05-24.10, P<0.001) respectively. Moreover, pCR rate can be achieved as high as 88.2% in HER2 overexpression patients with early clinical response, which was significantly higher than patients without early response (41.3%, P=0.001). Multivariate analysis showed that clinical early response was an independent factor associated with the pCR rate (OR=4.87, 95%CI=2.72-8.72, P<0.001). Conclusions: Early response was significantly associated with a higher pCR rate in breast cancer patients receiving NAT, especially for patients with HER2 overexpression subtype, which warrants further clinical evaluation.
Collapse
Affiliation(s)
- Jin Hong
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Jiayi Wu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Ou Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Jianrong He
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Li Zhu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Weiguo Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yafen Li
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| |
Collapse
|
12
|
Sharma P, Connolly RM, Roussos Torres ET, Thompson A. Best Foot Forward: Neoadjuvant Systemic Therapy as Standard of Care in Triple-Negative and HER2-Positive Breast Cancer. Am Soc Clin Oncol Educ Book 2020; 40:1-16. [PMID: 32315235 DOI: 10.1200/edbk_281381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Neoadjuvant systemic treatment of early-stage breast cancer has been used to improve resectability and reduce the extent of breast and axillary surgery. More recently, several other merits of neoadjuvant systemic treatment have emerged, including the ability to tailor clinically available adjuvant systemic therapy options based on pathologic response and to serve as a platform for early assessment of novel agents and response biomarkers and as an avenue for treatment optimization investigations (local and systemic therapy escalation and de-escalation trials guided by pathologic response). Attainment of a pathologic complete response (pCR) is associated with excellent long-term outcomes; conversely, the presence of residual disease is associated with a high risk of recurrence for patients with HER2-positive breast cancer and triple-negative breast cancer (TNBC). Treatment strategies in early-stage HER2-positive breast cancer include regimens incorporating trastuzumab, pertuzumab, ado-trastuzumab emtansine, and neratinib, resulting in high pCR rates and overall excellent long-term outcomes. Currently available cytotoxic regimens yield pCR for 35% to 55% of patients with TNBC, and immune checkpoint inhibition is showing early promise for this subtype. New drug and predictive biomarker evaluations in the neoadjuvant setting aim to develop optimal treatment strategies for the individual patient, with the ultimate goal of maximizing efficacy and minimizing toxicity. Research efforts involving novel agents are being undertaken to address the high risk of recurrence for patients with residual disease. Omission of breast surgery following neoadjuvant chemotherapy requires further development of imaging and biopsy techniques to accurately assess the extent of residual disease before clinical application.
Collapse
Affiliation(s)
- Priyanka Sharma
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Westwood, KS
| | | | | | - Alastair Thompson
- Department of Surgery, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
13
|
|
14
|
Cordoba O, Carrillo-Guivernau L, Reyero-Fernández C. Surgical Management of Breast Cancer Treated with Neoadjuvant Therapy. Breast Care (Basel) 2018; 13:238-243. [PMID: 30319325 PMCID: PMC6167713 DOI: 10.1159/000491760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neoadjuvant therapy (NAT) allows downstaging in some cases of breast cancer. By consequence, it may enable a more conservative surgical approach or make surgery possible in cases ineligible for surgery before NAT. In this article, we review the evidence and management recommendations for optimal surgical treatment in this setting.
Collapse
Affiliation(s)
- Octavi Cordoba
- Obstetrics and Gynecology Department, Hospital Universitari Son Espases, Palma, Spain
| | - Lourdes Carrillo-Guivernau
- Breast Cancer Unit, Obstetrics and Gynecology Department, Hospital Universitari Son Espases, Palma, Spain
| | | |
Collapse
|