1
|
Weiss A, Agnese DM, Al-Hilli Z, Cabioglu N, Farr D, Kantor O, Obeng-Gyasi S, Wilke L. An Overview of the Importance of Neoadjuvant Systemic Therapy for Breast Cancer Patients: From the Society of Surgical Oncology and the American Society of Breast Surgeons. Ann Surg Oncol 2025:10.1245/s10434-025-17405-7. [PMID: 40355803 DOI: 10.1245/s10434-025-17405-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025]
Affiliation(s)
- Anna Weiss
- Division of Surgical Oncology, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| | - Doreen M Agnese
- Division of Surgical Oncology, The Ohio State University, Columbus, OH, USA
| | - Zahraa Al-Hilli
- Breast Center, Integrated Surgical Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Deborah Farr
- Department of Surgery at UT Southwestern Medical Center, Dallas, TX, USA
| | - Olga Kantor
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Lee Wilke
- UW Health/Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
2
|
Talec H, Aubé C, Guerin-Charbonnel C, Berge P. The use of a clip prior to neoadjuvant chemotherapy for breast cancer with microcalcifications may not always be required. Breast Cancer Res Treat 2025; 209:585-593. [PMID: 39417908 PMCID: PMC11785642 DOI: 10.1007/s10549-024-07517-1] [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: 07/10/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Neoadjuvant chemotherapy is now a common first line therapy for breast cancer. International guidelines recommend placement of a clip before commencement of therapy to assist with localizing the tumor bed in the event of excellent response-this takes up time and resources. The microcalcifications associated usually persist after chemotherapy and could serve as an alternative marker. We investigated to determine prognostic criteria to avoid the need for a marker clip before neoadjuvant chemotherapy for breast tumors associated with microcalcifications. METHODS We performed a 7 year single-center bi-site retrospective analytical observational study of 88 women with calcified breast carcinoma treated by neoadjuvant chemotherapy at our bi-site institution between September 2015 and September 2022. This study includied two groups (clip-free tumor localization vs. clip-free tumor non-localization), and investigating quantitative and qualitative predictive factors. The clip-free tumor localization after neoadjuvant chemotherapy was defined by the visibility of residual calcifications on both views of the pre-operative mammogram on the day of or the day prior to surgery. RESULTS The mean age of the 88 women included in our population was 52.8 years (± 12.7 years standard deviation). Of the 90 tumors with microcalcifications, 64 carcinomas (71.1%) were localizable with no marker clip after neoadjuvant chemotherapy. The main predictive factors significantly associated with clip-free tumor localization were number of calcifications > 10 (P < 0.0001), grade 2 tumor (P = 0.003) with a probability of locating tumor after neoadjuvant chemotherapy of 97.9%, 95% CI [95.6; 99.0]. CONCLUSION More than 10 microcalcifications in a grade 2 breast tumor at the initial diagnosis may obviate the need for a marker clip.
Collapse
Affiliation(s)
- Henri Talec
- Department of Radiology and Medical Imaging, University Hospital, 49100, Angers, France.
| | - Christophe Aubé
- Department of Radiology and Medical Imaging, University Hospital, 49100, Angers, France
| | | | - Pierre Berge
- Institut de Cancérologie de l'Ouest, 49055, Angers, France
| |
Collapse
|
3
|
Allotey J, Ruparel V, McCallum A, Somal K, Simpson L, Gupta G, Lip G, Sharma R, Masannat Y. Residual microcalcifications after neoadjuvant systemic therapy for early breast cancer: Implications for surgical planning and long-term outcomes. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:108781. [PMID: 39486131 DOI: 10.1016/j.ejso.2024.108781] [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: 08/09/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Residual microcalcifications on mammograms after neoadjuvant chemotherapy (NACT) pose a challenge in surgical decision-making. This single-centre retrospective review of all patients who had NACT for breast cancer over five years, evaluated the relationship between pathological complete response and residual microcalcifications, controlling for tumour size, nodal stage, grade, and receptor status, as well as the impact of residual microcalcifications on recurrence and survival. There was no significant association between pathological complete response (pCR) and residual microcalcifications (p = 0.763). We computed hazard ratios (HR) for Time to recurrence (TTR) and overall survival (OS) which were both not significant, with HR = 2.599, [0.290, 23.264], p = 0.393 and HR = 1.362 [0.123, 15.062], p = 0.801 respectively. The predictive and prognostic significance of residual microcalcifications remains to be proven. The surgical excision of these lesions should be considered based on individual patient risk.
Collapse
Affiliation(s)
- Joel Allotey
- Department of Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK.
| | - Vinita Ruparel
- Department of Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - Anna McCallum
- Department of Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - Karendeep Somal
- Department of Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - Louise Simpson
- Breast Unit, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - Gaurav Gupta
- Department of Radiology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK; Diagnostic Imaging Department, St Marys Hospital, Isle of Wight NHS Trust, Newport, UK
| | - Gerald Lip
- Department of Radiology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK
| | - Ravi Sharma
- Department of Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, UK; School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Yazan Masannat
- Breast Unit, Broomfield Hospital, Mid and South Essex NHS Trust, Chelmsford, UK; School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
4
|
Kwon MR, Ko EY, Lee JE, Han BK, Ko ES, Choi JS, Kim H, Kim MK, Yu J, Lee H, Youn I. Prediction model for individualized precision surgery in breast cancer patients with complete response on MRI and residual calcifications on mammography after neoadjuvant chemotherapy. Breast Cancer 2025; 32:109-119. [PMID: 39348079 DOI: 10.1007/s12282-024-01638-7] [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: 07/12/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Identifying whether there is residual carcinoma in remaining suspicious calcifications after neoadjuvant chemotherapy (NAC) in breast cancer patients can provide crucial information for surgeons in determining the most appropriate surgical approach. Therefore, we investigated factors predicting calcifications without residual carcinoma (ypCalc_0) or with residual carcinoma (ypCalc_ca) and aimed to develop a prediction model for patients exhibiting residual suspicious calcifications on mammography but complete response on MRI after NAC. METHODS This retrospective study included breast cancer patients undergoing NAC, showing residual suspicious mammographic calcifications but complete response on MRI between January 2019 and December 2020 (development set) and between January 2021 and December 2022 (validation set). Multivariable logistic regression analysis identified significant factors associated with ypCalc_0. The prediction model, developed using a decision tree and factors from logistic regression analysis, was validated in the validation set. RESULTS The development set included 134 women (mean age, 50.6 years; 91 with ypCalc_0 and 43 with ypCalc_ca) and validation set included 146 women (mean age, 51.0 years; 108 with ypCalc_0 and 38 with ypCalc_ca). Molecular subtype (P = .0002) and high Ki-67 (P = .02) emerged as significant independent factors associated with ypCalc_0 in the development set. The prediction model, incorporating hormone receptor (HR)-/human epidermal growth factor receptor 2 (HER2)+ with high Ki-67 as ypCalc_0 predictors, and HR+/HER2- cancers or HR+/HER2+ or triple-negative (TN) cancers with low Ki-67, as ypCalc_ca predictors, achieved an area under receiver operating characteristic curve of 0.844 (95% CI 0.774-0.914) in the validation set. CONCLUSION Minimized surgery may be considered for managing residual calcifications in HR-/HER2+ with high Ki-67 cancers, while complete excision is recommended for HR+/HER2- breast cancers or for HR+/HER2+or TN breast cancers with low Ki-67.
Collapse
Affiliation(s)
- Mi-Ri Kwon
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Young Ko
- Department of Radiology and Center for Imaging Science, Samsung Medical Center,, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Jeong Eon Lee
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Boo-Kyung Han
- Department of Radiology and Center for Imaging Science, Samsung Medical Center,, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Sook Ko
- Department of Radiology and Center for Imaging Science, Samsung Medical Center,, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Soo Choi
- Department of Radiology and Center for Imaging Science, Samsung Medical Center,, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Haejung Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center,, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myoung Kyoung Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center,, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jonghan Yu
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyunwoo Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Inyoung Youn
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
5
|
Kim EY, Lee KH, Yun JS, Park YL, Park CH, Jang SY, Ryu JM, Lee SK, Chae BJ, Lee JE, Kim SW, Nam SJ, Yu JH. Impact of residual microcalcifcations on prognosis after neoadjuvant chemotherapy in breast cancer patients. BMC Womens Health 2024; 24:187. [PMID: 38509531 PMCID: PMC10956337 DOI: 10.1186/s12905-024-02973-9] [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: 06/29/2023] [Accepted: 02/14/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Residual microcalcifications after neoadjuvant chemotherapy (NAC) are challenging for deciding extent of surgery and questionable for impact on prognosis. We investigated changes in the extent and patterns of microcalcifications before and after NAC and correlated them with pathologic response. We also compared prognosis of patients depending on presence of residual microcalcifications after NAC. METHODS A total of 323 patients with invasive breast carcinoma treated with neoadjuvant chemotherapy at Kangbuk Samsung Hospital and Samsung Medical center from March 2015 to September 2018 were included. Patients were divided into four groups according to pathologic response and residual microcalcifications. Non-pCRw/mic group was defined as breast non-pCR with residual microcalcifications. Non-pCRw/o mic group was breast non-pCR without residual microcalcifications. pCRw/mic group was breast pCR with residual microcalcifications. pCRw/o mic group was breast pCR without residual microcalcifications. The first aim of this study is to investigate changes in the extent and patterns of microcalcifications before and after NAC and to correlate them with pathologic response. The second aim is to evaluate oncologic outcomes of residual microcalcifications according to pathologic response after NAC. RESULTS There were no statistical differences in the extent, morphology, and distribution of microcalcifications according to pathologic response and subtype after NAC (all p > 0.05). With a median follow-up time of 71 months, compared to pCRw/o mic group, the hazard ratios (95% confidence intervals) for regional recurrence were 5.190 (1.160-23.190) in non-pCRw/mic group and 5.970 (1.840-19.380) in non-pCRw/o mic group. Compared to pCRw/o mic group, the hazard ratios (95% CI) for distant metastasis were 8.520 (2.130-34.090) in non-pCRw/mic group, 9.120 (2.850-29.200) in non-pCRw/o mic group. Compared to pCRw/o mic, the hazard ratio (95% CI) for distant metastasis in pCRw/mic group was 2.240 (0.230-21.500) without statistical significance (p = 0.486). CONCLUSIONS Regardless of residual microcalcifications, patients who achieved pCR showed favorable long term outcome compared to non-pCR group.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kwan Ho Lee
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ji-Sup Yun
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yong Lai Park
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Chan Heun Park
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Yoon Jang
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Jai Min Ryu
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Se Kyung Lee
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Byung-Joo Chae
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Jeong Eon Lee
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Seok Won Kim
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Seok Jin Nam
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Jong Han Yu
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
6
|
Zhu C, Chen M, Liu Y, Li P, Ye W, Ye H, Ye Y, Liu Z, Liang C, Liu C. Value of mammographic microcalcifications and MRI-enhanced lesions in the evaluation of residual disease after neoadjuvant therapy for breast cancer. Quant Imaging Med Surg 2023; 13:5593-5604. [PMID: 37711784 PMCID: PMC10498223 DOI: 10.21037/qims-22-1170] [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: 10/27/2022] [Accepted: 07/17/2023] [Indexed: 09/16/2023]
Abstract
Background Microcalcifications persist even if a patient with breast cancer achieves pathologic complete response (pCR) as confirmed by surgery after neoadjuvant treatment (NAT). In practice, surgeons tend to remove all the microcalcifications. This study aimed to explore the correlation between changes in the extent of microcalcification after NAT and pathological tumor response and compare the accuracy of mammography (MG) and magnetic resonance imaging (MRI) in predicting the size of residual tumors. Methods This was a retrospective study which included a consecutive series of patients in Guangdong Provincial People's Hospital. Between January 2010 and January 2020, 127 patients with breast cancer and Breast Imaging Reporting and Data System (BI-RADS) 4-5 microcalcifications were included in this study. The maximum diameter of the microcalcifications on MG and lesion enhancement on MRI pre- and post-NAT were measured. The correlations between the changes in residual microcalcifications on MG and pCR were analyzed. Intraclass correlation coefficients (ICCs) were computed between the extent of the residual microcalcifications, residual enhancement, and residual tumor size. Results There were no statistically significant differences in the changes in microcalcifications after NAT according to the RECIST criteria on MRI (P=0.09) and Miller-Payne grade (P=0.14). MRI showed a higher agreement than did residual microcalcifications on MG in predicting residual tumor size (ICC: 0.771 vs. 0.097). Conclusions MRI is more accurate for evaluating residual tumor size in breast cancer. In our study, the extent of microcalcifications on MG after NAT had nearly no correlation with the pathological size of the residual tumor. Therefore, residual tumors with microcalcifications may not necessarily be a contraindication to breast-conserving surgery.
Collapse
Affiliation(s)
- Chao Zhu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
- Department of Radiology, Ningyuan County People’s Hospital, Yongzhou, China
| | - Minglei Chen
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yulin Liu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Pinxiong Li
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Weitao Ye
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Huifen Ye
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Yunrui Ye
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Chunling Liu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| |
Collapse
|
7
|
Wang Y, Zhao M, Ma Y, Liu A, Zhu Y, Yin L, Liang Z, Qu Z, Lu H, Ma Y, Ye Z. Accuracy of Preoperative Contrast-enhanced Cone Beam Breast CT in Assessment of Residual Tumor after Neoadjuvant Chemotherapy: A Comparative Study with Breast MRI. Acad Radiol 2023; 30:1805-1815. [PMID: 36610931 DOI: 10.1016/j.acra.2022.12.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023]
Abstract
RATIONALE AND OBJECTIVES To compare the accuracy of preoperative contrast-enhanced cone beam breast CT (CE-CBBCT) and MRI in assessment of residual tumor after neoadjuvant chemotherapy (NAC). MATERIALS AND METHODS Residual tumor assessments in 91 female patients were performed on preoperative CE-CBBCT and MRI images after NAC. The agreements of tumor size between imaging and pathology were tested by Intraclass Correlation Coefficient (ICC). Subgroup analyses were set according to ductal carcinoma in situ (DCIS), calcifications and molecular subtypes. Correlated-samples Wilcoxon Signed-rank test was used to analyze the difference between imaging and pathology in total and subgroups. AUC, sensitivity, specificity, PPV, and NPV were calculated to compare the performance of CE-CBBCT and MRI in predicting pathological complete response (pCR). RESULTS Comparing with pathology, the agreement on CE-CBBCT was good (ICC = 0.64, 95% CI, 0.35-0.78), whereas on MRI was moderate (ICC = 0.59, 95% CI, 0.36-0.77), and overestimation on CE-CBBCT was less than that on MRI (median (interquartile range, IQR): 0.24 [0.00, 1.31] cm vs. 0.67 [0.00, 1.81] cm; p = 0.000). In subgroup analysis, CE-CBBCT showed superior accuracy than MRI when residual DCIS (p = 0.000) and calcifications (p = 0.000) contained, as well as luminal A (p = 0.043) and luminal B (p = 0.009) breast cancer. CE-CBBCT and MRI performed comparable in predicting pCR, AUCs were 0.749 and 0.733 respectively (p > 0.05). CONCLUSION CE-CBBCT showed superior accuracy in assessment of residual tumor compared with MRI, especially when residual DCIS or calcifications contained and luminal subtype. The performance of preoperative CE-CBBCT in predicting pCR was comparable to MRI. CE-CBBCT could be an alternative method used for preoperative assessment after NAC.
Collapse
Affiliation(s)
- Yafei Wang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Mengran Zhao
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yue Ma
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Aidi Liu
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yueqiang Zhu
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Lu Yin
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhiran Liang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zhiye Qu
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Hong Lu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ying Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China..
| |
Collapse
|
8
|
Ploumen RAW, de Mooij CM, Gommers S, Keymeulen KBMI, Smidt ML, van Nijnatten TJA. Imaging findings for response evaluation of ductal carcinoma in situ in breast cancer patients treated with neoadjuvant systemic therapy: a systematic review and meta-analysis. Eur Radiol 2023; 33:5423-5435. [PMID: 37020070 PMCID: PMC10326113 DOI: 10.1007/s00330-023-09547-7] [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: 09/01/2022] [Revised: 12/23/2022] [Accepted: 02/23/2023] [Indexed: 04/07/2023]
Abstract
OBJECTIVES In approximately 45% of invasive breast cancer (IBC) patients treated with neoadjuvant systemic therapy (NST), ductal carcinoma in situ (DCIS) is present. Recent studies suggest response of DCIS to NST. The aim of this systematic review and meta-analysis was to summarise and examine the current literature on imaging findings for different imaging modalities evaluating DCIS response to NST. More specifically, imaging findings of DCIS pre- and post-NST, and the effect of different pathological complete response (pCR) definitions, will be evaluated on mammography, breast MRI, and contrast-enhanced mammography (CEM). METHODS PubMed and Embase databases were searched for studies investigating NST response of IBC, including information on DCIS. Imaging findings and response evaluation of DCIS were assessed for mammography, breast MRI, and CEM. A meta-analysis was conducted per imaging modality to calculate pooled sensitivity and specificity for detecting residual disease between pCR definition no residual invasive disease (ypT0/is) and no residual invasive or in situ disease (ypT0). RESULTS Thirty-one studies were included. Calcifications on mammography are related to DCIS, but can persist despite complete response of DCIS. In 20 breast MRI studies, an average of 57% of residual DCIS showed enhancement. A meta-analysis of 17 breast MRI studies confirmed higher pooled sensitivity (0.86 versus 0.82) and lower pooled specificity (0.61 versus 0.68) for detection of residual disease when DCIS is considered pCR (ypT0/is). Three CEM studies suggest the potential benefit of simultaneous evaluation of calcifications and enhancement. CONCLUSIONS AND CLINICAL RELEVANCE Calcifications on mammography can remain despite complete response of DCIS, and residual DCIS does not always show enhancement on breast MRI and CEM. Moreover, pCR definition effects diagnostic performance of breast MRI. Given the lack of evidence on imaging findings of response of the DCIS component to NST, further research is demanded. KEY POINTS • Ductal carcinoma in situ has shown to be responsive to neoadjuvant systemic therapy, but imaging studies mainly focus on response of the invasive tumour. • The 31 included studies demonstrate that after neoadjuvant systemic therapy, calcifications on mammography can remain despite complete response of DCIS and residual DCIS does not always show enhancement on MRI and contrast-enhanced mammography. • The definition of pCR has impact on the diagnostic performance of MRI in detecting residual disease, and when DCIS is considered pCR, pooled sensitivity was slightly higher and pooled specificity slightly lower.
Collapse
Affiliation(s)
- Roxanne A W Ploumen
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands.
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands.
| | - Cornelis M de Mooij
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Suzanne Gommers
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Marjolein L Smidt
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Thiemo J A van Nijnatten
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
9
|
Hu Y, Mao L, Wang M, Li Z, Li M, Wang C, Ji L, Zeng H, Zhang X. New insights into breast microcalcification for poor prognosis: NACT cohort and bone metastasis evaluation cohort. J Cancer Res Clin Oncol 2023; 149:7285-7297. [PMID: 36917189 DOI: 10.1007/s00432-023-04668-4] [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: 10/05/2022] [Accepted: 02/23/2023] [Indexed: 03/15/2023]
Abstract
OBJECTIVES The study aimed to analyze the poor prognosis of microcalcification in breast cancer (BC), including the pathological complete response (pCR) to neoadjuvant chemotherapy (NACT) and the risk of bone metastases. MATERIALS AND METHODS 313 breast cancer patients received NACT to evaluate pCR and 1182 patients from a multicenter database to assess bone metastases were retrospectively included. Two groups were divided according to the presence or absence of mammography microcalcification. Clinical data, image characteristics, neoadjuvant treatment response, bone involvement, and follow-up information were recorded. The pCR and bone metastases were compared between subgroups using the Mann-Whitney and χ2 tests and logistic regression, respectively. RESULTS Mammographic microcalcification was associated with a lower pCR than uncalcified BC in the NACT cohort (20.6% vs 31.6%, P = 0.029). Univariate and multivariate analysis suggested that calcification was a risk factor for poor NACT response [OR = 1.780, 95%CI (1.065-2.974), P = 0.028], [OR = 2.352, 95%CI (1.186-4.667), P = 0.014]. Microcalcification was more likely to be necrosis on MRI than those without microcalcification (53.0% vs 31.7%, P < 0.001), multivariate analysis indicated that tumor necrosis was also a risk factor for poor NACT response [OR = 2.325, 95%CI (1.100-4.911), P = 0.027]. Age, menopausal status, breast density, mass, molecular, and pathology type were not significantly associated with non-pCR risk assessment. In a multicenter cohort of 1182 patients with pathologically confirmed BC, those with microcalcifications had a higher proportion of bone metastases compared to non-calcified BC (11.6% vs 4.9%, P < 0.001). Univariate and multivariate analysis showed that microcalcification was an independent risk factor for bone metastasis [OR = 2.550, 95%CI (1.620-4.012), P < 0.001], [OR = 2.268(1.263-4.071), P = 0.006)]. Osteolytic bone metastases predominated but there was no statistical difference between the two groups (78.9% vs 60.7%, P = 0.099). Calcified BC was mainly involved in axial bone, but was more likely to involve the whole-body bone than non-calcified BC (33.8% vs 10.7%, P = 0.021). CONCLUSION This study provides important insights into the poor prognosis of microcalcification, not only in terms of poor response to NACT but also the risk factor of bone metastases.
Collapse
Affiliation(s)
- Yangling Hu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lijuan Mao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mengyi Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhenqiu Li
- Department of Radiology, The Panyu Fifth Hospital, Guangzhou, China
| | - Meizhi Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaoyang Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lin Ji
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui Zeng
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Xiaoling Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
10
|
Azam R, Lim D, Curpen B, Mulligan AM, Hong NL. Correlation of Mammographic Microcalcifications with Final Surgical Pathology After Neoadjuvant Chemotherapy for Breast Cancer. Ann Surg Oncol 2023:10.1245/s10434-023-13367-w. [PMID: 37029866 DOI: 10.1245/s10434-023-13367-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/27/2023] [Indexed: 04/09/2023]
Abstract
INTRODUCTION Imaging guidelines for post-neoadjuvant chemotherapy (NAC) breast cancer patients lack specificity on appropriateness and utility of individual modalities for surgical planning. Microcalcifications confound mammographic interpretation. We examined the correlation between the mammographic extent of microcalcifications present post-NAC, corresponding magnetic resonance imaging (MRI) lesions, and definitive surgical pathology. METHODS In this retrospective cohort study, patients with calcifications on mammography were collected from a database of consecutive breast cancer patients receiving NAC. The primary objective was to determine the correlation between maximum dimension of post-NAC calcifications with surgical pathology (invasive disease, tumor bed, and ductal carcinoma in situ [DCIS]), stratified by tumor receptor subgroup. Secondarily, we examined the correlation of residual disease with MRI mass enhancement (ME) and non-ME (NME). Pearson's correlation coefficient was used to evaluate statistical significance (strong: R2 ≥70%; moderate: R2=25-70%; weak: R2 ≤25%). RESULTS Overall, 186 patients met the inclusion criteria. Mammographic calcifications correlated poorly with invasive disease (R2 = 10.8%), overestimating by 57%. In patients with calcifications on mammography, MRI ME and NME correlated weakly with the maximum dimension of invasive disease and DCIS. In triple-negative breast cancer (TNBC) patients, invasive disease correlated strongly with the maximum dimension of calcifications (R2 = 83%) and moderately with ME (R2 = 37.7%) and NME (R2 = 28.4%). CONCLUSION Overall, current imaging techniques correlate poorly and overestimate final surgical pathology. This poor correlation may lead to uncertainty in the extent of required surgical excision and the exclusion of potential candidates for non-surgical management in ongoing trials. TNBCs would be good candidates for these trials given the stronger observed correlations between pathology and imaging.
Collapse
Affiliation(s)
- Riordan Azam
- PGME University of Toronto, Toronto, ON, Canada.
| | - David Lim
- PGME University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|
11
|
Hogan MP, Horvat JV, Ross DS, Sevilimedu V, Jochelson MS, Kirstein LJ, Goldfarb SB, Comstock CE, Sung JS. Contrast-enhanced mammography in the assessment of residual disease after neoadjuvant treatment. Breast Cancer Res Treat 2023; 198:349-359. [PMID: 36754936 PMCID: PMC10375516 DOI: 10.1007/s10549-023-06865-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/19/2023] [Indexed: 02/10/2023]
Abstract
PURPOSE To investigate the utility of contrast-enhanced mammography (CEM) as an alternative to breast MRI for the evaluation of residual disease after neoadjuvant treatment (NAT). METHODS This prospective study enrolled consecutive women undergoing NAT for breast cancer from July 2017-July 2019. Breast MRI and CEM exams performed after completion of NAT were read independently by two breast radiologists. Residual disease and lesion size on MRI and CEM recombined (RI) and low-energy images (LEI) were compared. Histopathology was considered the reference standard. Statistical analysis was performed using McNemar's and Leisenring's tests. Multiple comparison adjustment was made using Bonferroni procedure. Lesion sizes were correlated using Kendall's tau coefficient. RESULTS There were 110 participants with 115 breast cancers. Residual disease (invasive cancer or ductal carcinoma in situ) was detected in 83/115 (72%) lesions on pathology, 71/115 (62%) on MRI, 55/115 (48%) on CEM RI, and 75/115 (65%) on CEM LEI. When using multiple comparison adjustment, no significant differences were detected between MRI combined with CEM LEI and CEM RI combined with CEM LEI, in terms of accuracy (MRI: 77%, CEM: 72%; p ≥ 0.99), sensitivity (MRI: 88%, CEM: 81%; p ≥ 0.99), specificity (MRI: 47%, CEM: 50%; p ≥ 0.99), PPV (MRI: 81%, CEM: 81%; p ≥ 0.99), or NPV (MRI: 60%, CEM: 50%; p ≥ 0.99). Size correlation between pathology and both MRI combined with CEM LEI and CEM RI combined with CEM LEI was moderate: τ = 0. 36 vs 0.33 (p ≥ 0.99). CONCLUSION Contrast-enhanced mammography is an acceptable alternative to breast MRI for the detection of residual disease after neoadjuvant treatment.
Collapse
Affiliation(s)
- Molly P Hogan
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Joao V Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Varadan Sevilimedu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10017, USA
| | - Maxine S Jochelson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Laurie J Kirstein
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Shari B Goldfarb
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Christopher E Comstock
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Janice S Sung
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
12
|
Surgical Planning after Neoadjuvant Treatment in Breast Cancer: A Multimodality Imaging-Based Approach Focused on MRI. Cancers (Basel) 2023; 15:cancers15051439. [PMID: 36900231 PMCID: PMC10001061 DOI: 10.3390/cancers15051439] [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: 01/10/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Neoadjuvant chemotherapy (NACT) today represents a cornerstone in the treatment of locally advanced breast cancer and highly chemo-sensitive tumors at early stages, increasing the possibilities of performing more conservative treatments and improving long term outcomes. Imaging has a fundamental role in the staging and prediction of the response to NACT, thus aiding surgical planning and avoiding overtreatment. In this review, we first examine and compare the role of conventional and advanced imaging techniques in preoperative T Staging after NACT and in the evaluation of lymph node involvement. In the second part, we analyze the different surgical approaches, discussing the role of axillary surgery, as well as the possibility of non-operative management after-NACT, which has been the subject of recent trials. Finally, we focus on emerging techniques that will change the diagnostic assessment of breast cancer in the near future.
Collapse
|
13
|
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
|
14
|
Provenzano E, Shaaban AM. Pathology of neoadjuvant therapy and immunotherapy testing for breast cancer. Histopathology 2023; 82:170-188. [PMID: 36482270 DOI: 10.1111/his.14771] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 12/13/2022]
Abstract
Neoadjuvant chemotherapy (NACT) has become the standard of care for high-risk breast cancer, including triple-negative (TNBC) and HER2-positive disease. As a result, handling and reporting of breast specimens post-NACT is part of routine practice, and it is important for pathologists to recognise the changes in tumour cells, tumour-associated stroma and background breast tissue induced by NACT. Familiarity with characteristic stromal features enables identification of the pre-treatment tumour site and allows confident diagnosis of pathological complete response (pCR) which is important for decisions concerning adjuvant therapy. Neoadjuvant endocrine therapy (NAET) is used less frequently than NACT; however, the SARS-COVID-19 pandemic has changed practice, with increased use as bridging therapy if surgery is delayed. NAET also induces characteristic changes in the tumour and stroma. Changes in the tumour microenvironment following NACT and NAET are also described. Immunotherapy is approved for use in advanced TNBC, and there are several trials exploring its role in early TNBC in the neoadjuvant setting. The current biomarker to determine eligibility for treatment with immune checkpoint inhibitors is programmed death ligand-1 (PD-L1) immunohistochemistry; however, this is complicated by lack of standardisation with different drugs linked to tests using different antibodies with different scoring systems. The situation in the neoadjuvant setting is further complicated by improved pCR rates for PD-L1-positive tumours in both immune therapy and placebo arms. Alternative biomarkers are urgently needed to identify which patients will derive benefit from immunotherapy and key candidates are discussed.
Collapse
Affiliation(s)
- Elena Provenzano
- Department of Histopathology, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Abeer M Shaaban
- Queen Elizabeth Hospital Birmingham and University of Birmingham, Birmingham, UK
| |
Collapse
|
15
|
Lee J, Park NJY, Park HY, Kim WW, Kang B, Keum H, Kim HJ, Kim WH, Chae YS, Lee SJ, Lee IH, Park JY, Jung JH. Oncologic necessity for the complete removal of residual microcalcifications after neoadjuvant chemotherapy for breast cancer. Sci Rep 2022; 12:21535. [PMID: 36513704 PMCID: PMC9748126 DOI: 10.1038/s41598-022-24757-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
The surgical range of breast cancer that shows pathologic complete response (pCR) without change in microcalcifications after neoadjuvant chemotherapy (NAC) is controversial. This study examined whole breast specimens to evaluate the necessity of mastectomy in those cases. The viability of cancer cells around the residual microcalcification was assessed using prospectively collected breast samples to confirm the presence or absence of cancer cells. A total of 144 patients with breast cancer and diffuse microcalcifications were classified into the reduced mass with no change in residual microcalcification (RESMIN, n = 49) and non-RESMIN (n = 95) groups. Five specimens were prospectively evaluated to assess the presence of viable cancer cells around the microcalcification. Tumor responses to NAC were significantly better with high pCR rates in the RESMIN group (p = 0.005 and p = 0.002). The incidence of human epidermal growth factor receptor 2-positive and triple-negative breast cancers was significantly high in the RESMIN group (p = 0.007). Although five (10.2%) patients had locoregional recurrence in the RESMIN group, no local recurrence in the breast was reported. Although pCR was highly estimated, residual cancers, including ductal carcinoma in situ, remained in 80% cases. Therefore, given the weak scientific evidence available currently, complete removal of residual microcalcifications should be considered for oncologic safety.
Collapse
Affiliation(s)
- Jeeyeon Lee
- grid.258803.40000 0001 0661 1556Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Nora Jee-Young Park
- grid.258803.40000 0001 0661 1556Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ho Yong Park
- grid.258803.40000 0001 0661 1556Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Wan Wook Kim
- grid.258803.40000 0001 0661 1556Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Byeongju Kang
- grid.258803.40000 0001 0661 1556Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Heejung Keum
- grid.258803.40000 0001 0661 1556Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Hye Jung Kim
- grid.258803.40000 0001 0661 1556Department of Radiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Won Hwa Kim
- grid.258803.40000 0001 0661 1556Department of Radiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Yee Soo Chae
- grid.258803.40000 0001 0661 1556Department of Hematology/Oncology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Soo Jung Lee
- grid.258803.40000 0001 0661 1556Department of Hematology/Oncology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - In Hee Lee
- grid.258803.40000 0001 0661 1556Department of Hematology/Oncology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji-Young Park
- grid.258803.40000 0001 0661 1556Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Jin Hyang Jung
- grid.258803.40000 0001 0661 1556Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea ,grid.258803.40000 0001 0661 1556Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| |
Collapse
|
16
|
Locoregional Management of Breast Cancer Following Neoadjuvant Chemotherapy. CURRENT BREAST CANCER REPORTS 2022. [DOI: 10.1007/s12609-022-00452-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Nakhlis F, Portnow L, Gombos E, Daylan AEC, Leone JP, Kantor O, Richardson ET, Ho A, Dunn SA, Ohri N. Multidisciplinary Considerations in the Management of Breast Cancer Patients Receiving Neoadjuvant Chemotherapy. Curr Probl Surg 2022; 59:101191. [DOI: 10.1016/j.cpsurg.2022.101191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
18
|
Kong X, Zhang Q, Wu X, Zou T, Duan J, Song S, Nie J, Tao C, Tang M, Wang M, Zou J, Xie Y, Li Z, Li Z. Advances in Imaging in Evaluating the Efficacy of Neoadjuvant Chemotherapy for Breast Cancer. Front Oncol 2022; 12:816297. [PMID: 35669440 PMCID: PMC9163342 DOI: 10.3389/fonc.2022.816297] [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: 11/16/2021] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) is increasingly widely used in breast cancer treatment, and accurate evaluation of its response provides essential information for treatment and prognosis. Thus, the imaging tools used to quantify the disease response are critical in evaluating and managing patients treated with NAC. We discussed the recent progress, advantages, and disadvantages of common imaging methods in assessing the efficacy of NAC for breast cancer.
Collapse
Affiliation(s)
- Xianshu Kong
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Qian Zhang
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Xuemei Wu
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Tianning Zou
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Jiajun Duan
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Shujie Song
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Jianyun Nie
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Chu Tao
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Mi Tang
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Maohua Wang
- First Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Jieya Zou
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Yu Xie
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Zhen Li
- Third Department of the Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| |
Collapse
|
19
|
Lee SC, Mendez-Broomberg K, Eacobacci K, Vincoff NS, Gupta E, McElligott SE. Nipple-sparing Mastectomy: What the Radiologist Should Know. Radiographics 2022; 42:321-339. [PMID: 35179983 DOI: 10.1148/rg.210136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nipple-sparing mastectomy (NSM) is increasingly offered to patients undergoing treatment of breast cancer and prophylaxis treatment for reduction of breast cancer risk. NSM is considered oncologically safe for appropriately selected patients and is associated with improved cosmetic outcomes and quality of life. Accepted indications for NSM have expanded in recent years, and currently only inflammatory breast cancer or malignancy involving the nipple is considered an absolute contraindication. Neoplasms close to and involving the nipple areolar complex are common, and cancer of the lactiferous ducts can spread to the nipple. Therefore, accurate determination of nipple involvement at imaging examinations is critical to identifying appropriate candidates for NSM and preventing local recurrence. Multiple imaging features have been described as predictors of nipple involvement, with tumor to nipple distance, enhancement between the index malignancy and the nipple, and nipple retraction demonstrating the highest predictive values. These features can be assessed at multimodality breast imaging, particularly at breast MRI, which demonstrates high specificity and negative predictive value for determining nipple involvement in malignancy. Online supplemental material is available for this article. ©RSNA, 2022.
Collapse
Affiliation(s)
- Samantha C Lee
- From the Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra University/Northwell Health System, 300 Community Dr, Manhasset, NY 11030
| | - Karen Mendez-Broomberg
- From the Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra University/Northwell Health System, 300 Community Dr, Manhasset, NY 11030
| | - Katherine Eacobacci
- From the Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra University/Northwell Health System, 300 Community Dr, Manhasset, NY 11030
| | - Nina S Vincoff
- From the Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra University/Northwell Health System, 300 Community Dr, Manhasset, NY 11030
| | - Ekta Gupta
- From the Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra University/Northwell Health System, 300 Community Dr, Manhasset, NY 11030
| | - Suzanne E McElligott
- From the Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra University/Northwell Health System, 300 Community Dr, Manhasset, NY 11030
| |
Collapse
|
20
|
Logullo A, Prigenzi K, Nimir C, Franco A, Campos M. Breast microcalcifications: Past, present and future (Review). Mol Clin Oncol 2022; 16:81. [PMID: 35251632 PMCID: PMC8892454 DOI: 10.3892/mco.2022.2514] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/19/2021] [Indexed: 11/08/2022] Open
Abstract
Mammary microcalcifications (MCs) are calcium deposits that are considered as robust markers of breast cancer when identified on mammography. MCs are frequently associated with premalignant and malignant lesions. The aim of the present review was to describe the MC types and associated radiological and pathological aspects in detail, provide insights and approaches to the topic, and describe specific clinical scenarios. The primary MC types are composed of calcium oxalate, hydroxyapatite and hydroxyapatite associated with magnesium. The first type is usually associated with benign conditions, while the others remain primarily associated with malignancy. Radiologically, MCs are classified as benign or suspicious. MCs may represent an active pathological mineralization process rather than a passive process, such as degeneration or necrosis. Practical management of breast specimens requires finely calibrated radiological pathological procedures. Understanding the molecular and structural development of MCs may contribute to breast lesion detection and treatment.
Collapse
Affiliation(s)
- Angela Logullo
- Department of Pathology, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo 04023‑062, Brazil
| | - Karla Prigenzi
- Department of Pathology, Femme Laboratories, São Paulo 04004‑030, Brazil
| | - Cristiane Nimir
- Department of Pathology, Femme Laboratories, São Paulo 04004‑030, Brazil
| | - Andreia Franco
- Department of Pathology, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo 04023‑062, Brazil
| | - Mario Campos
- Breast Imaging Service, Femme Laboratories, São Paulo 04004‑030, Brazil
| |
Collapse
|
21
|
Murakami R, Tani H, Kumita S, Uchiyama N. Diagnostic performance of digital breast tomosynthesis for predicting response to neoadjuvant systemic therapy in breast cancer patients: A comparison with magnetic resonance imaging, ultrasound, and full-field digital mammography. Acta Radiol Open 2022; 10:20584601211063746. [PMID: 34992793 PMCID: PMC8725236 DOI: 10.1177/20584601211063746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Background The goals of neoadjuvant systemic therapy (NST) are to reduce tumor volume
and to provide a prognostic indicator in assessing treatment response.
Digital breast tomosynthesis (DBT) was developed and has increased interest
in clinical settings due to its higher sensitivity for breast cancer
detection compared to full-field digital mammography (FFDM). Purpose To evaluate the accuracy of DBT in assessing response to NST compared to
FFDM, ultrasound (US), and magnetic resonance imaging (MRI) in breast cancer
patients. Material and Methods In this retrospective study, 95 stages II–III breast cancer patients
undergoing NST and subsequent surgeries were enrolled. After NST, the
longest diameter of residual tumor measured by DBT, FFDM, US, and MRI was
compared with pathology. Agreements and correlations of tumor size were
assessed, and the diagnostic performance for predicting pathologic complete
response (pCR) was evaluated. Results Mean residual tumor size after NST was 19.9 mm for DBT, 18.7 mm for FFDM,
16.0 mm for US, and 18.4 mm for MRI, compared with 17.9 mm on pathology. DBT
and MRI correlated better with pathology than that of FFDM and US. The ICC
values were 0.85, 0.87, 0.74, and 0.77, respectively. Twenty-five patients
(26.3%) achieved pCR after NST. For predicting pCR, area under the receiver
operating characteristic (ROC) curve for DBT, FFDM, US, and MRI were 0.79,
0.66, 0.68, and 0.77, respectively. Conclusion DBT has good correlation with histopathology for measuring residual tumor
size after NST. DBT was comparable to MRI in assessing tumor response after
completion of NST.
Collapse
Affiliation(s)
- Ryusuke Murakami
- Department of Radiology, Nippon Medical School Hospital, Tokyo, Japan
| | - Hitomi Tani
- Department of Radiology, Nippon Medical School Hospital, Tokyo, Japan
| | - Shinichiro Kumita
- Department of Radiology, Nippon Medical School Hospital, Tokyo, Japan
| | | |
Collapse
|
22
|
Hadar T, Koretz M, Nawass M, Allweis TM. Innovative Standards in Surgery of the Breast after Neoadjuvant Systemic Therapy. Breast Care (Basel) 2021; 16:590-597. [PMID: 35087362 PMCID: PMC8739938 DOI: 10.1159/000520051] [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: 06/07/2021] [Accepted: 09/29/2021] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND The goal of neoadjuvant systemic therapy (NST) in breast cancer is to downstage tumors and downgrade treatment. Indications are constantly evolving. These changes raise practical questions for planning of surgery after NST. SUMMARY In this review we discuss current evolving aspects of surgery of the breast after NST. Breast-conserving surgery (BCS) eligibility increases after NST - both neoadjuvant chemotherapy (NAC) and neoadjuvant endocrine therapy. Adequate margin width in NST and upfront surgery are similar - "no tumor on ink" for invasive cancer. Oncoplastic breast surgery after NST is feasible - both for BCS and mastectomy with reconstruction. There is increasing interest in the possibility of omitting surgery in patients with a complete response to NAC. Several trials are being conducted in aim of achieving acceptable prediction of pathological complete response, by combination of imaging and percutaneous biopsy of the tumor bed, as well as assessing the safety of such an approach. KEY MESSAGES Surgery of the breast after NST should be determined not only according to biologic and anatomic parameters at diagnosis, but is dynamic, and must be tailored according to the response to therapy. The omission of surgery in exceptional responders after NAC is being explored.
Collapse
Affiliation(s)
- Tal Hadar
- Department of Breast Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Michael Koretz
- Department of Breast Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Mahmood Nawass
- Department of Breast Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Tanir M. Allweis
- Department of Breast Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
23
|
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
|
24
|
Cao L, Sugumar K, Keller E, Li P, Rock L, Simpson A, Freyvogel M, Montero AJ, Shenk R, Miller ME. Neoadjuvant Endocrine Therapy as an Alternative to Neoadjuvant Chemotherapy Among Hormone Receptor-Positive Breast Cancer Patients: Pathologic and Surgical Outcomes. Ann Surg Oncol 2021; 28:5730-5741. [PMID: 34342757 PMCID: PMC8330206 DOI: 10.1245/s10434-021-10459-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/22/2021] [Indexed: 12/22/2022]
Abstract
Background Neoadjuvant chemotherapy (NCT) is considered more effective in downstaging hormone receptor-positive (HR+) breast cancer than neoadjuvant endocrine therapy (NET), particularly in node-positive disease. This study compared breast and axillary response and survival after NCT and NET in HR+ breast cancer. Methods Based on American College of Surgeons Oncology Group (ACOSOG) Z1031 criteria, women age 50 years or older with cT2-4 HR+ breast cancer who underwent NET or NCT and surgery were identified in the National Cancer Database 2010–2016. Chi-square and logistic regression analysis determined differences between the NCT and NET groups and therapy response, including downstaging and pathologic complete response (pCR, ypT0/is and ypN0). Results Of 19,829 patients, 14,025 (70.7%) received NCT and 5804 (29.3%) received NET. The NET patients were older (mean age, 68.9 vs. 60.3; P < 0.001) and had greater comorbidity (1+ Charlson–Deyo score, 21% vs. 16%; P < 0.001). Therapy achieved T downstaging (any) for 58% of the patients with NCT versus 40.5% of the patients with NET, and in-breast pCR was achieved for 9.3% of the NCT versus 1.3% of the NET patients (P < 0.001). Approximately half of the mastectomy procedures could have been potentially avoided for the patients with in-breast pCR (53.6% of the NCT and 43.8% of the NET patients). For the cN+ patients, N downstaging (any) was 29% for the NCT patients versus 18.3% for the NET patients (P < 0.001), and nodal pCR was achieved for 20.3% of the NCT versus 13.5% of the NET patients (P < 0.001). Among those with nodal pCR, axillary lymph node dissection (ALND) still was performed for 56% of the patients after NCT and 45% of the patients after NET. Conclusions Although the response rates after NCT were higher, NET achieved both T and N downstaging and pCR. Neoadjuvant endocrine therapy can be used to de-escalate surgery for patients who cannot tolerate NCT or when chemotherapy may not be effective based on genomic testing. Supplementary Information The online version contains supplementary material available at 10.1245/s10434-021-10459-3.
Collapse
Affiliation(s)
- Lifen Cao
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA
| | - Kavin Sugumar
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA
| | - Eleanor Keller
- University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA.,Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Pamela Li
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA
| | - Lisa Rock
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA
| | - Ashley Simpson
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA
| | - Mary Freyvogel
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA
| | - Alberto J Montero
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Robert Shenk
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA
| | - Megan E Miller
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA. .,University Hospitals Research in Surgical Outcomes and Effectiveness (UH-RISES), Cleveland, OH, USA.
| |
Collapse
|
25
|
Iotti V, Ragazzi M, Besutti G, Marchesi V, Ravaioli S, Falco G, Coiro S, Bisagni A, Gasparini E, Giorgi Rossi P, Vacondio R, Pattacini P. Accuracy and Reproducibility of Contrast-Enhanced Mammography in the Assessment of Response to Neoadjuvant Chemotherapy in Breast Cancer Patients with Calcifications in the Tumor Bed. Diagnostics (Basel) 2021; 11:diagnostics11030435. [PMID: 33806306 PMCID: PMC7999407 DOI: 10.3390/diagnostics11030435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/16/2021] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
This study aimed to evaluate contrast-enhanced mammography (CEM) accuracy and reproducibility in the detection and measurement of residual tumor after neoadjuvant chemotherapy (NAC) in breast cancer (BC) patients with calcifications, using surgical specimen pathology as the reference. Pre- and post-NAC CEM images of 36 consecutive BC patients receiving NAC in 2012–2020, with calcifications in the tumor bed at diagnosis, were retrospectively reviewed by two radiologists; described were absence/presence and size of residual disease based on contrast enhancement (CE) only and CE plus calcifications. Twenty-eight patients (77.8%) had invasive and 5 (13.9%) in situ-only residual disease at surgical specimen pathology. Considering CE plus calcifications instead of CE only, CEM sensitivity for invasive residual tumor increased from 85.7% (95% CI = 67.3–96%) to 96.4% (95% CI = 81.7–99.9%) and specificity decreased from 5/8 (62.5%; 95% CI = 24.5–91.5%) to 1/8 (14.3%; 95% CI = 0.4–57.9%). For in situ-only residual disease, false negatives decreased from 3 to 0 and false positives increased from 1 to 2. CEM pathology concordance in residual disease measurement increased (R squared from 0.38 to 0.45); inter-reader concordance decreased (R squared from 0.79 to 0.66). Considering CE plus calcifications to evaluate NAC response in BC patients increases sensitivity in detection and accuracy in measurement of residual disease but increases false positives.
Collapse
Affiliation(s)
- Valentina Iotti
- Radiology Unit, Department of Imaging and Laboratory Medicine, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (V.I.); (V.M.); (S.R.); (R.V.); (P.P.)
| | - Moira Ragazzi
- Pathology Unit, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (M.R.); (A.B.)
| | - Giulia Besutti
- Radiology Unit, Department of Imaging and Laboratory Medicine, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (V.I.); (V.M.); (S.R.); (R.V.); (P.P.)
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Correspondence: ; Tel.: +39-0522-296369
| | - Vanessa Marchesi
- Radiology Unit, Department of Imaging and Laboratory Medicine, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (V.I.); (V.M.); (S.R.); (R.V.); (P.P.)
| | - Sara Ravaioli
- Radiology Unit, Department of Imaging and Laboratory Medicine, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (V.I.); (V.M.); (S.R.); (R.V.); (P.P.)
| | - Giuseppe Falco
- Breast Surgery Unit, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.F.); (S.C.)
| | - Saverio Coiro
- Breast Surgery Unit, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.F.); (S.C.)
| | - Alessandra Bisagni
- Pathology Unit, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (M.R.); (A.B.)
| | - Elisa Gasparini
- Oncology Unit, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Paolo Giorgi Rossi
- Epidemiology Unit, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Rita Vacondio
- Radiology Unit, Department of Imaging and Laboratory Medicine, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (V.I.); (V.M.); (S.R.); (R.V.); (P.P.)
| | - Pierpaolo Pattacini
- Radiology Unit, Department of Imaging and Laboratory Medicine, Azienda USL—IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (V.I.); (V.M.); (S.R.); (R.V.); (P.P.)
| |
Collapse
|
26
|
Plaza MJ, Cole DA. Could ultrasound-guided cryoablation be used to manage "low-risk" DCIS?: a feasibility case report. Radiol Case Rep 2020; 15:1301-1304. [PMID: 32595816 PMCID: PMC7306537 DOI: 10.1016/j.radcr.2020.05.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 11/29/2022] Open
Abstract
Management of ductal carcinoma in-situ (DCIS) is controversial as there is concern that the majority of diagnoses will never become life threatening such that a subset of patients may be overtreated with surgery. Active surveillance is an alternative proposed management strategy; however, we cannot accurately predict which DCIS will never progress to invasive disease potentially undertreating a large proportion of women. We present a case of a 58-year-old female with DCIS successfully treated with only ultrasound-guided cryoablation without resection. A follow-up needle biopsy of the ablation zone was benign and imaging follow-up has demonstrated no evidence of disease at 14-months. Cryoablation of DCIS is feasible with appropriate patient selection and warrants further investigation as an alternative to surgical resection or active surveillance.
Collapse
Affiliation(s)
- Michael J. Plaza
- Diagnostic Center for Women, LLC, 7500 SW 87th Ave, Suite 100, Miami, FL 33173 USA
| | | |
Collapse
|
27
|
Jones EF, Hathi DK, Freimanis R, Mukhtar RA, Chien AJ, Esserman LJ, van’t Veer LJ, Joe BN, Hylton NM. Current Landscape of Breast Cancer Imaging and Potential Quantitative Imaging Markers of Response in ER-Positive Breast Cancers Treated with Neoadjuvant Therapy. Cancers (Basel) 2020; 12:E1511. [PMID: 32527022 PMCID: PMC7352259 DOI: 10.3390/cancers12061511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Abstract
In recent years, neoadjuvant treatment trials have shown that breast cancer subtypes identified on the basis of genomic and/or molecular signatures exhibit different response rates and recurrence outcomes, with the implication that subtype-specific treatment approaches are needed. Estrogen receptor-positive (ER+) breast cancers present a unique set of challenges for determining optimal neoadjuvant treatment approaches. There is increased recognition that not all ER+ breast cancers benefit from chemotherapy, and that there may be a subset of ER+ breast cancers that can be treated effectively using endocrine therapies alone. With this uncertainty, there is a need to improve the assessment and to optimize the treatment of ER+ breast cancers. While pathology-based markers offer a snapshot of tumor response to neoadjuvant therapy, non-invasive imaging of the ER disease in response to treatment would provide broader insights into tumor heterogeneity, ER biology, and the timing of surrogate endpoint measurements. In this review, we provide an overview of the current landscape of breast imaging in neoadjuvant studies and highlight the technological advances in each imaging modality. We then further examine some potential imaging markers for neoadjuvant treatment response in ER+ breast cancers.
Collapse
Affiliation(s)
- Ella F. Jones
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Deep K. Hathi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Rita Freimanis
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Rita A. Mukhtar
- Department of Surgery, University of California, San Francisco, CA 94115, USA;
| | - A. Jo Chien
- School of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (A.J.C.); (L.J.v.V.)
| | - Laura J. Esserman
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA;
| | - Laura J. van’t Veer
- School of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (A.J.C.); (L.J.v.V.)
| | - Bonnie N. Joe
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| | - Nola M. Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94115, USA; (D.K.H.); (R.F.); (B.N.J.); (N.M.H.)
| |
Collapse
|
28
|
Petruolo O, Sevilimedu V, Montagna G, Le T, Morrow M, Barrio AV. How Often Does Modern Neoadjuvant Chemotherapy Downstage Patients to Breast-Conserving Surgery? Ann Surg Oncol 2020; 28:287-294. [PMID: 32514804 DOI: 10.1245/s10434-020-08593-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) has been proven to increase breast-conserving surgery (BCS) rates, but data are limited on conversion rates from BCS-ineligible (BCSi) to BCS-eligible (BCSe), specifically, in patients with large tumors. METHODS Consecutive patients with stage I-III breast cancer treated with NAC from November 2013 to March 2019 were identified. BCS eligibility before and after NAC was prospectively determined. Patients deemed BCSi before NAC due to large tumor size were studied. Statistical analyses were conducted using Student's t-test, Wilcoxon rank sum test, Chi-square test, Fisher's test, and logistic regression. RESULTS In this study, 600 of 1353 cancers were BCSi with large tumors; 69% were non-BCS candidates, 31% were borderline-BCS (bBCS) candidates. Of non-BCS candidates, 69% became BCSe after NAC; 66% chose BCS, and 90% were successful. Among bBCS candidates, 87% were BCSe after NAC, 73% chose BCS, and 96% were successful. On univariate analysis, bBCS candidacy, lower cT stage, cN0 status, absence of calcifications, human epidermal growth factor receptor 2 positive (HER2+)/triple negative (TN) receptor status, poor differentiation, ductal histology, and breast pCR were associated with conversion to BCS eligibility. On multivariable analysis, receptor status (hormone receptor positive [HR+]/HER2- ref; odds ratio [OR] HER2+ 1.63, P = 0.047; HR-/HER2- OR, 2.26, P = 0.003) and breast pCR (OR 2.62, P < 0.001) predicted successful downstaging, while larger clinical tumor size (OR 0.86, P = 0.003), non-BCS candidacy (OR 0.46, P = 0.003), cN+ status (OR 0.54, P = 0.008), and calcifications (OR 0.56, P = 0.007) predicted lower downstaging rates. CONCLUSION In patients with large tumors precluding BCS, conversion to BCS eligibility was high with NAC, particularly in bBCS candidates. HER2+/TN receptor status predicted successful downstaging, while lower downstaging rates were observed with larger tumors, cN+ status, and calcifications. These factors should be considered when selecting patients for NAC.
Collapse
Affiliation(s)
- Oriana Petruolo
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Varadan Sevilimedu
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giacomo Montagna
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tiana Le
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Monica Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea V Barrio
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
29
|
Morrow M, Khan AJ. Locoregional Management After Neoadjuvant Chemotherapy. J Clin Oncol 2020; 38:2281-2289. [PMID: 32442069 DOI: 10.1200/jco.19.02576] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Monica Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Atif J Khan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
30
|
Prediction of pathologic complete response using image-guided biopsy after neoadjuvant chemotherapy in breast cancer patients selected based on MRI findings: a prospective feasibility trial. Breast Cancer Res Treat 2020; 182:97-105. [DOI: 10.1007/s10549-020-05678-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/09/2020] [Indexed: 12/23/2022]
|
31
|
Kim EY, Do SI, Yun JS, Park YL, Park CH, Moon JH, Youn I, Choi YJ, Ham SY, Kook SH. Preoperative evaluation of mammographic microcalcifications after neoadjuvant chemotherapy for breast cancer. Clin Radiol 2020; 75:641.e19-641.e27. [PMID: 32291081 DOI: 10.1016/j.crad.2020.03.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
Abstract
AIM To assess the predictive value of preoperative residual mammographic microcalcifications for residual tumours after neoadjuvant chemotherapy (NAC) for breast cancer. MATERIALS AND METHODS This single-centre retrospective study included breast cancer patients who underwent NAC and demonstrated suspicious microcalcifications within or near the tumour bed on mammography from June 2015 to August 2018. The residual microcalcifications and remnant lesion on magnetic resonance imaging (MRI) were correlated with histopathological findings of residual tumours and immunohistochemical markers. RESULTS A total of 96 patients were included. Ten patients achieved pathological complete response (pCR) and previous suspicious microcalcifications were associated with benign pathology in 10.4% (10/96) of the patients. In the remaining 86 patients who did not achieve pCR, 61.5% (59/96) of the residual microcalcifications were associated with invasive or in situ carcinoma and 28.1% (27/96) with benign pathology. Hormone receptor-positive (HR+) patients had the highest proportion of residual malignant microcalcifications compared to HR- patients (48.9% versus 13.5%, respectively; p=0.019). MRI correlated better than residual microcalcifications on mammography in predicting residual tumour extent in all subtypes (ICC=0.709 versus 0.365). MRI also showed higher correlation with residual tumour size for the HR-/HER2+ and HR-/HER2- subtype (ICC=0.925 and 0.876, respectively). CONCLUSION The extent of microcalcifications on mammography after NAC did not correlate with the extent of residual cancer in 38.5% of women. Regardless of the extent of microcalcifications, residual tumour extent on MRI after NAC and molecular subtype could be an accurate tool in evaluating residual cancer after NAC.
Collapse
Affiliation(s)
- E Y Kim
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - S-I Do
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - J-S Yun
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Y L Park
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - C H Park
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - J H Moon
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - I Youn
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Y J Choi
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - S-Y Ham
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - S H Kook
- Department of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Chen CA, Hayward JH, Woodard GA, Ray KM, Starr CJ, Hylton NM, Joe BN, Lee AY. Complete Breast MRI Response to Neoadjuvant Chemotherapy and Prediction of Pathologic Complete Response. JOURNAL OF BREAST IMAGING 2019; 1:217-222. [PMID: 38424754 DOI: 10.1093/jbi/wbz028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Indexed: 03/02/2024]
Abstract
OBJECTIVE To assess the negative predictive value (NPV) of breast MRI in detecting residual disease after neoadjuvant chemotherapy (NAC) in women with invasive breast cancer, overall and by tumor subtype. METHODS An institutional review board approved retrospective study from January 2010 through December 2016 identified patients with invasive breast cancer who achieved complete MRI response to NAC, defined as the absence of residual enhancement in the tumor bed above background parenchymal enhancement. During the study period, it was our routine practice to assign a BI-RADS 1 or 2 assessment to these cases. The NPV was defined as the ability of a complete MRI response to predict pathologic complete response (pCR) at final surgical pathology. Statistical analyses were performed using a Fisher exact test. RESULTS Among 244 patients who underwent MRI to assess NAC response, 38 (16%) were determined to have complete MRI response by the interpreting radiologist. Of these, 20/38 (53%) had pCR. Complete MRI response did not significantly predict pCR for the total group (P < 0.9). However, NPVs significantly varied by molecular subtype (P < 0.004). True negative MRIs by tumor subtype were 2/10 (20%) for hormone receptor (HR)+/HER2-, 3/10 (30%) for HR+/HER2+, 6/8 (75%) for HR-/HER+, and 9/10 (90%) for triple negative (TN) subtypes. Complete MRI response significantly predicted pCR for only the TN subtype (NPV 90%; P < 0.02). CONCLUSIONS In patients with complete MRI response, 53% had pCR. While MRI lacks sufficient NPV to obviate the need for surgical excision, it may add prognostic value for certain molecular subtypes. The TN subtype demonstrated the highest NPV.
Collapse
Affiliation(s)
- Christina A Chen
- University of California San Francisco, Department of Radiology and Medical Imaging, San Francisco, CA
| | - Jessica H Hayward
- University of California San Francisco, Department of Radiology and Medical Imaging, San Francisco, CA
| | | | - Kimberly M Ray
- University of California San Francisco, Department of Radiology and Medical Imaging, San Francisco, CA
| | | | - Nola M Hylton
- University of California San Francisco, Department of Radiology and Medical Imaging, San Francisco, CA
| | - Bonnie N Joe
- University of California San Francisco, Department of Radiology and Medical Imaging, San Francisco, CA
| | - Amie Y Lee
- University of California San Francisco, Department of Radiology and Medical Imaging, San Francisco, CA
| |
Collapse
|
33
|
Complete response on MR imaging after neoadjuvant chemotherapy in breast cancer patients: Factors of radiologic-pathologic discordance. Eur J Radiol 2019; 118:114-121. [DOI: 10.1016/j.ejrad.2019.06.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 01/07/2023]
|
34
|
Fushimi A, Kudo R, Takeyama H. Do Decreased Breast Microcalcifications After Neoadjuvant Chemotherapy Predict Pathologic Complete Response? Clin Breast Cancer 2019; 20:e82-e88. [PMID: 31255548 DOI: 10.1016/j.clbc.2019.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/12/2019] [Accepted: 05/29/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Clinical response evaluation by image examination after neoadjuvant chemotherapy for breast cancer is important for determining drug response and progression. Mammography is less correlated with pathologic response assessment than magnetic resonance imaging or ultrasonography. The present study clarified characteristics of breast cancer patients with decreased microcalcifications after neoadjuvant chemotherapy to increase the accuracy of mammographic assessment. PATIENTS AND METHODS Consecutive patients who received neoadjuvant chemotherapy for breast cancer at our hospital from January 2013 to June 2017 were retrospectively reviewed. Characteristics of cases of microcalcifications before neoadjuvant chemotherapy and those showing a reduction after were examined. Stromal tumor-infiltrating lymphocytes were evaluated in the biopsy. Pathologic complete response was also explored in patients exhibiting a decrease in microcalcifications by neoadjuvant chemotherapy. RESULTS Seventy breast cancer cases were included; 37 had no malignant microcalcifications before neoadjuvant chemotherapy and 33 did. Breast cancer with microcalcifications was significantly more positive for human epidermal growth factor receptor 2 than those without microcalcifications (48% vs. 22%, P = .018). Only 6 of 33 patients with microcalcifications before neoadjuvant chemotherapy showed a decrease after treatment. These patients tended to have high stromal tumor-infiltrating lymphocytes (50% vs. 17%), segmental microcalcifications (66% vs. 33%), and pleomorphic/linear microcalcifications (83% vs. 37%). CONCLUSION These results suggest that neoadjuvant chemotherapy is able to decrease malignant calcifications, particularly segmental and pleomorphic/linear microcalcifications. By clarifying the mechanisms of formation and disappearance of microcalcifications, a consensus can be reached on whether microcalcifications apparent by mammography are useful for evaluating response to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Atsushi Fushimi
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan.
| | - Rei Kudo
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Takeyama
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Morigi C. Highlights of the 16th St Gallen International Breast Cancer Conference, Vienna, Austria, 20-23 March 2019: personalised treatments for patients with early breast cancer. Ecancermedicalscience 2019; 13:924. [PMID: 31281421 PMCID: PMC6546258 DOI: 10.3332/ecancer.2019.924] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Indexed: 12/15/2022] Open
Abstract
The 16th St Gallen International Breast Cancer Conference took place in Vienna for the third time, from 20–23 March 2019. More than 3000 people from all over the world were invited to take part in this important bi-annual critical review of the ‘state of the art’ in the primary care of breast cancer (BC), independent of political and industrial pressure, with the aim to integrate the most recent research data and most important developments in BC therapies since St Gallen International Breast Cancer Conference 2017, with the ultimate goal of drawing up a consensus for the current optimal treatment and prevention of BC. This year, the St Gallen Breast Cancer Award was won by Monica Morrow (Memorial Sloan Kettering Cancer Center, USA) for her extraordinary contribution in research and practise development in the treatment of BC. She opened the session with the lecture ‘Will surgery be a part of BC treatment in the future?’ Improved systemic therapy has decreased BC mortality and increased pathologic complete response (pCR) rates after neoadjuvant chemotherapy (NACT). Improved imaging and increased screening uptake have led to detect smaller cancers. These factors have highlighted two possible scenarios to omit surgery: for patients with small low-grade ductal carcinoma in situ (DCIS) and for those who have received NACT and had a clinical and radiological complete response. However, considering that 7%–20% of `low-risk’ DCIS patients have co-existing invasive cancer at diagnosis, that surgery has become progressively less morbid and less toxic than some systemic therapies with a lower cost-effectiveness ratio, and that identification of pathologic complete response (pCR) without surgery requires more intensive imaging follow-up (more biopsies, higher cost and more anxiety for the patient), surgery still appears to be an essential treatment for BC. The Umberto Veronesi Memorial Award went to Lesley Fallowfield (Brighton and Sussex Medical School, UK) for her important research and activity in the field of the development of patient outcome, of better communication skills and quality of life for women. In her lecture, she remarked on the importance of improving BC personalised treatments, especially through co-operation between scientists, always considering the whole woman and not just her breast disease. This award was given by Paolo Veronesi, after a moving introduction which culminated with the following words of Professor Umberto Veronesi: ‘It is not possible to take care of the people’s bodies without taking care of their mind. My duty, the duty of all doctors, is to listen and be part of the emotions of those we treat every day’.
Collapse
Affiliation(s)
- Consuelo Morigi
- Division of Senology, IRCCS European Institute of Oncology, 20141 Milan, Italy
| |
Collapse
|
36
|
Abstract
Breast cancer treatment is multidisciplinary. Most women with early stage breast cancer are candidates for breast-conserving surgery with radiotherapy or mastectomy. The risk of local recurrence and the chance of survival does not differ with these approaches. Sentinel node biopsy is used for axillary staging, and individualized approaches are minimizing the need for axillary dissection in women with positive sentinel nodes. Adjuvant systemic therapy is used in most women based on proven survival benefit, and molecular profiling to individualize treatment based on risk is now a clinical reality for patients with hormone receptor-positive cancers.
Collapse
Affiliation(s)
- Tracy-Ann Moo
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065, USA
| | - Rachel Sanford
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065, USA
| | - Chau Dang
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065, USA
| | - Monica Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065, USA.
| |
Collapse
|
37
|
Park J, Chae EY, Cha JH, Shin HJ, Choi WJ, Choi YW, Kim HH. Comparison of mammography, digital breast tomosynthesis, automated breast ultrasound, magnetic resonance imaging in evaluation of residual tumor after neoadjuvant chemotherapy. Eur J Radiol 2018; 108:261-268. [PMID: 30396666 DOI: 10.1016/j.ejrad.2018.09.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/17/2018] [Accepted: 09/29/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND To compare the accuracy of mammography (MG), digital breast tomosynthesis (DBT), automated breast ultrasound (ABUS) and magnetic resonance imaging (MRI) for the assessment of residual tumor extent in breast cancer after neoadjuvant chemotherapy (NAC). METHODS Fifty-one stage II-III breast cancer undergoing NAC were enrolled from March 2015 to December 2016. The longest diameter of residual tumor measured with MG, DBT, ABUS and MRI was compared with the pathologic tumor size. Statistical analysis was performed using intraclass correlation coefficients (ICC) and marginal homogeneity test. Receiver operating characteristics (ROC) analysis was used to evaluate the diagnostic performance for predicting pathologic complete response (pCR). RESULTS MRI size correlated well with pathology (ICC = 0.83), significantly better than MG, DBT and ABUS size (ICC = 0.56, ICC = 0.63 and ICC = 0.55, respectively). The discrepancy between MRI and pathology was statistical different from that of MG and ABUS (p = 0.0231 and 0.0039, respectively), but not different from that of DBT (p = 0.5727). For predicting pCR, MRI and DBT had a better performance compared to MG and US (area under the ROC curve: 0.92, 0.84, 0.72, 0.75, respectively; p = 0.3749 for DBT, p = 0.0972 for MG and p = 0.0596 for ABUS, when MRI being reference). CONCLUSIONS MRI and DBT allow more accurate assessment of tumor size compared to pathology compared with MG and ABUS. MRI and DBT outperform MG and ABUS in the prediction of pathologic complete response.
Collapse
Affiliation(s)
- Jiyoon Park
- Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Radiology, Research Institute of Radiology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Eun Young Chae
- Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joo Hee Cha
- Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Jung Shin
- Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Jung Choi
- Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Wook Choi
- Korea Electrotechnology Research Institute, Ansan, Republic of Korea
| | - Hak Hee Kim
- Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Vicini E, Invento A, Cuoghi M, Bafile A, Battaglia C, Biglia N, Busani M, Bussone R, Cianchetti E, Caruso F, Cucchi M, Dessena M, Di Filippo F, Fabi N, Folli S, Friedman D, Macellari G, Mainente P, Murgo R, Neri A, Pollini G, Palli D, Ricci F, Scalco G, Taffurelli M, Trunfio M, Galimberti V. Neoadjuvant systemic treatment for breast cancer in Italy: The Italian Society of Surgical Oncology (SICO) Breast Oncoteam survey. Eur J Surg Oncol 2018; 44:1157-1163. [DOI: 10.1016/j.ejso.2018.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 03/01/2018] [Accepted: 03/20/2018] [Indexed: 01/09/2023] Open
|
39
|
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
|
40
|
Rauch GM, Kuerer HM, Adrada B, Santiago L, Moseley T, Candelaria RP, Arribas E, Sun J, Leung JWT, Krishnamurthy S, Yang WT. Biopsy Feasibility Trial for Breast Cancer Pathologic Complete Response Detection after Neoadjuvant Chemotherapy: Imaging Assessment and Correlation Endpoints. Ann Surg Oncol 2018; 25:1953-1960. [PMID: 29667115 DOI: 10.1245/s10434-018-6481-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Indexed: 02/03/2023]
Abstract
PURPOSE This study was designed to present the secondary imaging endpoints of the trial for evaluating mammogram (MMG), ultrasound (US) and image guided biopsy (IGBx) assessment of pathologic complete response (pCR) in breast cancer (BC) patients undergoing neoadjuvant chemotherapy (NAC). METHODS Patients with T1-3, N0-3, M0 triple-negative or HER2-positive BC who received NAC were enrolled in an Institutional Review Board-approved prospective, clinical trial. Patients underwent US and MMG at baseline and after NAC. Images were evaluated for residual abnormality and to determine modality for IGBx [US-guided (USG) or stereotactic guided (SG)]. Fine-needle aspiration and 9-G, vacuum-assisted core biopsy (VACBx) of tumor bed was performed after NAC and was compared with histopathology at surgery. RESULTS Forty patients were enrolled. Median age was 50.5 (range 26-76) years; median baseline tumor size was 2.4 cm (range 0.8-6.3) and 1 cm (range 0-5.5) after NAC. Nineteen patients had pCR: 6 (32%) had residual Ca2+ presurgery, 5 (26%) residual mass, 1 (5%) mass with calcifications, and 7 (37%) no residual imaging abnormality. Sensitivity, specificity, and accuracy of US, MMG, and IGBx for pCR were 47/95/73%, 53/90/73%, and 100/95/98%, respectively. Twenty-five (63%) patients had SGBx and 15 (37%) had US-guided biopsy (USGBx). Median number of cores was higher with SGBx (12, range 6-14) than with USGBx (8, range 4-12), p < 0.002. Positive predictive value for pCR was significantly higher for SG VACBx than for USG VACBx (100 vs. 60%, p < 0.05). CONCLUSIONS SG VACBx is the preferred IGBx modality for identifying patients with pCR for trials testing the safety of eliminating surgery.
Collapse
Affiliation(s)
- Gaiane M Rauch
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Henry M Kuerer
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beatriz Adrada
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lumarie Santiago
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tanya Moseley
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rosalind P Candelaria
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elsa Arribas
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jessica W T Leung
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei T Yang
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
41
|
Um E, Kang JW, Lee S, Kim HJ, Yoon TI, Sohn G, Chung IY, Kim J, Lee JW, Son BH, Ahn SH, Ko BS. Comparing Accuracy of Mammography and Magnetic Resonance Imaging for Residual Calcified Lesions in Breast Cancer Patients Undergoing Neoadjuvant Systemic Therapy. Clin Breast Cancer 2018; 18:e1087-e1091. [PMID: 29703689 DOI: 10.1016/j.clbc.2018.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 01/08/2018] [Accepted: 03/10/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Neoadjuvant systemic therapy (NST) is performed to increase the rate of breast-conserving surgery in advanced breast cancer patients. Although magnetic resonance imaging (MRI) is accurate in predicting residual cancer, if calcification remains, the issue of whether to perform the surgery on the basis of the residual tumor prediction range in mammography (MMG) or MRI has not yet been elucidated. This study aimed to estimate the accuracy of predicting residual tumor after NST for residual microcalcification on mammographic and enhancing lesion on MRI. PATIENTS AND METHODS This was a single-center, retrospective study. We included breast cancer patients who underwent NST, had microcalcifications in the post-NST MMG, and underwent surgery from January 2, 2013 to December 30, 2014 at Asan Medical Center. Patients with post-NST MMG as well as MRI were included. Final pathologic tumor size with histopathology and biomarker status were obtained postoperatively. RESULTS In total, 151 patients were included in this study. Overall, MRI correlated better than MMG in predicting the tumor size (intraclass correlation coefficient [ICC], 0.769 vs. 0.651). For hormone receptor (HR)-positive (HR+)/HER2- subtype, MMG had higher correlation than MRI (ICC = 0.747 vs. 0.575). In HR- subtype, MRI had a strong correlation with pathology (HR-/HER2+ or triple negative (TN), ICC = 0.939 vs. 0.750), whereas MMG tended to overestimate the tumor size (HR-/HER2+ or TN, ICC = 0.543 vs. 0.479). CONCLUSION Post-NST residual microcalcifications on MMG have a lower correlation with residual tumor size than MRI. Other than HR+/HER2- subtype, the extent of calcifications on preoperative evaluation might not be accurate in evaluating the residual extent of the tumor after NST.
Collapse
Affiliation(s)
- Eunhae Um
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ji-Won Kang
- Department of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - SaeByul Lee
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Tae In Yoon
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Guiyun Sohn
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Il Yong Chung
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jisun Kim
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jong Won Lee
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Byung Ho Son
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sei Hyun Ahn
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Beom Seok Ko
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
42
|
An YY, Kim SH, Kang BJ. Residual microcalcifications after neoadjuvant chemotherapy for locally advanced breast cancer: comparison of the accuracies of mammography and MRI in predicting pathological residual tumor. World J Surg Oncol 2017; 15:198. [PMID: 29110671 PMCID: PMC5674773 DOI: 10.1186/s12957-017-1263-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 10/23/2017] [Indexed: 11/17/2022] Open
Abstract
Background The aims of this study were to correlate residual mammographic microcalcifications after neoadjuvant chemotherapy (NAC) with pathological results and to compare the accuracy of mammography (MG) and magnetic resonance imaging (MRI) in predicting the size of residual tumors. Methods The imaging findings and pathological results for 29 patients with residual microcalcifications after NAC were reviewed. We compared the agreement of the measured extent of residual microcalcifications based on MG and residual enhancement based on MRI with the residual tumor size based on pathology. Results At final pathology, residual microcalcifications were malignant in 55.2% of cases and benign in 44.8% of cases. In 36% of non-pCR cases, the remaining microcalcifications were benign. Compared with the measurements of residual tumor obtained from pathology, MG showed poor agreement, and MRI showed moderate agreement, for the entire group (concordance correlation coefficient [CCC] = 0.196 vs. 0.566). Regarding the receptor status, the agreement of measurements obtained by MG was superior to that obtained by MRI (CCC = 0.5629, 0.5472 vs. 0.4496, 0.4279) for ER(+) and HER2(−) tumors. In ER(−) tumors, the measurements obtained by MG showed the lowest agreement with the pathological tumor size, which had the highest agreement with those obtained by MRI (CCC = − 0.0162 vs. 0.8584). Conclusions Residual mammographic microcalcifications after NAC did not correlate with malignancy in 44.8% of cases. Residual microcalcifications on MG were poorly correlated with pathological tumor size, and MRI might be more reliable for predicting residual tumor size after NAC. Tumor receptor status affected the accuracy of both MG and MRI for predicting residual tumor size after NAC. Trial registration CRIS, KCT0002281; registered 6 April 2015, retrospectively registered
Collapse
Affiliation(s)
- Yeong Yi An
- Department of Radiology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, 93, Jungbu-daero, Paldal-gu, Suwon-si, 16247, Gyeonggi-do, Republic of Korea
| | - Sung Hun Kim
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Bong Joo Kang
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
43
|
Morigi C. Highlights from the 15th St Gallen International Breast Cancer Conference 15-18 March, 2017, Vienna: tailored treatments for patients with early breast cancer. Ecancermedicalscience 2017; 11:732. [PMID: 28491135 PMCID: PMC5406222 DOI: 10.3332/ecancer.2017.732] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Indexed: 01/16/2023] Open
Abstract
The 15th St Gallen International Breast Cancer Conference was held in Vienna for the second time, from 15th–18th March 2017. 4000 people from 105 countries all over the world were invited to take part in the event. The real highlight of the conference was the last day with the International Consensus Session which was chaired by around 50 experts on breast cancer worldwide. With reference to data from scientific research, the consensus panel tried to offer guidelines for the management of breast cancer with the aim of providing patients with optimal treatment. The topics covered focused on the treatment of breast cancer, consideration of surgery, radiotherapy, neo-adjuvant, and adjuvant systemic therapy for breast cancer, as well as genetics and prevention of breast cancer. In particular, in terms of precision medicine, an important topic of the conference was ‘is it possible to think that it could become routine in clinical practice to use immunotherapy and targeted therapy based on genetic signatures?’ In view of personalised therapy, it is important to take into consideration women’s treatment preferences. It is also important not only to offer guidelines which help breast cancer experts all over the world to choose the proper treatment for women with breast cancer but also to discuss the pros and cons of the therapy with the patient. This allows for a better understanding of the disease. ‘From the maximum tolerable to the minimum effective treatment: it is essential to escalate treatment when necessary and to de-escalate when unnecessary’. These few words could summarise the meaning of the 15th St Gallen International Breast Cancer Conference. Prof Martine Piccart-Gebhart was awarded with the St Gallen International Breast Cancer Award 2017 for her fundamental clinical research contribution and Prof Giuseppe Curigliano with the Umberto Veronesi Memorial Award which aims to recognise a physician’s leading role in advancing the science and care of breast cancer patients. Curigliano, in his lecture, spoke about the revolutionary immunotherapy in the clinical management of breast cancer (BC). For the development of these therapies, it is necessary to identify the genetic determinants of BC immune phenotypes in which The Cancer Genome Atlas (TCGA) has contributed towards this. For example, the T helper (Th-1) phenotype (ICR4), which also exhibits upregulation of immune-regulatory transcripts (eg. PDL1, PD1, FOXP3, IDO1, and CTLA4), was associated with prolonged patients’ survival. Chromosome segment 4q21, which includes genes encoding the Th-1 chemokines CXCL9-11, was significantly amplified only in the immune favourable phenotype (ICR4). The mutation and neo-antigen load progressively decreased from ICR4 to ICR1 but could not explain immune phenotypic differences. Mutations of TP53 were enriched in the immune favourable phenotype (ICR4). Instead, the presence of MAP3K1 and MAP2K4 mutations were closely associated with an immune unfavourable phenotype (ICR1). Using both the TCGA and the validation dataset, the degree of MAPK deregulation segregates BC according to their immune disposition. These findings suggest that mutational-driven deregulation of MAPK pathways is linked to the negative regulation of intratumoural immune response in BC. The main themes of this congress were: 1) Surgery of the primary tumour and margins; 2) Surgery of the axilla; 3) Radiotherapy: hypofractionated, ‘boost’ to tumour bed, partial breast, regional node, after mastectomy, advanced technology; 4) Pathology: subtypes, TILs; 5) Multi-gene signatures and therapy; 6) Endocrine therapy: pre- and post-menopausal and duration; 7) Chemotherapy: subtypes, stages; 8) Anti-HER-2 therapy; 9) Neo-adjuvant therapy; 10) Adjuvant bisphosponates; 11) Adjuvant diet and exercise.
Collapse
Affiliation(s)
- Consuelo Morigi
- Division of Breast Cancer Surgery, European Institute of Oncology, Via Ripamonti 435, 20146 Milano, Italy
| |
Collapse
|