1
|
Wang F, Wang Y, Xiong B, Yang Z, Wang J, Yao Y, Yu L, Fu Q, Li L, Zhang Q, Zheng C, Huang S, Liu L, Liu C, Sun H, Mao B, Liu DX, Yu Z. Neoadjuvant pyrotinib and trastuzumab in HER2-positive breast cancer with no early response (NeoPaTHer): efficacy, safety and biomarker analysis of a prospective, multicentre, response-adapted study. Signal Transduct Target Ther 2025; 10:45. [PMID: 39875376 PMCID: PMC11775149 DOI: 10.1038/s41392-025-02138-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
The potential benefits of pyrotinib for patients with trastuzumab-insensitive, HER2-positive early-stage breast cancer remain unclear. This prospective, multicentre, response-adapted study evaluated the efficacy and safety of adding pyrotinib to the neoadjuvant treatment of HER2-positive breast cancer patients with a poor response to initial docetaxel plus carboplatin and trastuzumab (TCbH). Early response was assessed using magnetic resonance imaging (MRI) after two cycles of treatment. Patients showing poor response, as defined by RECIST 1.1, could opt to receive additional pyrotinib or continue standard therapy. The primary endpoint was the total pathological complete response (tpCR; ypT0/isN0) rate. Of the 129 patients enroled, 62 (48.1%) were identified as MRI-responders (cohort A), 26 non-responders continued with four more cycles of TCbH (cohort B), and 41 non-responders received additional pyrotinib (cohort C). The tpCR rate was 30.6% (95% CI: 20.6-43.0%) in cohort A, 15.4% (95% CI: 6.2-33.5%) in cohort B, and 29.3% (95% CI: 17.6-44.5%) in cohort C. Multivariable logistic regression analyses demonstrated comparable odds of achieving tpCR between cohorts A and C (odds ratio = 1.04, 95% CI: 0.40-2.70). No new adverse events were observed with the addition of pyrotinib. Patients with co-mutations of TP53 and PIK3CA exhibited lower rates of early partial response compared to those without or with a single gene mutation (36.0% vs. 60.0%, P = 0.08). These findings suggest that adding pyrotinib may benefit patients who do not respond to neoadjuvant trastuzumab plus chemotherapy. Further investigation is warranted to identify biomarkers predicting patients' benefit from the addition of pyrotinib.
Collapse
Affiliation(s)
- Fei Wang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Yongjiu Wang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Bin Xiong
- Department of Breast Surgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Zhenlin Yang
- Department of Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Jingfen Wang
- Department of Breast Surgery, Linyi Cancer Hospital, Linyi, China
| | - Yumin Yao
- Department of Breast Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Lixiang Yu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Qinye Fu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Liang Li
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Qiang Zhang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Chao Zheng
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Shuya Huang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Liyuan Liu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Chun Liu
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Huaibo Sun
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Beibei Mao
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Dong-Xu Liu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Zhigang Yu
- Breast Center, The Second Hospital of Shandong University, Jinan, China.
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China.
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China.
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China.
| |
Collapse
|
2
|
Niu S, Sun T, Wang M, Yao L, He T, Wang Y, Zhang H, Li X, Xu Y. Multiple time points for detecting circulating tumor DNA to monitor the response to neoadjuvant therapy in breast cancer: a meta-analysis. BMC Cancer 2025; 25:115. [PMID: 39844103 PMCID: PMC11752932 DOI: 10.1186/s12885-025-13526-0] [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: 03/06/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Not all breast cancer (BC) patients can benefit from neoadjuvant therapy (NAT). A poor response may result in patients missing the best opportunity for treatment, ultimately leading to a poor prognosis. Thus, to identify an effective predictor that can assess and predict patient response at early time points, we focused on circulating tumor DNA (ctDNA), which is a vital noninvasive liquid biopsy biomarker. We performed a meta-analysis to explore the predictive value of response by monitoring ctDNA at four time points of NAT using pathologic complete response (pCR) and residual cancer burden (RCB). METHODS By searching Embase, PubMed, the Cochrane Library, and the Web of Science until December 24, 2023, we selected studies concerning the relationship between ctDNA and response or prognosis. We analysed the results at the following various time points: baseline (T0), first cycle of NAT (T1), mid-treatment (MT), and end of NAT (EOT). pCR and RCB were used to evaluate the response as the primary endpoint. The secondary endpoint was to investigate the relationship between ctDNA and prognosis. Odds ratios (ORs) and hazard ratios (HRs) were used as effect indicators. RESULTS Thirteen reports from twelve studies were eligible for inclusion in this meta-analysis. The results demonstrated that ctDNA negativity was associated with pCR at T1 (OR = 0.34; 95% CI: 0.21-0.57), MT (OR = 0.35; 95% CI: 0.20-0.60), and EOT (OR = 0.38; 95% CI: 0.22-0.66). When RCB was used to evaluate responses, ctDNA negativity was associated with RCB-0/I at the MT (OR = 0.34; 95% CI: 0.21-0.55) and EOT (OR = 0.26; 95% CI: 0.15-0.46). Furthermore, ctDNA positivity at T1 predicted a worse prognosis for patients (HR = 2.73; 95% CI: 1.29-5.75). We also performed a subgroup analysis to more accurately assess the predictive value of ctDNA for triple-negative breast cancer. CONCLUSIONS Our meta-analysis suggested that the ctDNA status at the early stage of NAT can predict patient response, which provides evidence for adjusting personalized treatment strategies and improving patient survival. PROSPERO REGISTRATION NUMBER CRD42024496465.
Collapse
Affiliation(s)
- Shuyi Niu
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tie Sun
- The Third Department of General Surgery, People's Hospital of China Medical University (Liaoning Provincial People's Hospital), Shenyang, Liaoning, 110001, China
| | - Mozhi Wang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Litong Yao
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tianyi He
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yusong Wang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Hengjun Zhang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Xiang Li
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Yingying Xu
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
3
|
Khan AA, Ahuja S, G. K, Zaheer S. Evaluating the Clinico-Pathological Relationship Between Stromal Tumor-Infiltrating Lymphocytes and Androgen Receptor Expression Across Molecular Subtypes of Invasive Breast Carcinoma. Indian J Surg Oncol 2024; 15:802-808. [PMID: 39555334 PMCID: PMC11564480 DOI: 10.1007/s13193-024-02001-0] [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: 02/17/2024] [Accepted: 06/20/2024] [Indexed: 11/19/2024] Open
Abstract
Breast cancer remains a significant cause of mortality globally, necessitating effective treatment strategies. Neoadjuvant chemotherapy (NAC) is widely employed to minimize tumor burden and prevent local spread, with treatment efficacy varying based on molecular subtypes. Despite advancements, resistance to conventional therapies persists, prompting the exploration of alternative approaches, including immune cell therapy. Tumor-infiltrating lymphocytes (TILs) have emerged as immunological biomarkers in breast cancer, exhibiting associations with molecular subtypes and treatment response. This retrospective study assessed the clinico-pathological relationship between stromal TILs and AR expression across molecular subtypes of invasive breast carcinoma in an Indian cohort. Thirty-seven patients receiving NAC followed by modified radical mastectomy were analyzed for TILs and molecular subtyping. Immunohistochemistry was used to determine hormone receptor status and AR expression. A higher AR positivity was observed in hormone receptor-positive/Her2neu-negative and hormone receptor-positive/Her2neu-positive tumors compared to triple-negative breast cancers (TNBCs). Significant associations were observed between AR expression and tumor grade, but not with age or Her2neu status. Although no significant correlation was found between AR and complete response to NAC, a weak negative correlation between AR and TILs was noted. Notably, TNBCs with negative AR and Ki67 index exhibited poorer responses to NAC, emphasizing the need for adjuvant therapy. These findings underscore the complex interplay between AR, TILs, and treatment response in breast cancer, highlighting the potential of personalized therapeutic approaches. Further research is warranted to elucidate the prognostic significance of AR and its implications for tailored treatment strategies in breast cancer management.
Collapse
Affiliation(s)
- Adil Aziz Khan
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Kiruthikasri G.
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
4
|
Nezhad NZ, Nezhad HZ, Shahpar A, Shahrebabak AG, Shahrebabak MG, Farokhi FR. From mammary mystery to parasitic surprise: a rare case of primary breast hydatid cyst. BMC Infect Dis 2024; 24:1369. [PMID: 39614184 DOI: 10.1186/s12879-024-10270-x] [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: 08/20/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Primary breast hydatid cyst is an exceedingly rare manifestation of echinococcosis, with an incidence of less than 0.27% among all hydatid cyst cases. CASE REPORT This report presents a unique case of a 51-year-old multiparous female who initially presented with a painless left breast mass. Initial imaging studies, including ultrasonography and mammography, revealed a 4.5 × 4 cm cyst classified as BI-RADS 3. The cyst was initially managed with fine-needle aspiration and conservative treatment. However, it recurred twice over a six-month period, necessitating surgical excision. Preoperative laboratory work ups revealed eosinophilia, a finding initially overlooked but later recognized as significant. Histopathological examination of the excised specimen confirmed the diagnosis of a hydatid cyst. Post-surgical management included albendazole therapy, regular imaging follow-ups, and patient education on hygiene practices to prevent reinfection. CONCLUSION This case highlights the importance of considering parasitic etiologies in the differential diagnosis of breast masses, particularly in endemic regions. It also underscores the value of a multidisciplinary approach in managing such rare cases. The unexpected diagnosis of a primary breast hydatid cyst in this case serves as a reminder of the diverse presentations of echinococcosis and the need for heightened clinical suspicion in atypical breast lesions. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Nazanin Zeinali Nezhad
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | | | - Amirhossein Shahpar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman, Iran
| | - Aazam Gholami Shahrebabak
- Department of Pediatrics Afzalipour Hospital, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Gholami Shahrebabak
- Department of Pediatrics, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
5
|
Duan X, Xue B, Xu Z, Niu Z. Multimodal Photodynamic Therapy by Inhibiting the Nrf2/ARE Signaling Pathway in Tumors. ACS Biomater Sci Eng 2024; 10:7018-7029. [PMID: 39417585 DOI: 10.1021/acsbiomaterials.4c01643] [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] [Indexed: 10/19/2024]
Abstract
Photodynamic therapy (PDT) has been widely used in the clinical therapy of various tumors, especially superficial tumors. However, the tumor microenvironment presents hypoxia, as well as the inherent antioxidant system (e.g., Nrf2) of tumor cells limits the therapeutic outcomes. Herein, a cascade-responsive "oxidative stress amplifier" (named EZ@TD) is designed by encapsulating manganese-doped carbon dots acting as a photosensitizer and catalase (CAT)-like nanozyme within pH-sensitive ZIF-8 and Zn2+-activated DNAzyme for relieving hypoxia and efficient Nrf2 gene disruption to enhance PDT. It is demonstrated that EZ@TD synergistically inhibited tumor growth and activated the antitumor immune response by inhibiting the Nrf2/ARE signaling pathway in tumors. We provide a new paradigm for amplifying intracellular oxidative stress by interfering with various signaling pathways.
Collapse
Affiliation(s)
- Xin Duan
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Bingjian Xue
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zimeng Xu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zixu Niu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
6
|
Khan AA, Ahuja S, Kiruthikasri K, Zaheer S. Assessment of stromal tumor-infiltrating lymphocytes in neoadjuvant chemotherapy for invasive breast carcinoma: Predictive insights across molecular subtypes. Pathol Res Pract 2024; 260:155382. [PMID: 38850879 DOI: 10.1016/j.prp.2024.155382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND AND AIMS Breast cancer, a leading cause of female mortality, has prompted the widespread adoption of Neoadjuvant chemotherapy (NAC) for its potential to minimize metastasis risk and downstaging tumors. Tumor Infiltrating Lymphocytes (TILs) have emerged as key immunological biomarkers, particularly in breast cancer research. This study focuses on evaluating Stromal TILs (sTILs) in pre-NAC core needle biopsies of Invasive Breast Carcinoma, No Special Type (IBC, NST) and correlating it with NAC response. MATERIALS AND METHODS A retrospective study spanning three years (October 2020 to September 2023) was conducted in a tertiary care hospital, involving 73 patients meeting specific inclusion criteria. Pathological assessments, including hormone receptor status, molecular subtyping, and TILs evaluation, were performed. Logistic regression and statistical analyses were conducted to determine associations between TILs, clinicopathological parameters, and complete response. RESULTS The study demonstrated excellent discriminatory power of TILs (>10 %) in predicting complete response. Univariate and multivariate logistic regression underscored the independent predictive value of TILs, emphasizing their significance across diverse molecular subtypes. CONCLUSION This study provides crucial insights into immune response assessment, particularly sTILs, in optimizing breast cancer treatment strategies and patient outcomes during NAC, contributing to the evolving landscape of personalized emphasising oncology.
Collapse
Affiliation(s)
- Adil Aziz Khan
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Kiruthikasri Kiruthikasri
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
7
|
Sun HK, Jiang WL, Zhang SL, Xu PC, Wei LM, Liu JB. Predictive value of tumor-infiltrating lymphocytes for neoadjuvant therapy response in triple-negative breast cancer: A systematic review and meta-analysis. World J Clin Oncol 2024; 15:920-935. [PMID: 39071463 PMCID: PMC11271722 DOI: 10.5306/wjco.v15.i7.920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND The association between tumor-infiltrating lymphocyte (TIL) levels and the response to neoadjuvant therapy (NAT) in patients with triple-negative breast cancer (TNBC) remains unclear. AIM To investigate the predictive potential of TIL levels for the response to NAT in TNBC patients. METHODS A systematic search of the National Center for Biotechnology Information PubMed database was performed to collect relevant published literature prior to August 31, 2023. The correlation between TIL levels and the NAT pathologic complete response (pCR) in TNBC patients was assessed using a systematic review and meta-analysis. Subgroup analysis, sensitivity analysis, and publication bias analysis were also conducted. RESULTS A total of 32 studies were included in this meta-analysis. The overall meta-analysis results indicated that the pCR rate after NAT treatment in TNBC patients in the high TIL subgroup was significantly greater than that in patients in the low TIL subgroup (48.0% vs 27.7%) (risk ratio 2.01; 95% confidence interval 1.77-2.29; P < 0.001, I 2 = 56%). Subgroup analysis revealed that the between-study heterogeneity originated from differences in study design, TIL level cutoffs, and study populations. Publication bias could have existed in the included studies. The meta-analysis based on different NAT protocols revealed that all TNBC patients with high levels of TILs had a greater rate of pCR after NAT treatment in all protocols (all P ≤ 0.01), and there was no significant between-protocol difference in the statistics among the different NAT protocols (P = 0.29). Additionally, sensitivity analysis demonstrated that the overall results of the meta-analysis remained consistent when the included studies were individually excluded. CONCLUSION TILs can serve as a predictor of the response to NAT treatment in TNBC patients. TNBC patients with high levels of TILs exhibit a greater NAT pCR rate than those with low levels of TILs, and this predictive capability is consistent across different NAT regimens.
Collapse
Affiliation(s)
- Hai-Kuan Sun
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Wen-Long Jiang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Shi-Lei Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Peng-Cheng Xu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Li-Min Wei
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Jiang-Bo Liu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| |
Collapse
|
8
|
Yang X, Liu Q, Guo Z, Yang X, Li K, Han B, Zhang M, Sun M, Huang L, Cai G, Wu Y. Promoter profiles in plasma CfDNA exhibits a potential utility of predicting the efficacy of neoadjuvant chemotherapy in breast cancer patients. Breast Cancer Res 2024; 26:112. [PMID: 38965610 PMCID: PMC11225256 DOI: 10.1186/s13058-024-01860-3] [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: 04/07/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Gene expression profiles in breast tissue biopsies contain information related to chemotherapy efficacy. The promoter profiles in cell-free DNA (cfDNA) carrying gene expression information of the original tissues may be used to predict the response to neoadjuvant chemotherapy in breast cancer as a non-invasive biomarker. In this study, the feasibility of the promoter profiles in plasma cfDNA was evaluated as a novel clinical model for noninvasively predicting the efficacy of neoadjuvant chemotherapy in breast cancer. METHOD First of all, global chromatin (5 Mb windows), sub-compartments and promoter profiles in plasma cfDNA samples from 94 patients with breast cancer before neoadjuvant chemotherapy (pCR = 31 vs. non-pCR = 63) were analyzed, and then classifiers were developed for predicting the efficacy of neoadjuvant chemotherapy in breast cancer. Further, the promoter profile changes in sequential cfDNA samples from 30 patients (pCR = 8 vs. non-pCR = 22) during neoadjuvant chemotherapy were analyzed to explore the potential benefits of cfDNA promoter profile changes as a novel potential biomarker for predicting the treatment efficacy. RESULTS The results showed significantly distinct promoter profile in plasma cfDNA of pCR patients compared with non-pCR patients before neoadjuvant chemotherapy. The classifier based on promoter profiles in a Random Forest model produced the largest area under the curve of 0.980 (95% CI: 0.978-0.983). After neoadjuvant chemotherapy, 332 genes with significantly differential promoter profile changes in sequential cfDNA samples of pCR patients was observed, compared with non-pCR patients, and their functions were closely related to treatment response. CONCLUSION These results suggest that promoter profiles in plasma cfDNA may be a powerful, non-invasive tool for predicting the efficacy of neoadjuvant chemotherapy breast cancer patients before treatment, and the on-treatment cfDNA promoter profiles have potential benefits for predicting the treatment efficacy.
Collapse
Affiliation(s)
- Xu Yang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qing Liu
- Department of Pathology, The First People's Hospital of Foshan, Foshan, China
| | - Zhiwei Guo
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xuexi Yang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Kun Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Bowei Han
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Min Zhang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Minying Sun
- Department of Primary Public Health, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
- Institute of Public Health, Guangzhou Medical University & Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Limin Huang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Gengxi Cai
- Department of Pathology, The First People's Hospital of Foshan, Foshan, China.
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Yingsong Wu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Ma T, Ma T, Wang L, Wang H. Neoadjuvant Chemotherapy Is Effective in Those Infected With SARS-CoV-2: The Real-World Experience of a Large Chinese Breast Cancer Center. J Breast Cancer 2024; 27:176-186. [PMID: 38769687 PMCID: PMC11221209 DOI: 10.4048/jbc.2023.0299] [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: 12/27/2023] [Revised: 02/27/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024] Open
Abstract
PURPOSE During the major shift in China's policies on coronavirus disease 2019 (COVID-19), many residents will be infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) over a short period, including a few patients with breast cancer undergoing neoadjuvant chemotherapy (NAC). Moreover, it is unknown whether this comorbidity affects the efficacy of NAC for breast cancer and the patient's psychological state and quality of life (QOL). This study aims to answer these questions. METHODS The clinical data of 2,793 patients with breast cancer who received NAC at The Affiliated Hospital of Qingdao University were retrospectively collected. The infected and non-infected groups were divided according to whether they were infected with COVID-19 during NAC. Propensity score matching was used to reduce patient selection bias. The effectiveness, psychological well-being, and QOL of the two groups were compared. RESULTS No discernible differences were observed in the pathological complete response rates (p = 0.307) and major histological responses rate (p = 0.398) between the infected and non-infected groups. Following the full course of NAC, the Functional Assessment of Cancer Treatment General (p < 0.001) and Functional Assessment of Cancer Therapy for Breast Cancer (p < 0.001) were lower in the infected group than the non-infected group, the Hospital Anxiety and Depression Scale (HADS) anxiety scale (p < 0.001) and HADS depression scale (p < 0.001) were considerably higher in the infected group than the non-infected group. CONCLUSION With timely treatment and effective medical management, SARS-CoV-2 does not appear to affect the efficacy of NAC; however, it can significantly affect the QOL of patients and increase their psychological distress. Therefore, in addition to a timely assessment of the efficacy of NAC, it is necessary to dynamically understand the patient's psychological state and QOL.
Collapse
Affiliation(s)
- Teng Ma
- Breast Disease Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tianyi Ma
- Breast Disease Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lulu Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haibo Wang
- Breast Disease Center, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
10
|
Baldassi B, Poladyan H, Shahi A, Maa-Hacquoil H, Rapley M, Komarov B, Stiles J, Freitas V, Waterston M, Aseyev O, Reznik A, Bubon O. Image quality evaluation for a clinical organ-targeted PET camera. Front Oncol 2024; 14:1268991. [PMID: 38590664 PMCID: PMC10999605 DOI: 10.3389/fonc.2024.1268991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction A newly developed clinical organ-targeted Positron Emission Tomography (PET) system (also known as Radialis PET) is tested with a set of standardized and custom tests previously used to evaluate the performance of Positron Emission Mammography (PEM) systems. Methods Imaging characteristics impacting standardized uptake value (SUV) and detectability of small lesions, namely spatial resolution, linearity, uniformity, and recovery coefficients, are evaluated. Results In-plane spatial resolution was measured as 2.3 mm ± 0.1 mm, spatial accuracy was 0.1 mm, and uniformity measured with flood field and NEMA NU-4 phantom was 11.7% and 8.3% respectively. Selected clinical images are provided as reference to the imaging capabilities under different clinical conditions such as reduced activity of 2-[fluorine-18]-fluoro-2-deoxy-D-glucose (18F-FDG) and time-delayed acquisitions. SUV measurements were performed for selected clinical acquisitions to demonstrate a capability for quantitative image assessment of different types of cancer including for invasive lobular carcinoma with comparatively low metabolic activity. Quantitative imaging performance assessment with phantoms demonstrates improved contrast recovery and spill-over ratio for this PET technology when compared to other commercial organ-dedicated PET systems with similar spatial resolution. Recovery coefficients were measured to be 0.21 for the 1 mm hot rod and up to 0.89 for the 5 mm hot rod of NEMA NU-4 Image Quality phantom. Discussion Demonstrated ability to accurately reconstruct activity in tumors as small as 5 mm suggests that the Radialis PET technology may be well suited for emerging clinical applications such as image guided assessment of response to neoadjuvant systemic treatment (NST) in lesions smaller than 2 cm. Also, our results suggest that, while spatial resolution greatly influences the partial volume effect which degrades contrast recovery, optimized count rate performance and image reconstruction workflow may improve recovery coefficients for systems with comparable spatial resolution. We emphasize that recovery coefficient should be considered as a primary performance metric when a PET system is used for accurate lesion size or radiotracer uptake assessments.
Collapse
Affiliation(s)
- Brandon Baldassi
- Department of Physics, Lakehead University, Thunder Bay, ON, Canada
| | | | - Anirudh Shahi
- Department of Physics, Lakehead University, Thunder Bay, ON, Canada
| | | | - Madeline Rapley
- Department of Physics, Lakehead University, Thunder Bay, ON, Canada
| | | | - Justin Stiles
- Department of Physics, Lakehead University, Thunder Bay, ON, Canada
| | - Vivianne Freitas
- Department of Medical Imaging, University Health Network, Sinai Health System, Women’s College Hospital, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | | | - Olexiy Aseyev
- Department of Medical Oncology, Thunder Bay Regional Health Sciences Center, Thunder Bay, ON, Canada
| | - Alla Reznik
- Department of Physics, Lakehead University, Thunder Bay, ON, Canada
- Radialis Inc., Thunder Bay, ON, Canada
| | - Oleksandr Bubon
- Department of Physics, Lakehead University, Thunder Bay, ON, Canada
- Radialis Inc., Thunder Bay, ON, Canada
- Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| |
Collapse
|
11
|
Li X, Yan F. Predictive value of background parenchymal enhancement on breast magnetic resonance imaging for pathological tumor response to neoadjuvant chemotherapy in breast cancers: a systematic review. Cancer Imaging 2024; 24:35. [PMID: 38462607 PMCID: PMC10926651 DOI: 10.1186/s40644-024-00672-0] [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: 07/19/2023] [Accepted: 02/09/2024] [Indexed: 03/12/2024] Open
Abstract
OBJECTIVES This review aimed to assess the predictive value of background parenchymal enhancement (BPE) on breast magnetic resonance imaging (MRI) as an imaging biomarker for pathologic complete response (pCR) after neoadjuvant chemotherapy (NACT). METHODS Two reviewers independently performed a systemic literature search using the PubMed, MEDLINE, and Embase databases for studies published up to 11 June 2022. Data from relevant articles were extracted to assess the relationship between BPE and pCR. RESULTS This systematic review included 13 studies with extensive heterogeneity in population characteristics, MRI follow-up points, MRI protocol, NACT protocol, pCR definition, and BPE assessment. Baseline BPE levels were not associated with pCR, except in 1 study that reported higher baseline BPE of the younger participants (< 55 years) in the pCR group than the non-pCR group. A total of 5 studies qualitatively assessed BPE levels and indicated a correlation between reduced BPE after NACT and pCR; however, among the studies that quantitatively measured BPE, the same association was observed only in the subgroup analysis of 2 articles that assessed the status of hormone receptor and human epidermal growth factor receptor 2. In addition, the predictive ability of early BPE changes for pCR was reported in several articles and remains controversial. CONCLUSIONS Changes in BPE may be a promising imaging biomarker for predicting pCR in breast cancer. Because current studies remain insufficient, particularly those that quantitatively measure BPE, prospective and multicenter large-sample studies are needed to confirm this relationship.
Collapse
Affiliation(s)
- Xue Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 DaHua Road, Dong Dan, Beijing, 100730, PR China
- Graduate School of Peking, Union Medical College, Beijing, PR China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
12
|
Hartup SM, Morgan JL, Cheng VWT, Barry PA, Copson E, Cutress RI, Dave R, Elsberger B, Fairbrother P, Hogan B, Horgan K, Kirwan CC, McIntosh SA, O’Connell RL, Patani N, Potter S, Rattay T, Sheehan L, Wyld L, Kim B. The MARECA (national study of management of breast cancer locoregional recurrence and oncological outcomes) study: protocol for a prospective, multicentre cohort study. Int J Surg Protoc 2024; 28:20-26. [PMID: 38433867 PMCID: PMC10905491 DOI: 10.1097/sp9.0000000000000018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 03/05/2024] Open
Abstract
Background Despite a UK 5-year breast cancer survival rate of 86.6%, patients may develop breast cancer recurrence within the same breast after breast conserving surgery, as well as in the remaining skin or chest wall after mastectomy or in the ipsilateral lymph glands. These recurrences, collectively termed locoregional recurrence (LRR), occur in around 8% of patients within 10 years of their original diagnosis. Currently, there is a lack of robust information on the presentation and prevalence of LRR with no UK-specific clinical guidelines available for the optimal management of this patient group. Additionally, there is a need to identify patterns of LRR presentation and their progression, which will enable prognostic factors to be determined. This will subsequently enable the tailoring of treatment and improve patient outcome. Methods The MARECA study is a prospective, multicentre cohort study recruiting patients diagnosed with breast cancer LRR +/- associated distant metastases. Over 50 UK breast units are participating in the study with the aim of recruiting at least 500 patients over a recruitment period of 24 months. The data collected will detail the tumour pathology, imaging results, surgical treatment, radiotherapy and systemic therapy of the primary and recurrent breast cancer. Study follow-up will be for up to 5 years following LRR diagnosis to determine subsequent oncological outcomes and evaluate potential prognostic factors. Discussion This study will address the current knowledge gap and identify subgroups of patients who have less successful treatment outcomes. The results will determine the current management of LRR and the prognosis of patients diagnosed with breast cancer LRR +/- distant metastases in the UK, with the aim of establishing best practice and informing future national guidelines. The results will direct future research and inform the design of additional interventional trials and translational studies.
Collapse
Affiliation(s)
- Sue M. Hartup
- The Breast Unit at the Leeds Cancer Centre, St James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Jenna L. Morgan
- Division of Clinical Medicine, University of Sheffield School of Medicine and Population Health, Beech Hill Road, Sheffield
| | - Vinton WT Cheng
- The Breast Unit at the Leeds Cancer Centre, St James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Peter A. Barry
- Department of Breast Surgery, The Royal Marsden NHS Foundation Trust, Sutton, Surrey
- The Institute of Cancer Research
| | - Ellen Copson
- Somers Cancer Research Building, University of Southampton and University Hospital Southampton, Southampton
| | - Ramsey I. Cutress
- Somers Cancer Research Building, University of Southampton and University Hospital Southampton, Southampton
| | - Rajiv Dave
- The Nightingale Breast Cancer Centre, Wythenshawe Hospital
| | - Beatrix Elsberger
- Aberdeen Breast Unit, Aberdeen Royal Infirmary, Foresterhill, Aberdeen
| | | | - Brian Hogan
- The Breast Unit at the Leeds Cancer Centre, St James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Kieran Horgan
- The Breast Unit at the Leeds Cancer Centre, St James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Cliona C. Kirwan
- The Nightingale Breast Cancer Centre, Wythenshawe Hospital
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oglesby Cancer Research Building, Manchester Cancer Research Centre, Manchester
| | - Stuart A. McIntosh
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast
| | - Rachel L. O’Connell
- Department of Breast Surgery, The Royal Marsden NHS Foundation Trust, Sutton, Surrey
- The Institute of Cancer Research
| | - Neill Patani
- Department of Breast Surgery, University College London Hospitals NHS Foundation Trust
| | - Shelley Potter
- Translational Health Sciences, Bristol Medical School, Learning and Research Building, Southmead Hospital
- Bristol Breast Care Centre, North Bristol NHS Trust, Bristol
| | - Tim Rattay
- Leicester Cancer Research Centre, Clinical Sciences Building, University of Leicester, Leicester Royal Infirmary
- Department of Breast Surgery, University Hospitals of Leicester NHS Trust, Leicester
| | - Lisa Sheehan
- Wessex Deanery, Southern House, Otterbourne, Winchester, UK
| | - Lynda Wyld
- Division of Clinical Medicine, University of Sheffield School of Medicine and Population Health, Beech Hill Road, Sheffield
| | - Baek Kim
- The Breast Unit at the Leeds Cancer Centre, St James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds
| |
Collapse
|
13
|
Xia G, Zhang Z, Jiang Q, Wang H, Wang J. Predictive value of stromal tumor-infiltrating lymphocytes in patients with breast cancer treated with neoadjuvant chemotherapy: A meta-analysis. Medicine (Baltimore) 2024; 103:e36810. [PMID: 38335394 PMCID: PMC10860995 DOI: 10.1097/md.0000000000036810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The predictive value of tumor-infiltrating lymphocytes (TILs) in response to neoadjuvant chemotherapy (NAC) for breast cancer (BC) has received increasing attention. Here, a meta-analysis was conducted to evaluate the correlation between the expression of stromal TILs and pathological complete response (pCR) after NAC in BC patients. METHODS The PubMed, Embase, Cochrane Library, and Web of Science databases were searched online by using a combination of keywords and free words to screen literature on the expression of stromal TILs and pCR after NAC in patients with BC. The data were extracted and evaluated for quality. Relative risk (RR) was used to evaluate the relationship between the expression of stromal TILs before NAC and pCR in BC patients. Meta-analysis was performed with Review Manager 5.3 and STATA 14.0 software. RESULTS Eleven studies involving 6039 BC patients were included in the meta-analysis. The results showed a generally high expression of stromal TILs in BC patients, and the pCR rate after NAC in BC patients with a high expression of stromal TILs was significantly higher than that in BC patients with a low expression of stromal TILs [RR = 1.83, 95% confidence interval (CI): 1.69-1.97]. Subgroup analysis based on the molecular subtypes of BC showed that the pCR rate was significantly higher in patients with a high expression of stromal TILs in hormone receptor (HR)-positive BC [RR = 3.23, 95% CI: 2.43-4.30], human epidermal growth factor receptor 2 (HER-2)-positive BC [RR = 1.41, 95% CI: 1.25-1.60], and triple-negative BC [RR = 1.70, 95% CI: 1.53-1.90] than in those with a low expression of stromal TILs. Subgroup analysis based on expression threshold showed that the pCR rate was higher in patients with a high expression of stromal TILs than in patients with a low expression of stromal TILs at different expression thresholds (10% [RR = 1.99, 95% CI: 1.55-2.55], 20%/30% [RR = 1.57, 95% CI: 1.37-1.81], 50%/60% [RR = 1.91, 95% CI: 1.73-2.11]. CONCLUSION TILs can be used as a predictor of pCR after NAC in patients with BC, and the appropriate high expression threshold of stromal TILs should be selected as the predictive value according to the molecular subtype of BC.
Collapse
Affiliation(s)
- Guangfa Xia
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Ziran Zhang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Qin Jiang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Huan Wang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Jie Wang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
14
|
Koppiker CB, Kelkar DA, Kulkarni M, Kadu S, Pai M, Dhar U, Deshmukh C, Varghese B, Zamre V, Jumle N, Gangurde N, Joshi A, Unde R, Banale R, Namewar N, Vaid P, Busheri L, Thomas G, Nare S, Pereira J, Badve S. Impact of oncoplasty in increasing breast conservation rates Post neo-adjuvant chemotherapy. Front Oncol 2023; 13:1176609. [PMID: 37746279 PMCID: PMC10514208 DOI: 10.3389/fonc.2023.1176609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/19/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction The essential goal of neoadjuvant chemotherapy (NACT) is to downstage the primary tumor making it amenable for breast conservation surgery (BCS). However, since the safety of this surgery is paramount, post-NACT breast conservation rates remain low. As per the recommendation of the 2018 Early Breast Cancer Trialists' Collaborative Group (EBCTCG) overview of long-term post-NACT follow-up, we have devised a protocol for imaging, localization, rad-path analysis, and documentation of radiotherapy techniques to ensure the safety of post-NACT breast conservation. Methods This is a retrospective cohort of 180 breast cancer patients who received NACT and were operated on by a single surgical oncologist from 2015 to 2020. After selection based on published guidelines, patients were treated with neoadjuvant systemic (chemo or hormone) therapy. In cases where primary tumors responded and reduced to 1-2 cm in size mid-NACT, the residual tumors were localized by clips under ultrasound guidance and calcification was wire localized. All patients were treated using appropriate surgical and oncoplastic techniques where indicated. Negative margins were ensured by intra-operative rad-path analysis. Adjuvant chemotherapy and radiotherapy were given as per protocol. Results In 81 cases that required mastectomy at presentation, we were able to achieve a 72.8% post-NACT BCS rate with the help of oncoplasty. Overall, 142 of 180 (80%) patients were treated with breast conserving surgery of which 80% (121 of 142) were oncoplasty. Margins were assessed on intra-operative frozen and re-excised in the same setting. No positive margins were reported in final histopath of 142 breast conservation procedures. Post-operative complication rates after breast conservation in the first year were at 17% (24 of 142 including two major complications). Patient reported outcomes were satisfactory with increased satisfaction for breast conservation compared with immediate breast reconstruction. Discussion Employing oncoplastic breast surgery (OBS) techniques following stringent protocols for accurate localization of the residual tumor, intra-operative rad-path analysis, and adjuvant treatments, we show successful breast conservation in 72.8% of our mastectomy-qualified patients after downstaging by NACT. We also report satisfactory outcomes for post-NACT surgery, patient-reported satisfaction, and survival.
Collapse
Affiliation(s)
- Chaitanyanand B. Koppiker
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
- Department of Onco-Sciences, Jehangir Hospital, Pune, India
- International School of Oncoplasty, Pune, India
- Orchids Breast Health Centre, A Prashanti Cancer Care Mission (PCCM) Initiative, Pune, India
| | - Devaki A. Kelkar
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Madhura Kulkarni
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Shweta Kadu
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Mugdha Pai
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Upendra Dhar
- Department of Onco-Sciences, Jehangir Hospital, Pune, India
- Orchids Breast Health Centre, A Prashanti Cancer Care Mission (PCCM) Initiative, Pune, India
| | - Chetan Deshmukh
- Department of Onco-Sciences, Jehangir Hospital, Pune, India
- Orchids Breast Health Centre, A Prashanti Cancer Care Mission (PCCM) Initiative, Pune, India
| | - Beenu Varghese
- Department of Onco-Sciences, Jehangir Hospital, Pune, India
- Orchids Breast Health Centre, A Prashanti Cancer Care Mission (PCCM) Initiative, Pune, India
| | | | - Nutan Jumle
- Department of Onco-Sciences, Jehangir Hospital, Pune, India
| | - Nutan Gangurde
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Anjali Joshi
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Rohini Unde
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Rituja Banale
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Namrata Namewar
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Pooja Vaid
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
- Ashoka University – Department of Biology, Ashoka University, Haryana, India
| | | | - George Thomas
- Orchids Breast Health Centre, A Prashanti Cancer Care Mission (PCCM) Initiative, Pune, India
| | - Smeeta Nare
- Prashanti Cancer Care Mission, Pune, India
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and Indian Institute of Science Education and Research (IISER), Pune, India
| | - Jerome Pereira
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Sunil Badve
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, United States
| |
Collapse
|
15
|
Gucin Z, Buyukpinarbasili N, Gecer MO, Ersoy YE, Turk HM, Yildiz S, Aksoy DO. Stem cell markers: A guide to neoadjuvant therapy in breast carcinomas. INDIAN J PATHOL MICR 2023; 66:495-501. [PMID: 37530329 DOI: 10.4103/ijpm.ijpm_1274_21] [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] [Indexed: 08/03/2023] Open
Abstract
Aim This study aims to investigate potential associations between the stem cell population and the degree of tumor regression in breast carcinomas treated with neoadjuvant therapy. Settings and Design The study included 92 patients with breast carcinoma who received neoadjuvant therapy. Tumor regression was defined based on Miller and Payne grading system. Patients with grade 1 or 2 regression on a 5-point scale were included in group 1 (n = 37), grade 3 regression in group 2 (n = 32), and grade 4 or 5 regression in group 3 (n = 23). Materials and Methods Immunohistochemical staining was performed on paraffin block sections of every case using CD44, CD24, CD29, CD133, ID4, and ALDH1 antibodies to detect stem cells. Statistical Analysis Used IBM Statistical Package for the Social Sciences (SPSS), version 23.0 (IBM Corp., Armonk, NY, USA) software was used for statistical analyses, and a P value less than 0.05 was considered statistically significant. Results Histologically high-grade tumors are more common in the near-complete/complete response group (P = 0.004). HER2-positive tumors were more common in the complete/near-complete response group (P = 0.054). Tumor cells positive for stem cell markers CD44 and CD24 were more common in the poor response group (P = 0.027 and P = 0.001, respectively). CD29 expression was reduced in the posttreatment residual tumor tissue in the near-complete/complete response group. Conclusion High CD44 and CD24 expression may be a predictor of poor response/nonresponse to neoadjuvant therapy in breast carcinomas. Background In recent years, stem cells have been defined as the main cell population responsible for resistance to anticancer therapies.
Collapse
Affiliation(s)
- Zuhal Gucin
- Department of Pathology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Nur Buyukpinarbasili
- Department of Ministry of Health, Cam Sakura City Hospital, Department of Pathology, Istanbul, Turkey
| | - Melin Ozgun Gecer
- Department of Pathology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Yeliz Emine Ersoy
- Department of General Surgery, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Haci Mehmet Turk
- Department of Medical Oncology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Seyma Yildiz
- Department of Radiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Direnc Ozlem Aksoy
- Department of Ministry of Health, Istanbul Training and Research Hospital, Department of Radiology, Istanbul, Turkey
| |
Collapse
|
16
|
Li Z, Li Y, Liu Y, Shang Y, Zhou Y, Ji X, Han H, Du K, Zhang J. Real‑world data indicated that neoadjuvant chemotherapy alone was associated with a higher risk of tumor recurrence in high‑risk breast cancer subgroup patients. Oncol Lett 2023; 25:193. [PMID: 37113400 PMCID: PMC10126659 DOI: 10.3892/ol.2023.13779] [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: 09/28/2022] [Accepted: 02/14/2023] [Indexed: 03/31/2023] Open
Abstract
Numerous clinical trials have reported equal effects of tumor control between neoadjuvant chemotherapy (NAC) and adjuvant chemotherapy (AC) in patients with breast cancer (BC). However, this conclusion has not been verified in practice. The present retrospective study evaluated if there were different risk profiles for NAC, AC and their combinative modes on disease-free survival (DFS) in patients with BC using real-world data. All women with primary unilateral Stage I-III BC and first recurrence in 2008-2018 at The Fourth Hospital of Hebei Medical University were retrospectively identified for enrollment. The four modes of chemotherapy administered for primary BC were classified as 'None', 'NAC only', 'NAC+AC' and 'AC only'. One multivariate Cox model was used to estimate the adjusted Hazard Ratio (HR) and P-value. Covariates included age, Easter Cooperative Oncology Group grade, T stage, N stage, pathology, grade, lymphovascular invasion (LVI), BC subtype, number of chemotherapy cycles and other therapies. Amongst 637 patients, who had a mean age of 48.2 years at BC diagnosis and 50.9 years at recurrence, the median DFS by the 'None' (n=27), 'NAC only' (n=47), 'NAC+AC' (n=118) and 'AC only' (n=445) modes were 31.4, 16.6, 22.6 and 28.4 months (P<0.001), respectively. Compared with the 'AC only', adjusted HR (P-value) of the 'None', 'NAC only' and 'NAC+AC' modes on tumor recurrence were 1.182 (0.551), 1.481 (0.037) and 1.102 (0.523), respectively. The adjusted HR of 'NAC only' vs. 'AC only' modes were 1.448 (P=0.157) for locoregional recurrence and 2.675 (P=0.003) for distant recurrence. Stratified analyses further indicated that the 'NAC only' mode was associated with a higher recurrence risk in T3-4, N2-3, LVI-positive, or HER2-negative subgroup patients. In conclusion, NAC alone was associated with a higher risk of tumor recurrence in high-risk BC subgroup patients in real-world data. Patient selection of chemotherapy mode was involved in practice but could not fully explain this finding. The 'inadequate' NAC was highly likely to have accounted for this observation.
Collapse
Affiliation(s)
- Zhensheng Li
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Yue Li
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Yunjiang Liu
- Department of Breast Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Yuguang Shang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Yarong Zhou
- Department of Breast Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Xiaohui Ji
- Department of Ultrasound, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Huina Han
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Kaiye Du
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| | - Jun Zhang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050035, P.R. China
| |
Collapse
|
17
|
Zeng L, Liu L, Chen D, Lu H, Xue Y, Bi H, Yang W. The innovative model based on artificial intelligence algorithms to predict recurrence risk of patients with postoperative breast cancer. Front Oncol 2023; 13:1117420. [PMID: 36959794 PMCID: PMC10029918 DOI: 10.3389/fonc.2023.1117420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Purpose This study aimed to develop a machine learning model to retrospectively study and predict the recurrence risk of breast cancer patients after surgery by extracting the clinicopathological features of tumors from unstructured clinical electronic health record (EHR) data. Methods This retrospective cohort included 1,841 breast cancer patients who underwent surgical treatment. To extract the principal features associated with recurrence risk, the clinical notes and histopathology reports of patients were collected and feature engineering was used. Predictive models were next conducted based on this important information. All algorithms were implemented using Python software. The accuracy of prediction models was further verified in the test cohort. The area under the curve (AUC), precision, recall, and F1 score were adopted to evaluate the performance of each model. Results A training cohort with 1,289 patients and a test cohort with 552 patients were recruited. From 2011 to 2019, a total of 1,841 textual reports were included. For the prediction of recurrence risk, both LSTM, XGBoost, and SVM had favorable accuracies of 0.89, 0.86, and 0.78. The AUC values of the micro-average ROC curve corresponding to LSTM, XGBoost, and SVM were 0.98 ± 0.01, 0.97 ± 0.03, and 0.92 ± 0.06. Especially the LSTM model achieved superior execution than other models. The accuracy, F1 score, macro-avg F1 score (0.87), and weighted-avg F1 score (0.89) of the LSTM model produced higher values. All P values were statistically significant. Patients in the high-risk group predicted by our model performed more resistant to DNA damage and microtubule targeting drugs than those in the intermediate-risk group. The predicted low-risk patients were not statistically significant compared with intermediate- or high-risk patients due to the small sample size (188 low-risk patients were predicted via our model, and only two of them were administered chemotherapy alone after surgery). The prognosis of patients predicted by our model was consistent with the actual follow-up records. Conclusions The constructed model accurately predicted the recurrence risk of breast cancer patients from EHR data and certainly evaluated the chemoresistance and prognosis of patients. Therefore, our model can help clinicians to formulate the individualized management of breast cancer patients.
Collapse
Affiliation(s)
- Lixuan Zeng
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Lei Liu
- Department of Breast Surgery, The Third Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongxin Chen
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Henghui Lu
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Xue
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Hongjie Bi
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Weiwei Yang
- Department of Pathology, Harbin Medical University, Harbin, China
| |
Collapse
|
18
|
Yang J, Deng Y, Liu D, Tan Y, Lin M, Zhou X, Zhang J, Yu H, Hu Y, Tang Y, Jiang S, Zhang J. Brain network deficits in breast cancer patients after early neoadjuvant chemotherapy: A longitudinal MRI study. J Neurosci Res 2023; 101:1138-1153. [PMID: 36791216 DOI: 10.1002/jnr.25178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
Breast cancer (BC) patients who undergo chemotherapy are likely to develop chemotherapy-related cognitive impairment (CRCI). Recent studies of BC patients after chemotherapy have used graph theory to investigate the topological properties of the brain functional connectome. However, little is known about structural morphological networks in BC patients after early neoadjuvant chemotherapy (NAC). Brain morphological network organization in 47 female participants with BC was investigated before and after NAC. Topological properties of brain networks were ascertained based on morphological similarities in regional gray matter using a graph theory approach based on 3D T1-weighted MRI data. Nonparametric permutation testing was used to assess longitudinal-group differences in topological metrics. Compared with BC patients before NAC, BC patients after early NAC showed significantly increased global efficiency (p = .048), decreased path length (p = .033), and abnormal nodal properties and connectivity, mainly located in the central executive network (CEN). The change in the network efficiency of the right caudate was negatively correlated with the change in the Self-Rating Anxiety Scale score (r = -.435, p = .008), and the change in the nodal degree of the left superior frontal gyrus (dorsolateral part) was positively correlated with the change in the Functional Assessment of Cancer Therapy score (r = .547, p = .002). BC participants showed randomization in global properties and dysconnectivity in the CEN after early NAC. NAC may disrupt the cognitive balance of the brain morphological network in individuals with BC.
Collapse
Affiliation(s)
- Jing Yang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yongchun Deng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, School of Medicine, Chongqing, China.,Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, School of Medicine, Chongqing, China
| | - Daihong Liu
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yong Tan
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Meng Lin
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Xiaoyu Zhou
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Jing Zhang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Hong Yu
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yixin Hu
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yu Tang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Shixi Jiang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
19
|
Memişoğlu E, Sarı R. The effect of positron emission tomography/computed tomography in axillary surgery approach after neoadjuvant treatment in breast cancer. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:37-43. [PMID: 36629643 PMCID: PMC9937613 DOI: 10.1590/1806-9282.20220097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The aim of this study was to determine the role of positron emission tomography/computed tomography in the decision to perform axillary surgery by comparing positron emission tomography/computed tomography findings with pathology consistency after neoadjuvant chemotherapy. METHODS Patients who were diagnosed for T1-4, cN1/2 breast cancer receiving neoadjuvant chemotherapy in our clinic between January 2016 and February 2021 were evaluated. Clinical and radiological responses, axillary surgery, and histopathological results after neoadjuvant chemotherapy were evaluated. RESULTS Axillary involvement was not detected in positron emission tomography/computed tomography after neoadjuvant chemotherapy in 140 (60.6%) of 231 node-positive patients. In total, 88 (62.8%) of these patients underwent sentinel lymph node biopsy, and axillary lymph node dissection was performed in 29 (33%) of these patients upon detection of 1 or 2 positive lymph nodes. The other 52 (37.1%) patients underwent direct axillary lymph node dissection, and no metastatic lymph nodes were detected in 33 (63.4%) patients. No metastatic lymph node was found pathologically in a total of 92 patients without involvement in positron emission tomography/computed tomography, and the negative predictive value was calculated as 65.7%. Axillary lymph node dissection was performed in 91 (39.4%) patients with axillary involvement in positron emission tomography/computed tomography after neoadjuvant chemotherapy. Metastatic lymph nodes were found pathologically in 83 of these patients, and the positive predictive value was calculated as 91.2%. CONCLUSION Positron emission tomography/computed tomography was found to be useful in the evaluation of clinical response, but it was not sufficient enough to predict a complete pathological response. When planning axillary surgery, axillary lymph node dissection should not be decided only with a positive positron emission tomography/computed tomography. Other radiological images should also be evaluated, and a positive sentinel lymph node biopsy should be the determinant of axillary lymph node dissection.
Collapse
Affiliation(s)
- Ecem Memişoğlu
- Kartal Dr. Lütfi Kirdar Şehir Hastanesi, Department of General Surgery – Istanbul, Turkey
| | - Ramazan Sarı
- Kartal Dr. Lütfi Kirdar Şehir Hastanesi, Department of General Surgery – Istanbul, Turkey
| |
Collapse
|
20
|
Lv Y, Bai Z, Wang X, Liu J, Li Y, Zhang X, Shan Y. Comprehensive evaluation of breast cancer immunotherapy and tumor microenvironment characterization based on interleukin genes-related risk model. Sci Rep 2022; 12:20524. [PMID: 36443508 PMCID: PMC9705306 DOI: 10.1038/s41598-022-25059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Breast cancer (BRCA) is the most prevalent malignancy and the leading cause of death in women. Interleukin (IL) genes are critical in tumor initiation and control. Nevertheless, the prognosis value of the IL in BRCA remains unclear. We collected data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), and 94 IL genes were identified from GeneCard. Based on the random forest (RF), least absolute shrinkage and selection operator (LASSO) analysis, and multivariate Cox regression analysis, we constructed an IL signature. GSE22219, GSE25065, and GSE21653 were derived as validation sets. The expression differences in the tumor microenvironment (TME), immunotherapy, and chemosensitivity of BRCA between the high- and low-risk groups were evaluated. Overall, 21 IL genes were selected to construct an IL risk model, of which IL18BP, IL17D, and IL23A were the first time identified as prognostic genes in BRCA. IL score could distinguish BRCA patients with inferior outcomes, and AUC of it was 0.70, 0.76, and 0.72 for 1-,3- and 5- years, respectively, which was also verified in GSE22219, GSE25065, and GSE21653 cohorts. Meanwhile, compared to luminal A and luminal B, HER2-positive and TNBC had significantly higher IL score. Besides, the high-risk group had a significantly higher prevalence of TP53 and TTN but a lower prevalence of PIK3CA, as well as higher tumor mutation burden (TMB) and neoantigen level. High- and low-risk groups exhibited notable differences in immunomodulators and tumor infiltrates immune cells (TIICs), and the high-risk group had significantly lower Tumor Immune Dysfunction and Exclusion (TIDE) score. Additionally, the high-risk group has more responders to immune or anti-HER2 combination therapy, whereas the low-risk group has higher sensitivity to docetaxel and paclitaxel. Consequently, we constructed a reliable risk model based on the IL genes, which can provide more information on both the risk stratification and personalizing management strategies for BRCA.
Collapse
Affiliation(s)
- Yalei Lv
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China
| | - Zihe Bai
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China
| | - Xiaoyan Wang
- The Fifth Ward of Medical Oncology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Jiayin Liu
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China
| | - Yuntao Li
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, Hebei Medical University, Shijiazhuang, China
| | - Yujie Shan
- Department of Medical Oncology, Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, China.
| |
Collapse
|
21
|
Liu Q, Tang L, Chen M. Ultrasound Strain Elastography and Contrast-Enhanced Ultrasound in Predicting the Efficacy of Neoadjuvant Chemotherapy for Breast Cancer: A Nomogram Integrating Ki-67 and Ultrasound Features. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:2191-2201. [PMID: 34888900 DOI: 10.1002/jum.15900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/27/2021] [Accepted: 11/19/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVES To explore whether conventional elastography and contrast-enhanced ultrasound (CEUS) combined with histopathology can monitor the efficacy of neoadjuvant chemotherapy (NAC) for breast cancer (BC), and develop a Nomogram prediction model monitoring response to NAC. METHODS From February 2010 to November 2015, 91 BC patients who received NAC were recruited. The maximum diameter, stiffness, and CEUS features were assessed. Core biopsy, surgical pathology immunophenotype, and Miller-Payne (MP) evaluation were documented. Univariate and multivariate analysis was performed using receiver operating characteristic (ROC) analysis and logistic regression analysis. RESULTS There were 37 cases showing pathological complete response (pCR) and 54 of non-pCR. The changes of maximal diameter were correlated with MP (P < .05). The sensitivity (SEN), specificity (SPE), and area under the ROC curve (AUC) of baseline size predicting pCR were 57.40%, 70.30%, and 0.64 (P = .024). Baseline Ki-67 index of pCR group is significantly higher than that of non-pCR group (P = .029), and the ROC analysis of baseline Ki-67 indicates the SEN, SPE, and AUC of 51.70%, 78.00%, and 0.638 (P = .050). When combined with size, CEUS features, stiffness, and Ki-67 of baseline, the ROC curve shows good performance with SEN, SPE, and AUC of 70.00%, 76.19%, 0.821 (P = .004). Incorporating the change of characteristics into multivariate regression analysis, the results demonstrate excellent performance (SEN 100.00%, SPE 95.24%, AUC 0.986, P = .000). CONCLUSIONS The change of the maximum size was correlated with MP score, which can provide reference to predict efficacy of NAC and evaluate residual lesions. When combining with elastography, CEUS, and Ki-67, better performance in predicting pathological response was shown.
Collapse
Affiliation(s)
- Qi Liu
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Tang
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man Chen
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Zhou Y, Tian Q, Gao H, Zhu L, Yang J, Zhang J, Yang J. Correlation Between Immune-Related Genes and Tumor-Infiltrating Immune Cells With the Efficacy of Neoadjuvant Chemotherapy for Breast Cancer. Front Genet 2022; 13:905617. [PMID: 35754838 PMCID: PMC9214242 DOI: 10.3389/fgene.2022.905617] [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: 03/27/2022] [Accepted: 05/03/2022] [Indexed: 11/15/2022] Open
Abstract
Background: In the absence of targeted therapy or clear clinically relevant biomarkers, neoadjuvant chemotherapy (NAC) is still the standard neoadjuvant systemic therapy for breast cancer. Among the many biomarkers predicting the efficacy of NAC, immune-related biomarkers, such as immune-related genes and tumor-infiltrating lymphocytes (TILs), play a key role. Methods: We analyzed gene expression from several datasets in the Gene Expression Omnibus (GEO) database and evaluated the relative proportion of immune cells using the CIBERSORT method. In addition, mIHC/IF detection was performed on clinical surgical specimens of triple-negative breast cancer patients after NAC. Results: We obtained seven immune-related genes, namely, CXCL1, CXCL9, CXCL10, CXCL11, IDO1, IFNG, and ORM1 with higher expression in the pathological complete response (pCR) group than in the non-pCR group. In the pCR group, the levels of M1 and γδT macrophages were higher, while those of the M2 macrophages and mast cells were lower. After NAC, the proportions of M1, γδT cells, and resting CD4 memory T cells were increased, while the proportions of natural killer cells and dendritic cells were decreased with downregulated immune-related genes. The results of mIHC/IF detection and the prognostic information of corresponding clinical surgical specimens showed the correlation of proportions of natural killer cells, CD8-positive T cells, and macrophages with different disease-free survival outcomes. Conclusion: The immune-related genes and immune cells of different subtypes in the tumor microenvironment are correlated with the response to NAC in breast cancer, and the interaction between TILs and NAC highlights the significance of combining NAC with immunotherapy to achieve better clinical benefits.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qi Tian
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lizhe Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiao Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Juan Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Mercado C, Chhor C, Scheel JR. MRI in the Setting of Neoadjuvant Treatment of Breast Cancer. JOURNAL OF BREAST IMAGING 2022; 4:320-330. [PMID: 38422421 DOI: 10.1093/jbi/wbab059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 03/02/2024]
Abstract
Neoadjuvant therapy may reduce tumor burden preoperatively, allowing breast conservation treatment for tumors previously unresectable or requiring mastectomy without reducing disease-free survival. Oncologists can also use the response of the tumor to neoadjuvant chemotherapy (NAC) to identify treatment likely to be successful against any unknown potential distant metastasis. Accurate preoperative estimations of tumor size are necessary to guide appropriate treatment with minimal delays and can provide prognostic information. Clinical breast examination and mammography are inaccurate methods for measuring tumor size after NAC and can over- and underestimate residual disease. While US is commonly used to measure changes in tumor size during NAC due to its availability and low cost, MRI remains more accurate and simultaneously images the entire breast and axilla. No method is sufficiently accurate at predicting complete pathological response that would obviate the need for surgery. Diffusion-weighted MRI, MR spectroscopy, and MRI-based radiomics are emerging fields that potentially increase the predictive accuracy of tumor response to NAC.
Collapse
Affiliation(s)
- Cecilia Mercado
- NYU Grossman School of Medicine, Department of Radiology, New York, NY, USA
| | - Chloe Chhor
- NYU Grossman School of Medicine, Department of Radiology, New York, NY, USA
| | - John R Scheel
- University of Washington, Department of Radiology, Seattle, WA, USA
| |
Collapse
|
24
|
Li F, Zhao Y, Wei Y, Xi Y, Bu H. Tumor-Infiltrating Lymphocytes Improve Magee Equation-Based Prediction of Pathologic Complete Response in HR-Positive/HER2-Negative Breast Cancer. Am J Clin Pathol 2022; 158:291-299. [PMID: 35486808 DOI: 10.1093/ajcp/aqac041] [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: 11/13/2021] [Accepted: 04/07/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Magee equation 3 (ME3) is predictive of the pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) in patients with hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer but with insufficient predictive performance. This study was designed to improve predictive ability by combining ME3 with additional clinicopathologic markers. METHODS We retrospectively enrolled 460 patients with HR-positive/HER2-negative breast cancer from 2 centers. We obtained baseline characteristics, the ME3 score, and the number of stromal tumor-infiltrating lymphocytes (sTILs). After performing a logistic regression analysis, a predictive nomogram was built and validated externally. RESULTS ME3 score (adjusted odds ratio [OR], 1.14 [95% confidence interval (CI), 1.10-1.17]; P < .001) and TILs (adjusted OR, 5.21 [95% CI, 3.33-8.14]; P < .001) were independently correlated with pCR. The nomogram (named ME3+) was established using ME3 and sTILs, and it demonstrated an area under the curve of 0.816 and 0.862 in internal and external validation, respectively, outperforming the ME3 score alone. sTILs and ME3 scores were also found to be positively correlated across the entire cohort (P < .001). CONCLUSIONS The combination of sTILs and ME3 score potentially shows better performance for predicting pCR than ME3 alone. Larger validations are required for widespread application of ME3+ nomogram in NAC settings for HR-positive/HER2-negative breast cancer.
Collapse
Affiliation(s)
- Fengling Li
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Zhao
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, China
| | - Yani Wei
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Zheng CH, Xu K, Shan WP, Zhang YK, Su ZD, Gao XJ, Wang YJ, Qi JY, Ding XY, Wang CP, Wang YS. Meta-Analysis of Shrinkage Mode After Neoadjuvant Chemotherapy for Breast Cancers: Association With Hormonal Receptor. Front Oncol 2022; 11:617167. [PMID: 35444932 PMCID: PMC9014257 DOI: 10.3389/fonc.2021.617167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background Patients with concentric shrinkage mode after neoadjuvant chemotherapy (NAC) is considered to be ideal candidates for breast conserving treatment (BCT). While, what proportion of patients would represent CSM have not been well defined. This study was conducted to pool the rates of concentric shrinkage mode (CSM) in patients undergoing NAC, determine the impact of hormonal receptor on the shrinkage mode after NAC and estimate the rates of the CSM in various subgroups. Methods We conducted a systematic review following the guidelines for Meta-Analyses and Systematic reviews for the PRISMA guidelines. We systematically searched the literature about shrinkage mode after NAC from PubMed, Web of Science, Embase, The Cochrane Library, CNKI, Wanfang database published from January 2002 to June 2020 on breast cancer shrinkage mode after NAC and carefully screened the literature by using eligibility criteria: (1) patients with primary breast cancer treated with NAC; (2) publications with available data of shrinkage mode measured by magnetic resonance imaging (MRI), or data of pathology and hormonal receptor. The association between shrinkage mode and hormonal receptor was estimated using Stata 15.1 software. Results This analysis included a total of 2434 tumors from 23 papers. The included studies were heterogeneous (I2 = 89.4%, P<0.01). Random effects model was used to estimate the overall rates of CSM: 56.6% [95%CI (50.5%, 62.7%)]. According to the analysis of hormonal receptor, 10 of the paper was included for HR+ (hormone receptor positive) type analysis and the rate of CSM for HR+ type was 45.7% [95%CI (36.4%, 55.0%)]; 9 of the paper was used for HR- type (hormone receptor negative) analysis and the incidence of HR-CSM is 63.1% [95%CI (50.0%, 76.1%)]; with HR+ type as the control, the OR of the HR- CSM rate is 2.32 (1.32, 4.08) folds of HR+ type. From subgroup analyses, the CSM% of luminal A, luminal B, Her2+, and triple negative were 29.7% (16.5%, 42.8%); 47.2% (19.1%, 75.3%); 59.0% (39.7%, 78.3%); 66.2% (52.8%, 79.6%), respectively. Conclusions Breast cancer patients undergoing NAC did not get an ideal odds ratio of CSM. The incidence of CSM in breast cancer after NAC is associated with hormonal receptor. Patients with triple-negative breast cancers have the highest rates of CSM after NAC. More care should be taken to select patients with the luminal subtypes for BCT throughout NAC.
Collapse
Affiliation(s)
- Chun-Hui Zheng
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Department of Breast Surgery, Weifang People's Hospital, Weifang, China
| | - Kai Xu
- Department of Preventive Medicine, Weifang Medical University, Weifang, China.,Department of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan, China
| | - Wen-Ping Shan
- Department of Preventive Medicine, Weifang Medical University, Weifang, China
| | - Ya-Kun Zhang
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Zhi-De Su
- Department of Pharmacy, Weifang People's Hospital, Weifang, China
| | - Xiang-Jin Gao
- Department of Preventive Medicine, Weifang Medical University, Weifang, China
| | - Yu-Jue Wang
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, United States
| | - Jian-Yu Qi
- Department of Preventive Medicine, Weifang Medical University, Weifang, China
| | - Xiao-Yan Ding
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang, China
| | - Chun-Ping Wang
- Department of Preventive Medicine, Weifang Medical University, Weifang, China
| | - Yong-Sheng Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
26
|
Deng H, Xiao B, Huang Y, Weng K, Chen J, Li K, Wu H, Luo S, Hao W. The Combined Use of Orf Virus and PAK4 Inhibitor Exerts Anti-tumor Effect in Breast Cancer. Front Microbiol 2022; 13:845259. [PMID: 35401439 PMCID: PMC8984157 DOI: 10.3389/fmicb.2022.845259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
The parapoxvirus Orf virus (ORFV) has long been recognized as one of the valuable vectors in researches of oncolytic virus. In order to develop a potential therapeutic strategy for breast cancer based on the oncolytic virotherapy via ORFV, firstly we explore the oncolytic effects of ORFV. Our research showed that ORFV exerts anti-tumor effects in vitro by inducing breast cancer cell G2/M phase arrest and cell apoptosis. In vivo experiments were carried out, in which we treated 4T1 tumor-bearing BALB/C mice via intratumoral injection of ORFV. ORFV can exert anti-tumor activity by regulating tumor microenvironment (TME) and inducing a host immune response plus directly oncolytic effect. The CRISPR-Cas9 knockout library targeting 507 kinases was used to screen out PAK4, which is beneficial to the anti-tumor effect of ORFV on breast cancer cells. PF-3758309 is a potent PAK4-targeted inhibitor. Co-using of ORFV and PF-3758309 as a combination treatment produces its anti-tumor effects through inhibition of cell viability, induction of apoptosis and suppression of cell migration and invasion in vitro. The results of in vivo experiments showed that the tumor growth of mice in the combination treatment group was significantly inhibited, which proved that the combination treatment exerts an effective anti-tumor effect in vivo. In summary, we have clarified the oncolytic effect of ORFV on breast cancer, and found that the combination of ORFV and PAK4 inhibitor can effectively improve the oncolytic effect of ORFV. We hope our research could provide a new idea for the development of new treatment strategies for breast cancer.
Collapse
Affiliation(s)
- Hao Deng
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Southern Medical University, Guangzhou, China
| | - Bin Xiao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Yinger Huang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Southern Medical University, Guangzhou, China
| | - Kongyan Weng
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Southern Medical University, Guangzhou, China
- Department of Transfusion Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jialing Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Southern Medical University, Guangzhou, China
| | - Kun Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Southern Medical University, Guangzhou, China
| | - Hongfeng Wu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Southern Medical University, Guangzhou, China
| | - Shuhong Luo
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, China
- *Correspondence: Shuhong Luo,
| | - Wenbo Hao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, China
- Wenbo Hao,
| |
Collapse
|
27
|
Bhardwaj D, Dasgupta A, DiCenzo D, Brade S, Fatima K, Quiaoit K, Trudeau M, Gandhi S, Eisen A, Wright F, Look-Hong N, Curpen B, Sannachi L, Czarnota GJ. Early Changes in Quantitative Ultrasound Imaging Parameters during Neoadjuvant Chemotherapy to Predict Recurrence in Patients with Locally Advanced Breast Cancer. Cancers (Basel) 2022; 14:cancers14051247. [PMID: 35267555 PMCID: PMC8909335 DOI: 10.3390/cancers14051247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND This study was conducted to explore the use of quantitative ultrasound (QUS) in predicting recurrence for patients with locally advanced breast cancer (LABC) early during neoadjuvant chemotherapy (NAC). METHODS Eighty-three patients with LABC were scanned with 7 MHz ultrasound before starting NAC (week 0) and during treatment (week 4). Spectral parametric maps were generated corresponding to tumor volume. Twenty-four textural features (QUS-Tex1) were determined from parametric maps acquired using grey-level co-occurrence matrices (GLCM) for each patient, which were further processed to generate 64 texture derivatives (QUS-Tex1-Tex2), leading to a total of 95 features from each time point. Analysis was carried out on week 4 data and compared to baseline (week 0) data. ∆Week 4 data was obtained from the difference in QUS parameters, texture features (QUS-Tex1), and texture derivatives (QUS-Tex1-Tex2) of week 4 data and week 0 data. Patients were divided into two groups: recurrence and non-recurrence. Machine learning algorithms using k-nearest neighbor (k-NN) and support vector machines (SVMs) were used to generate radiomic models. Internal validation was undertaken using leave-one patient out cross-validation method. RESULTS With a median follow up of 69 months (range 7-118 months), 28 patients had disease recurrence. The k-NN classifier was the best performing algorithm at week 4 with sensitivity, specificity, accuracy, and area under curve (AUC) of 87%, 75%, 81%, and 0.83, respectively. The inclusion of texture derivatives (QUS-Tex1-Tex2) in week 4 QUS data analysis led to the improvement of the classifier performances. The AUC increased from 0.70 (0.59 to 0.79, 95% confidence interval) without texture derivatives to 0.83 (0.73 to 0.92) with texture derivatives. The most relevant features separating the two groups were higher-order texture derivatives obtained from scatterer diameter and acoustic concentration-related parametric images. CONCLUSIONS This is the first study highlighting the utility of QUS radiomics in the prediction of recurrence during the treatment of LABC. It reflects that the ongoing treatment-related changes can predict clinical outcomes with higher accuracy as compared to pretreatment features alone.
Collapse
Affiliation(s)
- Divya Bhardwaj
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Archya Dasgupta
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Daniel DiCenzo
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Stephen Brade
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Kashuf Fatima
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Karina Quiaoit
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Maureen Trudeau
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Sonal Gandhi
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Andrea Eisen
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (M.T.); (S.G.); (A.E.)
- Department of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Frances Wright
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.W.); (N.L.-H.)
- Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Nicole Look-Hong
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.W.); (N.L.-H.)
- Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Medical Imaging, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Lakshmanan Sannachi
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; (D.B.); (A.D.); (D.D.); (S.B.); (K.F.); (K.Q.); (L.S.)
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M4N 3M5, Canada
- Correspondence: ; Tel.: +416-480-6128
| |
Collapse
|
28
|
Liu H, Lv L, Gao H, Cheng M. Pathologic Complete Response and Its Impact on Breast Cancer Recurrence and Patient's Survival after Neoadjuvant Therapy: A Comprehensive Meta-Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:7545091. [PMID: 35003324 PMCID: PMC8741368 DOI: 10.1155/2021/7545091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Earlier research has illustrated prognostic significance of pathologic complete response (pCR) in neoadjuvant therapy (NAT) for breast cancer, whereas correlation between treatment after achieving pCR and survival improvement remains underexplored. We attempted to measure the relation between pCR achieved after NAT and breast cancer recurrence or patient's survival. METHODS We searched PubMed, EMBASE, Web of Science, and The Cochrane Library databases to find relevant articles from their inception to November 2020. According to eligibility criteria, studies were selected and basic data were extracted. The primary endpoint was the correlation between pCR achieved after NAT and event-free survival (EFS) or overall survival (OS). The results were obtained by directly extracting specific information from the literature or estimating individual data by survival curves on DigitizeIt software, presented with HR and 95% CI. All data were processed on Stata 14.0 software. RESULTS Among 4338 articles, there were 25 eligible articles involving 8767 patients. The EFS of patients achieved pCR after NAT improved obviously (HR = 0.27; 95% CI, 0.24-0.31), especially in triple negative (HR = 0.17; 95% CI, 0.12-0.24) and HER2 positive (HR = 0.24; 95% CI, 0.20-0.30) breast cancer patients. As such, pCR after NAT was implicated in significantly increased OS (HR = 0.32; 95% CI, 0.27-0.37). CONCLUSION Achieving pCR after NAT was notably related to the improvement of EFS and OS, especially for patients with triple-negative and HER2-positive breast cancer. pCR can be a surrogate indicator for outcome of breast cancer patients after NAT, as well as a predictor of treatment efficacy after NAT. Besides, well-designed studies are still warranted for confirmation.
Collapse
Affiliation(s)
- Hui Liu
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| | - Liqiong Lv
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| | - Hui Gao
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| | - Ming Cheng
- Department of Breast Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi Province, China
| |
Collapse
|
29
|
Li Y, Wang K, Chen Y, Cai J, Qin X, Lu A, Guan D, Qin G, Chen W. A System Pharmacology Model for Decoding the Synergistic Mechanisms of Compound Kushen Injection in Treating Breast Cancer. Front Pharmacol 2021; 12:723147. [PMID: 34899291 PMCID: PMC8660088 DOI: 10.3389/fphar.2021.723147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022] Open
Abstract
Breast cancer (BC) is one of the most common malignant tumors among women worldwide and can be treated using various methods; however, side effects of these treatments cannot be ignored. Increasing evidence indicates that compound kushen injection (CKI) can be used to treat BC. However, traditional Chinese medicine (TCM) is characterized by “multi-components” and “multi-targets”, which make it challenging to clarify the potential therapeutic mechanisms of CKI on BC. Herein, we designed a novel system pharmacology strategy using differentially expressed gene analysis, pharmacokinetics synthesis screening, target identification, network analysis, and docking validation to construct the synergy contribution degree (SCD) and therapeutic response index (TRI) model to capture the critical components responding to synergistic mechanisms of CKI in BC. Through our designed mathematical models, we defined 24 components as a high contribution group of synergistic components (HCGSC) from 113 potentially active components of CKI based on ADME parameters. Pathway enrichment analysis of HCGSC targets indicated that Rhizoma Heterosmilacis and Radix Sophorae Flavescentis could synergistically target the PI3K-Akt signaling pathway and the cAMP signaling pathway to treat BC. Additionally, TRI analysis showed that the average affinity of HCGSC and targets involved in the key pathways reached -6.47 kcal/mmol, while in vitro experiments proved that two of the three high TRI-scored components in the HCGSC showed significant inhibitory effects on breast cancer cell proliferation and migration. These results demonstrate the accuracy and reliability of the proposed strategy.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kexin Wang
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong SAR, China.,Neurosurgery Center, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yupeng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Key Laboratory of Biochip Technology, Southern Medical University, Guangzhou, China
| | - Jieqi Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Key Laboratory of Biochip Technology, Southern Medical University, Guangzhou, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Aiping Lu
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong SAR, China
| | - Daogang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Key Laboratory of Biochip Technology, Southern Medical University, Guangzhou, China
| | - Genggeng Qin
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiguo Chen
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Elevated Level of Nerve Growth Factor (NGF) in Serum-Derived Exosomes Predicts Poor Survival in Patients with Breast Cancer Undergoing Neoadjuvant Chemotherapy. Cancers (Basel) 2021; 13:cancers13215260. [PMID: 34771423 PMCID: PMC8582365 DOI: 10.3390/cancers13215260] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Exosomes and cytokines play crucial roles in the process of tumor progression. Recent studies have reported that cytokines can be packaged into exosomes, leading to drug resistance. The aim of this study is to evaluate the potential value of cytokines in both serum and exosomes as prognostic biomarkers of long-term outcomes in patients with breast cancer treated with neoadjuvant chemotherapy. We observed significant differences in expression patterns between serum cytokines and exosomal cytokines. Elevated levels of serum IP-10, serum MMP-1, and exosomal NGF were associated with poor overall survival. In multivariate analysis, exosomal NGF was an independent prognostic factor for overall survival. These findings suggest that exosomal NGF is useful for identifying patients with poor survival outcomes. Abstract Neoadjuvant chemotherapy (NAC) is a standard treatment strategy for patients with locally advanced breast cancer (LABC). However, there are no established predictors of chemosensitivity and survival in LABC patients who undergo NAC. Many studies have demonstrated that exosomes and cytokines are important players in intercellular communication between tumors and their environments, and are involved in chemotherapy resistance. Recently, it was reported that cytokines can be packaged into exosomes, but whether exosomal cytokines serve as biomarkers in breast cancer patients is still unclear. In this study, we examined the roles of cytokines in both serum and exosomes as prognostic biomarkers for long-term outcomes in patients with breast cancer who undergo NAC. We isolated exosomes from the blood of 129 patients with early breast cancer who were receiving neoadjuvant chemotherapy between 2008 and 2011 at Samsung Medical Center. The levels of cytokines and growth factors in serum and exosomes were measured with ProcartaPlex immune-related panels. We investigated correlations between clinic-pathologic variables and patient survival, and Cox proportional hazards regression analysis was performed for prognostic evaluation. We detected significant differences in expression patterns between serum cytokines and exosomal cytokines. In both serum and exosomes, many cytokines were positively correlated with age. In univariate analysis, patients with high serum IP-10, serum MMP-1, and exosomal NGF had shorter overall survival. Exosomal NGF showed significantly poorer overall survival in multivariate analysis. These findings suggest that exosomal NGF is useful for identifying patients with poor survival outcomes.
Collapse
|
31
|
Hong J, Rui W, Fei X, Chen X, Shen K. Association of tumor-infiltrating lymphocytes before and after neoadjuvant chemotherapy with pathological complete response and prognosis in patients with breast cancer. Cancer Med 2021; 10:7921-7933. [PMID: 34562054 PMCID: PMC8607245 DOI: 10.1002/cam4.4302] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/25/2021] [Accepted: 08/27/2021] [Indexed: 12/30/2022] Open
Abstract
Purpose To evaluate the predictive and prognostic value of tumor‐infiltrating lymphocytes (TILs) before and after neoadjuvant chemotherapy (NAC) in patients with breast cancer. Patients and methods Consecutive breast cancer patients treated with NAC between August 2008 and November 2019 were retrospectively analyzed. TIL levels were evaluated of invasive tumor samples, and high expression was defined as TILs >10%. Total pathological complete response (pCR) was defined as no invasive tumor in the breast or lymph nodes. Univariate and multivariate analyses were used to assess factors associated with pCR rate, disease‐free survival (DFS), and overall survival. Results A total of 461 patients were included. The mean pre‐NAC TIL level was higher among patients with pCR than among patients without pCR (24.28% ± 2.34% vs. 11.34% ± 0.60%, respectively, p < 0.0001). The multivariate analysis demonstrated that a high pre‐NAC TIL level was an independent risk factor for a higher pCR (odds ratio = 3.92, 95% CI = 2.23–6.90, p < 0.001). Patients with high pre‐NAC TIL levels had a better 5‐year DFS than those with low pre‐NAC TIL levels (84.5% vs. 68.9%, HR = 0.50, 95% CI = 0.31–0.81, p = 0.005). The multivariate analysis showed that pre‐NAC TIL (HR = 0.48; 95% CI = 0.29–0.81, p = 0.006) but not post‐NAC TIL (HR = 0.89, 95% CI = 0.50–1.59, p = 0.699) was significantly associated with DFS among patients without pCR. Furthermore, patients with low pre‐ and post‐NAC TIL levels had a worse 5‐year DFS than those with high pre‐NAC TIL levels (HR = 2.09, 95% CI = 1.23–3.56, p = 0.007). Conclusions Pre‐NAC TIL level can predict pCR and DFS in patients with breast cancer receiving NAC. For patients without pCR, pre‐NAC TIL, and TIL category change, but not post‐NAC TIL, were significantly associated with DFS.
Collapse
Affiliation(s)
- Jin Hong
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Weiwei Rui
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Steinhof-Radwańska K, Grażyńska A, Lorek A, Gisterek I, Barczyk-Gutowska A, Bobola A, Okas K, Lelek Z, Morawska I, Potoczny J, Niemiec P, Szyluk K. Contrast-Enhanced Spectral Mammography Assessment of Patients Treated with Neoadjuvant Chemotherapy for Breast Cancer. Curr Oncol 2021; 28:3448-3462. [PMID: 34590596 PMCID: PMC8482113 DOI: 10.3390/curroncol28050298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Evaluating the tumor response to neoadjuvant chemotherapy is key to planning further therapy of breast cancer. Our study aimed to evaluate the effectiveness of low-energy and subtraction contrast-enhanced spectral mammography (CESM) images in the detection of complete response (CR) for neoadjuvant chemotherapy (NAC) in breast cancer. Methods: A total of 63 female patients were qualified for our retrospective analysis. Low-energy and subtraction CESM images just before the beginning of NAC and as a follow-up examination 2 weeks before the end of chemotherapy were compared with one another and assessed for compliance with the postoperative histopathological examination (HP). The response to preoperative chemotherapy was evaluated based on the RECIST 1.1 criteria (Response Evaluation Criteria in Solid Tumors). Results: Low-energy images tend to overestimate residual lesions (6.28 mm) and subtraction images tend to underestimate them (2.75 mm). The sensitivity of low-energy images in forecasting CR amounted to 33.33%, while the specificity was 92.86%. In the case of subtraction CESM, the sensitivity amounted to 85.71% and the specificity to 71.42%. Conclusions: CESM is characterized by high sensitivity in the assessment of CR after NAC. The use of only morphological assessment is insufficient. CESM correlates well with the size of residual lesions on histopathological examination but tends to underestimate the dimensions.
Collapse
Affiliation(s)
- Katarzyna Steinhof-Radwańska
- Department of Radiology and Nuclear Medicine, Prof. Kornel Gibiński Independent Public Central Clinical Hospital, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland;
- Correspondence: ; Tel.: +48-32-358-1350
| | - Anna Grażyńska
- Students’ Scientific Society, Department of Radiology and Nuclear Medicine, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland; (A.G.); (K.O.); (Z.L.); (I.M.); (J.P.)
| | - Andrzej Lorek
- Department of Oncological Surgery, Prof. Kornel Gibiński Independent Public Central Clinical Hospital, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland;
| | - Iwona Gisterek
- Department of Oncology and Radiotherapy, Prof. Kornel Gibiński Independent Public Central Clinical Hospital, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland; (I.G.); (A.B.)
| | - Anna Barczyk-Gutowska
- Department of Radiology and Nuclear Medicine, Prof. Kornel Gibiński Independent Public Central Clinical Hospital, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland;
| | - Agnieszka Bobola
- Department of Oncology and Radiotherapy, Prof. Kornel Gibiński Independent Public Central Clinical Hospital, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland; (I.G.); (A.B.)
| | - Karolina Okas
- Students’ Scientific Society, Department of Radiology and Nuclear Medicine, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland; (A.G.); (K.O.); (Z.L.); (I.M.); (J.P.)
| | - Zuzanna Lelek
- Students’ Scientific Society, Department of Radiology and Nuclear Medicine, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland; (A.G.); (K.O.); (Z.L.); (I.M.); (J.P.)
| | - Irmina Morawska
- Students’ Scientific Society, Department of Radiology and Nuclear Medicine, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland; (A.G.); (K.O.); (Z.L.); (I.M.); (J.P.)
| | - Jakub Potoczny
- Students’ Scientific Society, Department of Radiology and Nuclear Medicine, Medical University of Silesia, Medyków 18, 40-514 Katowice, Poland; (A.G.); (K.O.); (Z.L.); (I.M.); (J.P.)
| | - Paweł Niemiec
- Department of Biochemistry and Medical Genetics, School of Health Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland;
| | - Karol Szyluk
- 1st Department of Orthopaedic and Trauma Surgery, District Hospital of Orthopaedics and Trauma Surgery, Bytomska 62, 41-940 Piekary Śląskie, Poland;
| |
Collapse
|
33
|
Murphy J, Gandhi A. Does Mastectomy Reduce Overall Survival in Early Stage Breast Cancer? Clin Oncol (R Coll Radiol) 2021; 33:440-447. [DOI: 10.1016/j.clon.2021.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/22/2020] [Accepted: 03/05/2021] [Indexed: 01/12/2023]
|
34
|
Yan H, Luo B, Wu X, Guan F, Yu X, Zhao L, Ke X, Wu J, Yuan J. Cisplatin Induces Pyroptosis via Activation of MEG3/NLRP3/caspase-1/GSDMD Pathway in Triple-Negative Breast Cancer. Int J Biol Sci 2021; 17:2606-2621. [PMID: 34326697 PMCID: PMC8315016 DOI: 10.7150/ijbs.60292] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Cisplatin (DDP) was reported to improve pathological complete response (pCR) rates in triple-negative breast cancer (TNBC) patients, however, the molecular mechanism still remains largely unknown. Emerging evidence suggested that some chemotherapeutic drugs played anti-tumor effects by inducing cell pyroptosis. Nevertheless, whether pyroptosis contributes to the DDP-induced anti-tumor effect in TNBC remains unexploited. In the present study, NLRP3/caspase-1/GSDMD pyroptosis pathway was involved in the DDP-induced anti-tumor effect of TNBC in vitro and in vivo, providing evidence that DDP might induce pyroptosis in TNBC. Moreover, DDP activated NLRP3/caspase-1/GSDMD pyroptosis pathway by up-regulating the long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3). Furthermore, knockdown of MEG3 not only partly abolished the activation effect of DDP on NLRP3/caspase-1/GSDMD pathway-mediated pyroptosis, but also reversed the suppression of DDP on tumor growth and metastasis ability in vitro and in vivo, further confirming that MEG3 may partially mediate the pyroptotic signaling upon DDP treatment. Thus, our data uncovered a novel mechanism that DDP induced pyroptosis via activation of MEG3/NLRP3/caspase-1/GSDMD pathway in TNBC to exert anti-tumor effects, which may help to develop new strategies for the therapeutic interventions in TNBC.
Collapse
Affiliation(s)
- Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bin Luo
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoyan Wu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Feng Guan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xinxin Yu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lina Zhao
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaokang Ke
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Juan Wu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
35
|
Neoadjuvant chemotherapy: practice and thinking for Chinese patients with early breast cancer. Chin Med J (Engl) 2021; 133:2368-2369. [PMID: 32769502 PMCID: PMC7546848 DOI: 10.1097/cm9.0000000000000940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
36
|
Zheng CH, Liu ZY, Yuan CX, Dong XY, Li HM, Wang JJ, Zhang ZP, Liu HY, Ding XY, Wu W, Wang RR, Wang YS. Mutant Allele Frequency-Based Intra-Tumoral Genetic Heterogeneity Related to the Tumor Shrinkage Mode After Neoadjuvant Chemotherapy in Breast Cancer Patients. Front Med (Lausanne) 2021; 8:651904. [PMID: 33869255 PMCID: PMC8044356 DOI: 10.3389/fmed.2021.651904] [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: 01/11/2021] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
The shrinkage mode of tumor extent after neoadjuvant chemotherapy (NAC) is an important index to evaluate the odds of breast-conserving surgery. However, there is no sufficient measurement to predict the shrinkage mode after NAC. In this study, we analyzed 24 patients' formalin-fixed, paraffin-embedded samples before and after treatment and analyzed 456 cancer-related genes panel by using target next-generation sequencing. Meanwhile, the pathological shrinkage mode was reconstructed in three dimensions after surgery, and the genetic heterogeneity level was estimated by mutant-allele tumor heterogeneity (MATH). We measured the genetic intra-tumor heterogeneity and explored its correlation with the shrinkage mode after NAC. A total of 17 matched pair samples of primary tumor tissue and residual tumor tissue were successfully accessed. It was found that the most common mutated genes were TP53 and PIK3CA in both samples before and after NAC, and no recurrent mutations were significantly associated with the shrinkage mode. Besides, the MATH value of formalin-fixed, paraffin-embedded samples before and after NAC was analyzed by the area under the curve of the receiver operating characteristic, and it is feasible to classify patients into concentric shrinkage mode and non-concentric shrinkage mode in NAC based on the MATH threshold of 58. Our findings indicate that the MATH value was associated with the shrinkage mode of breast cancer in a non-linear model. Patients with the MATH value below the threshold of 58 before and after NAC displayed a concentric shrinkage mode. The area under the curve was 0.89, with a sensitivity of 0.69 and specificity of 1. Our study might provide a promising application of intra-tumor heterogeneity that is measured by MATH to make a choice of surgery.
Collapse
Affiliation(s)
- Chun-Hui Zheng
- Breast Cancer Center, Shandong Cancer Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Departments of Oncology Surgery, People's Hospital, Weifang, China
| | - Zhao-Yun Liu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Chen-Xi Yuan
- Department of Radiation Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xiao-Yun Dong
- Genetics Department in School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Hai-Mei Li
- Department of Molecular Biology in School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Jin-Jin Wang
- Berry Oncology Corporation, Digital Fujian Park, Fuzhou, China
| | - Zhao-Peng Zhang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Hong-Ying Liu
- Genetics Department in School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Xiao-Yan Ding
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang, China
| | - Wendy Wu
- Berry Oncology Corporation, Digital Fujian Park, Fuzhou, China
| | - Rui-Ru Wang
- Berry Oncology Corporation, Digital Fujian Park, Fuzhou, China
| | - Yong-Sheng Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
37
|
Untch M. Local and regional therapy considerations after preoperative therapy in patients with breast cancer. Curr Opin Obstet Gynecol 2021; 33:59-63. [PMID: 33122576 DOI: 10.1097/gco.0000000000000672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The starting point of neoadjuvant therapy was to use preoperative chemotherapy in order to provide surgeons and radiotherapists with the possibility of local treatment in patients with locally advanced, primary inoperable or inflammatory disease. Since then, this treatment approach has dramatically evolved and is now a standard of care in patients with high-risk early breast cancer. RECENT FINDINGS The role of surgery after neoadjuvant therapy is to remove residual disease in the breast and or in the lymph nodes and to provide further treatment possibilities according to pathohistologic findings at surgery. SUMMARY Innovative medical treatments are now being used for neoadjuvant treatment in order to reduce the extent of locoregional surgery in the breast and the axilla and also to adjust further medical treatment after neoadjuvant therapy and surgery.
Collapse
Affiliation(s)
- Michael Untch
- Helios Klinikum Berlin-Buch, Germany, Clinic for Gynecology, Gynecologic Oncology and Obstetrics, Berlin, Germany
| |
Collapse
|
38
|
Song D, Man X, Jin M, Li Q, Wang H, Du Y. A Decision-Making Supporting Prediction Method for Breast Cancer Neoadjuvant Chemotherapy. Front Oncol 2021; 10:592556. [PMID: 33469514 PMCID: PMC7813988 DOI: 10.3389/fonc.2020.592556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/16/2020] [Indexed: 01/02/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) may increase the resection rate of breast cancer and shows promising effects on patient prognosis. It has become a necessary treatment choice and is widely used in the clinical setting. Benefitting from the clinical information obtained during NAC treatment, computational methods can improve decision-making by evaluating and predicting treatment responses using a multidisciplinary approach, as there are no uniformly accepted protocols for all institutions for adopting different treatment regiments. In this study, 166 Chinese breast cancer cases were collected from patients who received NAC treatment at the First Bethune Hospital of Jilin University. The Miller–Payne grading system was used to evaluate the treatment response. Four machine learning multiple classifiers were constructed to predict the treatment response against the 26 features extracted from the patients’ clinical data, including Random Forest (RF) model, Convolution Neural Network (CNN) model, Support Vector Machine (SVM) model, and Logistic Regression (LR) model, where the RF model achieved the best performance using our data. To allow a more general application, the models were reconstructed using only six selected features, and the RF model achieved the highest performance with 54.26% accuracy. This work can efficiently guide optimal treatment planning for breast cancer patients.
Collapse
Affiliation(s)
- Dong Song
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, China
| | - Xiaxia Man
- Department of Oncological Gynecology, The First Hospital, Jilin University, Changchun, China
| | - Meng Jin
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institute of Computational Biology, Northeast Normal University, Changchun, China
| | - Qian Li
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, China
| | - Han Wang
- School of Information Science and Technology, Northeast Normal University, Changchun, China.,Institute of Computational Biology, Northeast Normal University, Changchun, China
| | - Ye Du
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
39
|
Asaoka M, Gandhi S, Ishikawa T, Takabe K. Neoadjuvant Chemotherapy for Breast Cancer: Past, Present, and Future. Breast Cancer (Auckl) 2020; 14:1178223420980377. [PMID: 33402827 PMCID: PMC7747102 DOI: 10.1177/1178223420980377] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) had been developed as a systematic approach before definitive surgery for the treatment of locally advanced or inoperable breast cancer such as inflammatory breast cancer in the past. In addition to its impact on surgery, the neoadjuvant setting has a benefit of providing the opportunity to monitor the individual drug response. Currently, the subject of NAC has expanded to include patients with early-stage, operable breast cancer because it is revealed that the achievement of a pathologic complete response (pCR) is associated with excellent long-term outcomes, especially in patients with aggressive phenotype breast cancer. In addition, this approach provides the unique opportunity to escalate adjuvant therapy in those with residual disease after NAC. Neoadjuvant chemotherapy in breast cancer is a rapidly evolving topic with tremendous interest in ongoing clinical trials. Here, we review the improvements and further challenges in the NAC setting in translational breast cancer research.
Collapse
Affiliation(s)
- Mariko Asaoka
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Shipra Gandhi
- Breast Medicine, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
40
|
Dasgupta A, Brade S, Sannachi L, Quiaoit K, Fatima K, DiCenzo D, Osapoetra LO, Saifuddin M, Trudeau M, Gandhi S, Eisen A, Wright F, Look-Hong N, Sadeghi-Naini A, Tran WT, Curpen B, Czarnota GJ. Quantitative ultrasound radiomics using texture derivatives in prediction of treatment response to neo-adjuvant chemotherapy for locally advanced breast cancer. Oncotarget 2020; 11:3782-3792. [PMID: 33144919 PMCID: PMC7584238 DOI: 10.18632/oncotarget.27742] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
Background: To investigate quantitative ultrasound (QUS) based higher-order texture derivatives in predicting the response to neoadjuvant chemotherapy (NAC) in patients with locally advanced breast cancer (LABC). Materials and Methods: 100 Patients with LABC were scanned before starting NAC. Five QUS parametric image-types were generated from radio-frequency data over the tumor volume. From each QUS parametric-image, 4 grey level co-occurrence matrix-based texture images were derived (20 QUS-Tex1), which were further processed to create texture derivatives (80 QUS-Tex1-Tex2). Patients were classified into responders and non-responders based on clinical/pathological responses to treatment. Three machine learning algorithms based on linear discriminant (FLD), k-nearest-neighbors (KNN), and support vector machine (SVM) were used for developing radiomic models of response prediction. Results: A KNN-model provided the best results with sensitivity, specificity, accuracy, and area under curve (AUC) of 87%, 81%, 82%, and 0.86, respectively. The most helpful features in separating the two response groups were QUS-Tex1-Tex2 features. The 5-year recurrence-free survival (RFS) calculated for KNN predicted responders and non-responders using QUS-Tex1-Tex2 model were comparable to RFS for the actual response groups. Conclusions: We report the first study demonstrating QUS texture-derivative methods in predicting NAC responses in LABC, which leads to better results compared to using texture features alone.
Collapse
Affiliation(s)
- Archya Dasgupta
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Stephen Brade
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Lakshmanan Sannachi
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Karina Quiaoit
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Kashuf Fatima
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Daniel DiCenzo
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Laurentius O Osapoetra
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Murtuza Saifuddin
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Maureen Trudeau
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Sonal Gandhi
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Andrea Eisen
- Department of Medical Oncology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Frances Wright
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada
| | - Nicole Look-Hong
- Department of Surgical Oncology, Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada
| | - Ali Sadeghi-Naini
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Department of Electrical Engineering and Computer Sciences, Lassonde School of Engineering, York University, Toronto, Canada
| | - William T Tran
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Evaluative Clinical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Belinda Curpen
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Gregory J Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Department of Electrical Engineering and Computer Sciences, Lassonde School of Engineering, York University, Toronto, Canada
| |
Collapse
|
41
|
Hong J, Wu J, Huang O, He J, Zhu L, Chen W, Li Y, Chen X, Shen K. Early response and pathological complete remission in Breast Cancer with different molecular subtypes: a retrospective single center analysis. J Cancer 2020; 11:6916-6924. [PMID: 33123282 PMCID: PMC7591996 DOI: 10.7150/jca.46805] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/06/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose: To evaluate the association of clinical early response and pathological complete remission (pCR) in breast cancer patients with different molecular subtypes. Materials and methods: Breast cancer patients who received neoadjuvant treatment (NAT) with clinical early response assessment from October 2008 to October 2018 were retrospectively analyzed. Clinical early response was defined as tumor size decreasing ≥30% evaluated by ultrasound after two cycles of NAT. Chi-square test was used to compare the pCR rates between the responder and non-responder groups with different molecular subtypes. Multivariate logistic regression was used to identify independent factors associated with the pCR. Results: A total of 328 patients were included: 100 responders and 228 non-responders. The progesterone receptor (PR) expression was an independent factor associated with clinical early response (OR=2.39, 95%CI=1.41-4.05, P=0.001). The pCR rate of breast was 50.0% for responders and 18.0% for non-responders (P<0.001). Regarding different molecular subtypes, responders had higher pCR rates than non-responders for patients with HER2 overexpression (OR=10.66, 95%CI=2.18-52.15, P=0.001), triple negative (OR=3.29, 95%CI=1.23-8.84, P=0.016) and Luminal (HER2-) subtypes (OR=8.58, 95%CI=3.05-24.10, P<0.001) respectively. Moreover, pCR rate can be achieved as high as 88.2% in HER2 overexpression patients with early clinical response, which was significantly higher than patients without early response (41.3%, P=0.001). Multivariate analysis showed that clinical early response was an independent factor associated with the pCR rate (OR=4.87, 95%CI=2.72-8.72, P<0.001). Conclusions: Early response was significantly associated with a higher pCR rate in breast cancer patients receiving NAT, especially for patients with HER2 overexpression subtype, which warrants further clinical evaluation.
Collapse
Affiliation(s)
- Jin Hong
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Jiayi Wu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Ou Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Jianrong He
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Li Zhu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Weiguo Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Yafen Li
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| |
Collapse
|
42
|
Valmadrid AC, Kaoutzanis C, Wormer BA, Farinas AF, Wang L, Al Kassis S, Perdikis G, Braun SA, Higdon KK. Comparison of Telfa Rolling and a Closed Washing System for Autologous Fat Processing Techniques in Postmastectomy Breast Reconstruction. Plast Reconstr Surg 2020; 146:486-497. [PMID: 32842097 DOI: 10.1097/prs.0000000000007053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The purpose of this study was to compare the commonly used fat grafting techniques-Telfa rolling and a closed washing system-in breast reconstruction patients. METHODS Consecutive patients undergoing fat grafting were retrospectively reviewed and grouped by technique. Patients with less than 180 days of follow-up were excluded. Demographics, operative details, and complications were compared using univariate analysis with significance set at p < 0.05. RESULTS Between January of 2013 and September of 2017, 186 women underwent a total of 319 fat grafting procedures. There was no difference in demographics, number of procedures performed, volume of fat grafted, and number of days after reconstruction that fat grafting was performed between groups (p > 0.05). Telfa rolling patients had longer operative times for second fat grafting procedures (implant exchange often completed prior) [100.0 minutes (range, 60.0 to 150.0 minutes) versus 79.0 minutes (range, 64.0 to 94.0 minutes); p = 0.03]. Telfa rolling breasts had more palpable masses requiring imaging (26.0 percent versus 14.4 percent; p = 0.01) and an increased incidence of fat necrosis (20.6 percent versus 8.0 percent; p < 0.01). The closed washing system was found to be an independent predictor of decreased rates of imaging-confirmed fat necrosis (OR, 0.29; p = 0.048). There was no difference in fat necrosis excision or cancer recurrence between the groups. CONCLUSION The closed washing system was independently associated with decreased rates of imaging-confirmed fat necrosis compared to Telfa rolling without an increase in other complications. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, III.
Collapse
Affiliation(s)
- Al C Valmadrid
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Christodoulos Kaoutzanis
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Blair A Wormer
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Angel F Farinas
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Li Wang
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Salam Al Kassis
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Galen Perdikis
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Stephane A Braun
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| | - Kent K Higdon
- From the Department of Plastic Surgery, Vanderbilt University Medical Center; and Department of Biostatistics, Vanderbilt University School of Medicine
| |
Collapse
|
43
|
Dediu M, Zielinski C. Reply to the Correspondence by Untch et al. "Concerning Dediu M, Zielinski C: A Proposal to Redefine Pathologic Complete Remission as Endpoint following Neoadjuvant Chemotherapy in Early Breast Cancer" [Breast Care 2019; DOI 10.1159/000500624]. Breast Care (Basel) 2020; 15:314-316. [PMID: 32774227 DOI: 10.1159/000502508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/05/2019] [Indexed: 11/19/2022] Open
Affiliation(s)
| | - Christoph Zielinski
- Vienna Cancer Center, Vienna Hospital Association and Medical University Vienna, Vienna, Austria
| |
Collapse
|
44
|
Jabagi MJ, Vey N, Goncalves A, Le Tri T, Zureik M, Dray-Spira R. Risk of secondary hematologic malignancies associated with breast cancer chemotherapy and G-CSF support: A nationwide population-based cohort. Int J Cancer 2020; 148:375-384. [PMID: 32683691 DOI: 10.1002/ijc.33216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022]
Abstract
Our study aimed to analyze the risk of hematologic malignancies (HM) associated with the use of G-CSF with chemotherapy for BC. Using the French National Health Data System, we examined the HM risks in patients diagnosed with an incident breast cancer between 2007 and 2015, who received chemotherapy for BC. Main outcomes were acute myeloid leukemia (AML), Myelodysplastic syndrome (MDS), myeloproliferative neoplasms (MPNs), multiple myeloma (MM), Hodgkin lymphoma or non-Hodgkin lymphoma (HL/NHL) and acute lymphoblastic leukemia or lymphocytic lymphoma (ALL/LL). Among a total of 122 373 BC survivors, 38.9% received chemotherapy only and 61.1% received chemotherapy + G-CSF. Overall, 781 cases of hematologic malignancies occurred. We observed a nonsignificant increase in the risk of AML (aHR, 1.3; 95% CI, 1.0-1.7), of MDS (aHR, 1.3; 95% CI, 0.9-1.8) and of ALL/LL (aHR, 2.0; 95% CI, 1.0-4.4) among patients treated by chemotherapy + G-CSF compared to chemotherapy only. In analyses by dose, we observed a slight increase in the risk of AML (1-3 doses: aHR, 1.2; 95% CI, 0.8-1.7/4+ doses: aHR, 1.3; 95% CI, 1.0-1.8) and of MDS (1-3 doses: aHR, 1.1; 95% CI, 0.7-1.7/4+ doses: aHR, 1.4; 95% CI, 1.0-1.9), a significant increase in risk of ALL (1-3 doses: aHR, 1.5; 95% CI, 0.5-3.9 / 4+ doses: aHR, 2.3; 95% CI, 1.0-5.1) with increasing cycles of G-CSF. Our population-based study showed that the ALL/LL was the only HM at increased risk with the use of growth factors with a possible dose-effect relationship. Our data regarding the risk of all the other HM are reassuring.
Collapse
Affiliation(s)
- Marie Joelle Jabagi
- EPI-PHARE (French National Agency for Medicines and Health Products Safety (ANSM) and French National Health Insurance (CNAM)), Saint-Denis, France
| | - Norbert Vey
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France
| | - Anthony Goncalves
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France
| | - Thien Le Tri
- EPI-PHARE (French National Agency for Medicines and Health Products Safety (ANSM) and French National Health Insurance (CNAM)), Saint-Denis, France
| | - Mahmoud Zureik
- EPI-PHARE (French National Agency for Medicines and Health Products Safety (ANSM) and French National Health Insurance (CNAM)), Saint-Denis, France
| | - Rosemary Dray-Spira
- EPI-PHARE (French National Agency for Medicines and Health Products Safety (ANSM) and French National Health Insurance (CNAM)), Saint-Denis, France
| |
Collapse
|
45
|
Woitek R, McLean MA, Gill AB, Grist JT, Provenzano E, Patterson AJ, Ursprung S, Torheim T, Zaccagna F, Locke M, Laurent MC, Hilborne S, Frary A, Beer L, Rundo L, Patterson I, Slough R, Kane J, Biggs H, Harrison E, Lanz T, Basu B, Baird R, Sala E, Graves MJ, Gilbert FJ, Abraham JE, Caldas C, Brindle KM, Gallagher FA. Hyperpolarized 13C MRI of Tumor Metabolism Demonstrates Early Metabolic Response to Neoadjuvant Chemotherapy in Breast Cancer. Radiol Imaging Cancer 2020; 2:e200017. [PMID: 32803167 PMCID: PMC7398116 DOI: 10.1148/rycan.2020200017] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/24/2020] [Accepted: 04/15/2020] [Indexed: 04/12/2023]
Abstract
Purpose To compare hyperpolarized carbon 13 (13C) MRI with dynamic contrast material-enhanced (DCE) MRI in the detection of early treatment response in breast cancer. Materials and Methods In this institutional review board-approved prospective study, a woman with triple-negative breast cancer (age, 49 years) underwent 13C MRI after injection of hyperpolarized [1-carbon 13 {13C}]-pyruvate and DCE MRI at 3 T at baseline and after one cycle of neoadjuvant therapy. The 13C-labeled lactate-to-pyruvate ratio derived from hyperpolarized 13C MRI and the pharmacokinetic parameters transfer constant (K trans) and washout parameter (k ep) derived from DCE MRI were compared before and after treatment. Results Exchange of the 13C label between injected hyperpolarized [1-13C]-pyruvate and the endogenous lactate pool was observed, catalyzed by the enzyme lactate dehydrogenase. After one cycle of neoadjuvant chemotherapy, a 34% reduction in the 13C-labeled lactate-to-pyruvate ratio resulted in correct identification of the patient as a responder to therapy, which was subsequently confirmed via a complete pathologic response. However, DCE MRI showed an increase in mean K trans (132%) and mean k ep (31%), which could be incorrectly interpreted as a poor response to treatment. Conclusion Hyperpolarized 13C MRI enabled successful identification of breast cancer response after one cycle of neoadjuvant chemotherapy and may improve response prediction when used in conjunction with multiparametric proton MRI.Published under a CC BY 4.0 license.
Collapse
Affiliation(s)
- Ramona Woitek
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Mary A. McLean
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Andrew B. Gill
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - James T. Grist
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Elena Provenzano
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Andrew J. Patterson
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Stephan Ursprung
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Turid Torheim
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Fulvio Zaccagna
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Matthew Locke
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Marie-Christine Laurent
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Sarah Hilborne
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Amy Frary
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Lucian Beer
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Leonardo Rundo
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Ilse Patterson
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Rhys Slough
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Justine Kane
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Heather Biggs
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Emma Harrison
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Titus Lanz
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Bristi Basu
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Richard Baird
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Evis Sala
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Martin J. Graves
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Fiona J. Gilbert
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Jean E. Abraham
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Carlos Caldas
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Kevin M. Brindle
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| | - Ferdia A. Gallagher
- From the Departments of Radiology (R.W., A.B.G., J.T.G., A.J.P., S.U., F.Z., M.L., M.C.L., S.H., A.F., L.B., L.R., E.S., M.J.G., F.J.G., F.A.G.), Oncology (J.K., H.B., E.H., B.B., R.B., J.E.A., C.C.), and Biochemistry (K.M.B.), the Cambridge Breast Cancer Research Unit (E.P., J.K., H.B., E.H., R.B., J.E.A., C.C.), University of Cambridge, Cambridge, England; Departments of Radiology (A.J.P., I.P., R.S., M.J.G., F.J.G., F.A.G.) and Histopathology (E.P.), Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, England; Cancer Research UK Cambridge Centre, Cambridge, England (R.W., M.A.M., E.P., T.T., L.B., L.R., E.S., J.E.A., C.C., K.M.B., F.A.G.); Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, Vienna 1090, Austria (R.W., L.B.); Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, England (M.A.M., T.T., C.C., K.M.B.); and RAPID Biomedical, Rimpar, Germany (T.L.)
| |
Collapse
|
46
|
Zhao Y, Schaafsma E, Cheng C. Gene signature-based prediction of triple-negative breast cancer patient response to Neoadjuvant chemotherapy. Cancer Med 2020; 9:6281-6295. [PMID: 32692484 PMCID: PMC7476842 DOI: 10.1002/cam4.3284] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/24/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
Neoadjuvant chemotherapy is the current standard of care for large, advanced, and/or inoperable tumors, including triple‐negative breast cancer. Although the clinical benefits of neoadjuvant chemotherapy have been illustrated through numerous clinical trials, more than half of the patients do not experience therapeutic benefit and needlessly suffer from side effects. Currently, no clinically applicable biomarkers are available for predicting neoadjuvant chemotherapy response in triple‐negative breast cancer; the discovery of such a predictive biomarker or marker profile is an unmet need. In this study, we introduce a generic computational framework to calculate a response‐probability score (RPS), based on patient transcriptomic profiles, to predict their response to neoadjuvant chemotherapy. We first validated this framework in ER‐positive breast cancer patients and showed that it predicted neoadjuvant chemotherapy response with equal performance to several clinically used gene signatures, including Oncotype DX and MammaPrint. Then, we applied this framework to triple‐negative breast cancer data and, for each patient, we calculated a response probability score (TNBC‐RPS). Our results indicate that the TNBC‐RPS achieved the highest accuracy for predicting neoadjuvant chemotherapy response compared to previously proposed 143 gene signatures. When combined with additional clinical factors, the TNBC‐RPS achieved a high prediction accuracy for triple‐negative breast cancer patients, which was comparable to the prediction accuracy of Oncotype DX and MammaPrint in ER‐positive patients. In conclusion, the TNBC‐RPS accurately predicts neoadjuvant chemotherapy response in triple‐negative breast cancer patients and has the potential to be clinically used to aid physicians in stratifying patients for more effective neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Yanding Zhao
- Department of Molecular and Systems Biology, The Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA.,Department of Biomedical Data Science, The Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA
| | - Evelien Schaafsma
- Department of Molecular and Systems Biology, The Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA.,Department of Biomedical Data Science, The Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA
| | - Chao Cheng
- Department of Molecular and Systems Biology, The Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA.,Department of Biomedical Data Science, The Geisel School of Medicine at Dartmouth College, Lebanon, NH, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
47
|
Wang H, Mao X. Evaluation of the Efficacy of Neoadjuvant Chemotherapy for Breast Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2423-2433. [PMID: 32606609 PMCID: PMC7308147 DOI: 10.2147/dddt.s253961] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
Neoadjuvant chemotherapy is increasingly used in breast cancer, especially for downstaging the primary tumor in the breast and the metastatic axillary lymph node. Accurate evaluations of the response to neoadjuvant chemotherapy provide important information on the impact of systemic therapies on breast cancer biology, prognosis, and guidance for further therapy. Moreover, pathologic complete response is a validated and valuable surrogate prognostic factor of survival after therapy. Evaluations of neoadjuvant chemotherapy response are very important in clinical work and basic research. In this review, we will elaborate on evaluations of the efficacy of neoadjuvant chemotherapy in breast cancer and provide a clinical evaluation procedure for neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Huan Wang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| |
Collapse
|
48
|
Mazalovska M, Kouokam JC. Transiently Expressed Mistletoe Lectin II in Nicotiana benthamiana Demonstrates Anticancer Activity In Vitro. Molecules 2020; 25:E2562. [PMID: 32486427 PMCID: PMC7321061 DOI: 10.3390/molecules25112562] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/24/2022] Open
Abstract
Mistletoe (Viscum album) extracts have been used as alternative and complementary therapeutic preparations in multiple cancers for decades. Mistletoe lectins (ML-I, ML-II, and ML-III) are considered to be the main anticancer components of such preparations. In the present study, ML-II was transiently expressed in Nicotiana benthamiana using the pEAQ-HT expression system. Expression levels of up to 60 mg/kg of the infiltrated plant tissue were obtained, and a three-fold increase was achieved by adding the endoplasmic reticulum (ER) retention signal KDEL to the native ML-II sequence. The native protein containing His-tag and KDEL was purified by immobilized metal affinity chromatography (IMAC) and gel filtration. We found that the recombinant ML-II lectin was glycosylated and retained its carbohydrate-binding activity. In addition, we demonstrated that plant produced ML-II displayed anticancer activity in vitro, inhibiting non-small cell lung cancer H460 and A549 cells with EC50 values of 4 and 3.5 µg/mL, respectively. Annexin V-448A and PI double staining revealed that cell cytotoxicity occurred via apoptosis induction. These results indicate that ML-II transiently expressed in N. benthamiana plants is a promising candidate as an anticancer agent, although further optimization of production and purification methods is required to enable further in vitro testing, as well as in vivo assays.
Collapse
Affiliation(s)
- Milena Mazalovska
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - J. Calvin Kouokam
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
49
|
Plant-Derived Lectins as Potential Cancer Therapeutics and Diagnostic Tools. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1631394. [PMID: 32509848 PMCID: PMC7245692 DOI: 10.1155/2020/1631394] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/27/2020] [Indexed: 12/19/2022]
Abstract
Cancer remains a global health challenge, with high morbidity and mortality, despite the recent advances in diagnosis and treatment. Multiple compounds assessed as novel potential anticancer drugs derive from natural sources, including microorganisms, plants, and animals. Lectins, a group of highly diverse proteins of nonimmune origin with carbohydrate-binding abilities, have been detected in virtually all kingdoms of life. These proteins can interact with free and/or cell surface oligosaccharides and might differentially bind cancer cells, since malignant transformation is tightly associated with altered cell surface glycans. Therefore, lectins could represent a valuable tool for cancer diagnosis and be developed as anticancer therapeutics. Indeed, several plant lectins exert cytotoxic effects mainly by inducing apoptotic and autophagic pathways in malignant cells. This review summarizes the current knowledge regarding the basis for the use of lectins in cancer diagnosis and therapy, providing a few examples of plant-derived carbohydrate-binding proteins with demonstrated antitumor effects.
Collapse
|
50
|
O'Toole SA, Spillane C, Huang Y, Fitzgerald MC, Ffrench B, Mohamed B, Ward M, Gallagher M, Kelly T, O'Brien C, Ruttle C, Bogdanska A, Martin C, Mullen D, Connolly E, McGarrigle SA, Kennedy J, O'Leary JJ. Circulating tumour cell enumeration does not correlate with Miller-Payne grade in a cohort of breast cancer patients undergoing neoadjuvant chemotherapy. Breast Cancer Res Treat 2020; 181:571-580. [PMID: 32378053 PMCID: PMC7220879 DOI: 10.1007/s10549-020-05658-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/27/2020] [Indexed: 12/16/2022]
Abstract
Purpose The association between pathological complete response (pCR) in patients receiving neoadjuvant chemotherapy (NAC) for breast cancer and Circulating Tumour Cells (CTCs) is not clear. The aim of this study was to assess whether CTC enumeration could be used to predict pathological response to NAC in breast cancer as measured by the Miller–Payne grading system. Methods Twenty-six patients were recruited, and blood samples were taken pre- and post-NAC. CTCs were isolated using the ScreenCell device and stained using a modified Giemsa stain. CTCs were enumerated by 2 pathologists and classified as single CTCs, doublets, clusters/microemboli and correlated with the pathological response as measured by the Miller–Payne grading system. χ2 or ANOVA was performed in SPSS 24.0 statistics software for associations. Results 89% of patients had invasive ductal carcinoma (IDC) and 11% invasive lobular carcinoma (ILC). At baseline 85% of patients had CTCs present, median 7 (0–161) CTCs per 3 ml of whole blood. Post-chemotherapy, 58% had an increase in CTCs. This did not correlate with the Miller–Payne grade of response. No significant association was identified between the number of CTCs and clinical characteristics; however, we did observe a correlation between pre-treatment CTC counts and body mass index, p < 0.05. Conclusions Patients with a complete response to NAC still had CTCs present, suggesting enumeration is not sufficient to aid surgery stratification. Additional characterisation and larger studies are needed to further characterise CTCs isolated pre- and post-chemotherapy. Long-term follow-up of these patients will determine the significance of CTCs in NAC breast cancer patients. Electronic supplementary material The online version of this article (10.1007/s10549-020-05658-7) contains supplementary material, which is available to authorized users.
Collapse
MESH Headings
- Adult
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/drug therapy
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Cohort Studies
- Female
- Follow-Up Studies
- Humans
- Middle Aged
- Neoadjuvant Therapy/mortality
- Neoplasm Grading
- Neoplastic Cells, Circulating/drug effects
- Neoplastic Cells, Circulating/pathology
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Survival Rate
Collapse
Affiliation(s)
- Sharon A O'Toole
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland.
- Department of Obstetrics and Gynaecology, Trinity College, Dublin, Ireland.
- Trinity St James's Cancer Institute, Dublin 8, Ireland.
| | - Cathy Spillane
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Yanmei Huang
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Department of Obstetrics and Gynaecology, Trinity College, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Marie C Fitzgerald
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Department of Obstetrics and Gynaecology, Trinity College, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Brendan Ffrench
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
| | - Bashir Mohamed
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Mark Ward
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Michael Gallagher
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Tanya Kelly
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Cathal O'Brien
- Cancer Molecular Diagnostics, St. James's Hospital, Dublin 8, Ireland
| | - Carmel Ruttle
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
| | - Anna Bogdanska
- Department of Obstetrics and Gynaecology, Trinity College, Dublin, Ireland
| | - Cara Martin
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Dorinda Mullen
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| | - Elizabeth Connolly
- Trinity St James's Cancer Institute, Dublin 8, Ireland
- Department of Surgery, St James's Hospital, Dublin 8, Ireland
| | - Sarah A McGarrigle
- Trinity St James's Cancer Institute, Dublin 8, Ireland
- Department of Surgery, St James's Hospital, Dublin 8, Ireland
| | - John Kennedy
- Trinity St James's Cancer Institute, Dublin 8, Ireland
- HOPE Directorate, St. James's Hospital, Dublin 8, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin and Emer Casey Molecular Pathology Research Laboratory, Coombe Women's and Infants University Hospital, Dublin, Ireland
- Trinity St James's Cancer Institute, Dublin 8, Ireland
| |
Collapse
|