51
|
Wang Y, Zou J, Cao Q, Dai G, Fan P, Gong X, Jiang J, Kong Y, Liu C, Liu C, Lu C, Li M, Lang Z, Lin Y, Peng Y, Shi H, Wang Y, Wang J, Xie B, Yang B, Yu G, Zhang C, Zhang H, Zhou L, Zhang Z, Zhu Z, Hao J. Multi-institutional evaluation comparing guidance from International Ki67 Working Group vs National Health Commission of China on immunohistochemistry-based Ki67 assessment alongside the Quantitative Dot Blot method. Front Oncol 2025; 14:1510273. [PMID: 39931209 PMCID: PMC11808281 DOI: 10.3389/fonc.2024.1510273] [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: 10/12/2024] [Accepted: 12/23/2024] [Indexed: 02/13/2025] Open
Abstract
Purpose Recommendations from the National Health Commission of China (NHCC) and the International Ki67 Working Group (IKWG) were issued to guide immunohistochemistry (IHC)-based Ki67 scoring for breast cancer patients in daily clinical practice. They were evaluated in this multi-institutional study alongside the results from the Quantitative Dot Blot (QDB) method. Methods Three alternative adjacent sections from 40 primary ER+ breast cancer resection blocks were randomly assigned a number from 1 to 120 for Ki67 staining and reviewed by 21 pathologists, while the other three alternative sections were sent for QDB analysis of Ki67 protein levels. Ki67 scores were grouped by 5/30% (IKWG), 10/30% (NHCC) and 20/30% (NHCC appendix 9, NHCCa9), respectively while QDB results were grouped by C5-C95 of 2.31 nmol/g defined in previous study as low-, equivocal-, and high-risk groups. Results The overall Intraclass Correlation Coefficient (ICC) was 0.785 for IHC evaluations from 21 pathologists, with Fleiss Kappa values of 0.555, 0.628, and 0.480 when Ki67 scores were grouped by guidance from IKWG, NHCC, and NHCCa9, respectively. In comparison, the ICC and Fleiss kappa values for the QDB analysis were 0.939 and 0.831, respectively. When IHC and QDB results were cross-referenced, more specimens were grouped as high-risk by QDB than IHC, and NHCCa9 led to the highest percentage of disagreement between the two methods. Conclusion The IKWG recommendation was harder to achieve categorized agreement among pathologists than the NHCC recommendation, yet it led to the best agreement with the QDB to define the low-risk group. The QDB method offers significantly improved consistency compared to the current IHC-based Ki67 assessment.
Collapse
Affiliation(s)
- Yin Wang
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Jiarui Zou
- Department of Pathology, Hangzhou Red Cross Hospital, Hangzhou, China
| | - Qinghua Cao
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Guihong Dai
- Department of Pathology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Panhong Fan
- Department of Pathology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xue Gong
- Department of Pathology, Qinghai University Affiliated Hospital, Xining, China
| | - Jinyan Jiang
- Department of Pathology, Chenzhou No. 1 People Hospital, Chenzhou, China
| | - Yanqing Kong
- Department of Pathology, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Chao Liu
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chunhui Liu
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Chenjia Lu
- Department of Pathology, The People’s Hospital of LongHua, Shenzhen, China
| | - Meiren Li
- Department of Pathology, Jiujiang University Affiliated Hospital, Jiujiang, China
| | - Zhiqiang Lang
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yang Lin
- Department of Health statistics, School of Public Health, Binzhou Medical University, Yantai, China
| | - Yan Peng
- Department of Pathology, The Third Affiliated Hospital of Soochow University/Changzhou First People’s Hospital, Changzhou, China
| | - Haiyan Shi
- Department of Pathology, Guangdong Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Yuhuan Wang
- Department of Pathology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urymqi, China
| | - Jiu Wang
- Department of Health statistics, School of Public Health, Binzhou Medical University, Yantai, China
| | - Bichen Xie
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Bing Yang
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Guohua Yu
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Cuiping Zhang
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Hengming Zhang
- Department of Pathology, Weifang People’s Hospital, Weifang, China
| | - Luting Zhou
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zilan Zhang
- Department of Pathology, Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Zhenli Zhu
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Junmei Hao
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| |
Collapse
|
52
|
Macarrón V, Losantos-García I, Peláez-García A, Yébenes L, Berjón A, Frías L, Martí C, Zamora P, Sánchez-Méndez JI, Hardisson D. A Novel Nomogram for Estimating a High-Risk Result in the EndoPredict ® Test for Estrogen Receptor-Positive/Human Epidermal Growth Factor Receptor 2 (HER2)-Negative Breast Carcinoma. Cancers (Basel) 2025; 17:273. [PMID: 39858055 PMCID: PMC11763868 DOI: 10.3390/cancers17020273] [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: 12/04/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The EndoPredict® assay has been widely used in recent years to estimate the risk of distant recurrence and the absolute chemotherapy benefit for patients with estrogen (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer. However, there are no well-defined criteria for selecting patients who may benefit from the test. The aim of this study was to develop a novel nomogram to estimate the probability of obtaining a high-risk EndoPredict® result in clinical practice. Methods: The study cohort comprised 348 cases of T1-3/N0-1a/M0 ER-positive/HER2-negative breast carcinoma. A multivariate analysis was conducted using a training cohort (n = 270) based on clinicopathological features that demonstrated a statistically significant correlation with the EndoPredict® result in a univariate analysis. The predictive model was subsequently represented as a nomogram to estimate the probability of obtaining a high-risk result in the EndoPredict® assay. The predictive model was then validated using a separate validation cohort (n = 78). Results: The clinicopathological features incorporated into the nomogram included tumor size, tumor grade, sentinel lymph node status, pN stage, and Ki67. The internal validation of the model yielded an area under the curve (AUC) of 0.803 (95% CI = 0.751, 0.855) in the receiver operating characteristic (ROC) curve for the training cohort, with an optimal sensitivity and specificity at a threshold of 0.536. The external validation yielded an AUC of 0.789 (95% CI = 0.689, 0.890) in its ROC curve, with optimal sensitivity and specificity achieved at a threshold of 0.393. Conclusions: This study presents, for the first time, the development of a clinically accessible nomogram designed to estimate the probability of obtaining a high-risk result in the EndoPredict® assay. The use of easily available clinicopathological features allows for the optimization of patient selection for the EndoPredict® assay, ensuring that those who would most benefit from undergoing the test are identified.
Collapse
Affiliation(s)
- Víctor Macarrón
- Department of Pathology, Hospital Universitario La Paz, 28046 Madrid, Spain; (V.M.); (L.Y.); (A.B.)
| | | | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
| | - Laura Yébenes
- Department of Pathology, Hospital Universitario La Paz, 28046 Madrid, Spain; (V.M.); (L.Y.); (A.B.)
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
| | - Alberto Berjón
- Department of Pathology, Hospital Universitario La Paz, 28046 Madrid, Spain; (V.M.); (L.Y.); (A.B.)
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
| | - Laura Frías
- Breast Unit, Department of Gynecology and Obstetrics, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.F.); (C.M.)
| | - Covadonga Martí
- Breast Unit, Department of Gynecology and Obstetrics, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.F.); (C.M.)
| | - Pilar Zamora
- Department of Medical Oncology, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | - José Ignacio Sánchez-Méndez
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
- Breast Unit, Department of Gynecology and Obstetrics, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.F.); (C.M.)
- Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, 28046 Madrid, Spain; (V.M.); (L.Y.); (A.B.)
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital (IdiPAZ), 28046 Madrid, Spain;
- Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Center for Biomedical Research in the Cancer Network (Centro de Investigación Biomédica en Red de Cáncer, CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
53
|
Ding X, Zhang L, Fan M, Li L. Network-based transfer of pan-cancer immunotherapy responses to guide breast cancer prognosis. NPJ Syst Biol Appl 2025; 11:4. [PMID: 39788975 PMCID: PMC11720706 DOI: 10.1038/s41540-024-00486-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025] Open
Abstract
Breast cancer prognosis is complicated by tumor heterogeneity. Traditional methods focus on cancer-specific gene signatures, but cross-cancer strategies that provide deeper insights into tumor homogeneity are rarely used. Immunotherapy, particularly immune checkpoint inhibitors, results from variable responses across cancers, offering valuable prognostic insights. We introduced a network-based transfer (NBT) of pan-cancer immunotherapy responses to enhance breast cancer prognosis using node embedding and heat diffusion algorithms, identifying gene signatures netNE and netHD. Our results showed that netHD and netNE outperformed seven established breast cancer signatures in prognostic metrics, with netHD excelling. All nine gene signatures were grouped into three clusters, with netHD and netNE enriching the immune-related interferon-gamma pathway. Stratifying TCGA patients into two groups based on netHD revealed significant immunological differences and variations in 20 of 50 cancer hallmarks, emphasizing immune-related markers. This approach leverages pan-cancer insights to enhance breast cancer prognosis, facilitating insight transfer and improving tumor homogeneity understanding.Abstract graph of network-based insights translating pan-cancer immunotherapy responses to breast cancer prognosis. This abstract graph illustrates the conceptual framework for transferring immunotherapy response insights from pan-cancer studies to breast cancer prognosis. It highlights the integration of PPI networks to bridge genetic data and clinical phenotypes. The network-based method facilitates the identification of prognostic gene signatures in breast cancer by leveraging immunotherapy response information, providing a novel perspective on tumor homogeneity and its implications for clinical outcomes.
Collapse
Affiliation(s)
- Xiaobao Ding
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou, China
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, China
| | - Lin Zhang
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China.
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, China.
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, China.
| |
Collapse
|
54
|
Taube JM, Sunshine JC, Angelo M, Akturk G, Eminizer M, Engle LL, Ferreira CS, Gnjatic S, Green B, Greenbaum S, Greenwald NF, Hedvat CV, Hollmann TJ, Jiménez-Sánchez D, Korski K, Lako A, Parra ER, Rebelatto MC, Rimm DL, Rodig SJ, Rodriguez-Canales J, Roskes JS, Schalper KA, Schenck E, Steele KE, Surace MJ, Szalay AS, Tetzlaff MT, Wistuba II, Yearley JH, Bifulco CB. Society for Immunotherapy of Cancer: updates and best practices for multiplex immunohistochemistry (IHC) and immunofluorescence (IF) image analysis and data sharing. J Immunother Cancer 2025; 13:e008875. [PMID: 39779210 PMCID: PMC11749220 DOI: 10.1136/jitc-2024-008875] [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: 04/05/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES Multiplex immunohistochemistry and immunofluorescence (mIHC/IF) are emerging technologies that can be used to help define complex immunophenotypes in tissue, quantify immune cell subsets, and assess the spatial arrangement of marker expression. mIHC/IF assays require concerted efforts to optimize and validate the multiplex staining protocols prior to their application on slides. The best practice guidelines for staining and validation of mIHC/IF assays across platforms were previously published by this task force. The current effort represents a complementary manuscript for mIHC/IF analysis focused on the associated image analysis and data management. METHODS The Society for Immunotherapy of Cancer convened a task force of pathologists and laboratory leaders from academic centers as well as experts from pharmaceutical and diagnostic companies to develop best practice guidelines for the quantitative image analysis of mIHC/IF output and data management considerations. RESULTS Best-practice approaches for image acquisition, color deconvolution and spectral unmixing, tissue and cell segmentation, phenotyping, and algorithm verification are reviewed. Additional quality control (QC) measures such as batch-to-batch correction and QC for assembled images are also discussed. Recommendations for sharing raw outputs, processed results, key analysis programs and source code, and representative photomicrographs from mIHC/IF assays are included. Lastly, multi-institutional harmonization efforts are described. CONCLUSIONS mIHC/IF technologies are maturing and are routinely included in research studies and moving towards clinical use. Guidelines for how to perform and standardize image analysis on mIHC/IF-stained slides will likely contribute to more comparable results across laboratories and pave the way for clinical implementation. A checklist encompassing these two-part guidelines for the generation of robust data from quantitative mIHC/IF assays will be provided in a third publication from this task force. While the current effort is mainly focused on best practices for characterizing the tumor microenvironment, these principles are broadly applicable to any mIHC/IF assay and associated image analysis.
Collapse
Affiliation(s)
- Janis M Taube
- Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Joel C Sunshine
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | | | - Margaret Eminizer
- Johns Hopkins University, Baltimore, Maryland, USA
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Logan L Engle
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Cláudia S Ferreira
- Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Benjamin Green
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Shirley Greenbaum
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Noah F Greenwald
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Cyrus V Hedvat
- Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Travis J Hollmann
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Daniel Jiménez-Sánchez
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
| | - Konstanty Korski
- Department of Personalized Healthcare, Data, Analytics and Imaging Group, F Hoffmann-La Roche AG, Basel, Switzerland
| | - Ana Lako
- Dana Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Edwin R Parra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Scott J Rodig
- Dana Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Jeffrey S Roskes
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | - Alexander S Szalay
- Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Bloomberg~Kimmel Institute of Cancer Immunotherapy, Johns Hopkins University SOM, Baltimore, Maryland, USA
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael T Tetzlaff
- Departments of Pathology and Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | |
Collapse
|
55
|
Shanthala S, Amirtham U, Lokesh KN, Jacob L, Babu G. Clinicopathological Evaluation of Patients with Hormone Receptor-Positive HER2-Negative Metastatic Breast Cancer Progressing on Endocrine Treatment: A Real-World Retrospective Study from a Regional Cancer Center. South Asian J Cancer 2025; 14:15-22. [PMID: 40124154 PMCID: PMC11925626 DOI: 10.1055/s-0043-1775806] [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] [Indexed: 03/25/2025] Open
Abstract
Metastatic breast cancer (MBC) is an incurable disease with the primary aim of treatment being the improvement of the patient's quality of life and the delay of disease progression. A substantial proportion of patients with hormone receptor (HR)-positive MBC eventually experience progression despite endocrine treatment. As endocrine resistance remains a significant challenge, we aim to comprehend the intricate relationship between clinicopathological characteristics and the utility of various parameters as predictive markers for hormonal treatment response. This study, conducted at a single center, is ambispective in nature and includes hormone receptor (HR)-positive, human epidermal growth factor 2-negative MBC patients who progressed while on endocrine treatment, selected through purposeful sampling. Nominal data were analyzed in terms of frequency distribution, and continuous variables were represented as median/mean ± standard deviation. Spearman's correlation test and chi-square test were employed to examine variable dependencies. Data comparisons were performed using the independent t-test, one-way analysis of variance, or Mann-Whitney's test. The majority of our study participants ( n = 44, 64.70%) presented with de novo metastasis, while the remainder ( n = 24, 35.29%) were patients who progressed from early-stage breast cancer to metastasis. The overall mean age of our study population at presentation was 47 ± 11 years. Patients with upfront stage 4 tumors presented at an older age, exhibited grade 2 tumors, had a higher frequency of bone-only metastasis, and experienced longer progression-free survival (PFS) compared to patients who progressed from the early stage to metastasis. Multiple visceral involvements had a significant negative impact on PFS in contrast to cases with single visceral or bone-only involvement. No significant associations with PFS were observed for the Ki-67 index, first-line chemotherapy, or endocrine therapy. The extent of metastasis to various organs emerged as the most influential factor in determining PFS. Consequently, we propose the necessity for larger prospective studies aimed at identifying superior or additional biomarkers.
Collapse
Affiliation(s)
- S. Shanthala
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Usha Amirtham
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - K. N. Lokesh
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Linu Jacob
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Govinda Babu
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| |
Collapse
|
56
|
Chen XC, Jiao DC, Qiao JH, Wang CZ, Sun XF, Lu ZD, Li LF, Zhang CJ, Yan M, Wei Y, Chen B, Feng YQ, Deng M, Ma MD, Plichta JK, He YW, Liu ZZ. De-escalated neoadjuvant weekly nab-paclitaxel with trastuzumab and pertuzumab versus docetaxel, carboplatin, trastuzumab, and pertuzumab in patients with HER2-positive early breast cancer (HELEN-006): a multicentre, randomised, phase 3 trial. Lancet Oncol 2025; 26:27-36. [PMID: 39612919 DOI: 10.1016/s1470-2045(24)00581-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND A previous phase 2 trial showed promising outcomes for patients with HER2-positive early-stage breast cancer using neoadjuvant de-escalation chemotherapy with paclitaxel, trastuzumab, and pertuzumab. We aimed to evaluate the efficacy of weekly nab-paclitaxel compared with the standard regimen of docetaxel plus carboplatin, both with trastuzumab and pertuzumab, as neoadjuvant therapies for patients with HER2-positive breast cancer. METHODS HELEN-006 was a multicentre, randomised, phase 3 trial done at six hospitals in China. We enrolled patients aged 18-70 years with untreated, histologically confirmed stage II-III invasive HER2-positive breast cancer and an Eastern Cooperative Oncology Group performance status of 0 or 1. Using an interactive response system, patients were randomly assigned (1:1) under a permuted block randomisation scheme (block size of four), stratified by tumour stage, nodal status, and hormone receptor status. Patients received either intravenous nab-paclitaxel (125 mg/m2 on days 1, 8, and 15) for six 3-week cycles, or intravenous docetaxel (75 mg/m2 on day 1) plus intravenous carboplatin (area under the concentration-time curve 6 mg/mL per min on day 1) for six 3-week cycles. Both groups also received concurrent intravenous trastuzumab, with an initial loading dose of 8 mg/kg and a maintenance dose of 6 mg/kg on day 1, as well as intravenous pertuzumab with a loading dose of 840 mg and a maintenance dose of 420 mg on day 1. This report is the final analysis of the primary endpoint, pathological complete response (ypT0/is ypN0), analysed in all patients who started treatment (modified intention to treat). The trial is registered with ClinicalTrials.gov, NCT04547907, and follow-up of the adjuvant phase is ongoing. FINDINGS Between Sept 20, 2020, and March 1, 2023, 789 patients were screened for eligibility, 689 of whom were randomly assigned (343 to the nab-paclitaxel group and 346 to the docetaxel plus carboplatin group). All 689 patients were Asian women. 669 patients received at least one dose of the study treatment and were included in the full analysis set (332 in the nab-paclitaxel group and 337 in the docetaxel plus carboplatin group). Median age of the patients was 50 years (IQR 43-55). Median follow-up time was 26 months (IQR 19-32). 220 (66·3% [95% CI 61·2-71·4]) patients in the nab-paclitaxel group had a pathological complete response, compared with 194 (57·6% [52·3-62·9]) in the docetaxel plus carboplatin group (combined odds ratio 1·54 [95% CI 1·10-2·14]; stratified p=0·011). 100 (30%) patients in the nab-paclitaxel group and 128 (38%) in the docetaxel plus carboplatin group had grade 3-4 adverse events. The most common grade 3-4 adverse events were nausea (22 [7%] in the nab-paclitaxel group vs 76 [23%] in the docetaxel plus carboplatin group), diarrhoea (25 [8%] vs 55 [16%]), and neuropathy (43 [13%] vs eight [2%]). Serious drug-related adverse events were reported in three (1%) patients in the nab-paclitaxel group and five (2%) in the docetaxel plus carboplatin group. No treatment-related deaths were reported in either group. INTERPRETATION These findings might suggest a potential advantage of nab-paclitaxel combined with trastuzumab and pertuzumab compared with the standard regimen in neoadjuvant therapy for patients with HER2-positive early breast cancer, suggesting that this new combination might establish a new standard for neoadjuvant treatment in this patient population. FUNDING National Natural Science Foundation of China, and Science and Technology Research Projects of Henan Province, China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Xiu-Chun Chen
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - De-Chuang Jiao
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiang-Hua Qiao
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Cheng-Zheng Wang
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xian-Fu Sun
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Zhen-Duo Lu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Lian-Fang Li
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Chong-Jian Zhang
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Min Yan
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Ya Wei
- Department of Breast Surgery, Anyang Tumor Hospital, Anyang, China
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yue-Qing Feng
- Department of Breast Surgery, Xinxiang Central Hospital, Xinxiang, China
| | - Miao Deng
- Department of Breast Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Ming-De Ma
- Department of Thyroid and Breast Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jennifer K Plichta
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - You-Wen He
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Zhen-Zhen Liu
- Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
57
|
Hassing CMS, Tvedskov THF, Kroman N, Knoop AS, Lænkholm AV. Evaluating the Prognostic Role of the PAM50 Signature and Selected Immune-Related Signatures for Recurrence in Patients With T1abN0 Breast Cancer. Clin Breast Cancer 2025; 25:e71-e78.e2. [PMID: 39209597 DOI: 10.1016/j.clbc.2024.08.003] [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: 03/05/2024] [Revised: 07/15/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND De-escalation of adjuvant treatment in patients with T1abN0 breast cancer is discussed internationally. Identification of new prognostic factors in these patients may assist this de-escalation. The PAM50 signature and tumor inflammation signature (TIS), Programmed Cell Death Protein 1 (PD-1) and Programmed Cell Death Ligand 1 (PD-L1) signatures are possible prognostic factors for recurrence. MATERIALS AND METHODS Danish patients with T1abN0 breast cancer diagnosed between 2007-2016 were identified, the NanoString Breast Cancer 360 Panel was performed on tissue samples from cases with recurrence matched 1:1 with controls without recurrence (n = 234). The association between gene signatures and recurrence was analyzed with conditional logistic regression. RESULTS Patients with the basal-like subtype had higher values of TIS, PD-1 and PD-L1 scores compared with other subtypes. Patients with higher PD-L1 score had significantly lower odds of recurrence (odds ratio [OR] 0.61, P = .01). Likewise, an increased TIS score was associated to lower, but nonsignificant odds of recurrence (OR 0.76, P = .07). Patients with human epidermal growth factor receptor 2 (HER2)-enriched subtype had significantly higher odds of recurrence compared with patients with luminal A subtype (OR 4.8, P = .03). DISCUSSION PAM50 and immune-related signatures provide important prognostic information in patients with T1abN0 breast cancer, which may refine the risk assessment in these patients.
Collapse
Affiliation(s)
- Christina M S Hassing
- Department of Breast Surgery, Herlev and Gentofte Hospital, Copenhagen University Hospital, Gentofte Hospitalsvej 1, 2900 Hellerup, Denmark.
| | - Tove Holst Filtenborg Tvedskov
- Department of Breast Surgery, Herlev and Gentofte Hospital, Copenhagen University Hospital, Gentofte Hospitalsvej 1, 2900 Hellerup, Denmark
| | - Niels Kroman
- Department of Breast Surgery, Herlev and Gentofte Hospital, Copenhagen University Hospital, Gentofte Hospitalsvej 1, 2900 Hellerup, Denmark; Danish Cancer Society, Strandboulevarden 49, 2100 Copenhagen Ø, Denmark
| | - Ann Søegaard Knoop
- Department of Oncology, Section 4262, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - Anne-Vibeke Lænkholm
- Department of Surgical Pathology, Zealand University Hospital, Sygehusvej 9 (postal: Sygehusvej 10), 4000 Roskilde, Denmark
| |
Collapse
|
58
|
Zwager MC, Yu S, Buikema HJ, de Bock GH, Ramsing TW, Thagaard J, Koopman T, van der Vegt B. Advancing Ki67 hotspot detection in breast cancer: a comparative analysis of automated digital image analysis algorithms. Histopathology 2025; 86:204-213. [PMID: 39104219 PMCID: PMC11649514 DOI: 10.1111/his.15294] [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: 02/27/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 08/07/2024]
Abstract
AIM Manual detection and scoring of Ki67 hotspots is difficult and prone to variability, limiting its clinical utility. Automated hotspot detection and scoring by digital image analysis (DIA) could improve the assessment of the Ki67 hotspot proliferation index (PI). This study compared the clinical performance of Ki67 hotspot detection and scoring DIA algorithms based on virtual dual staining (VDS) and deep learning (DL) with manual Ki67 hotspot PI assessment. METHODS Tissue sections of 135 consecutive invasive breast carcinomas were immunohistochemically stained for Ki67. Two DIA algorithms, based on VDS and DL, automatically determined the Ki67 hotspot PI. For manual assessment; two independent observers detected hotspots and calculated scores using a validated scoring protocol. RESULTS Automated hotspot detection and assessment by VDS and DL could be performed in 73% and 100% of the cases, respectively. Automated hotspot detection by VDS and DL led to higher Ki67 hotspot PIs (mean 39.6% and 38.3%, respectively) compared to manual consensus Ki67 PIs (mean 28.8%). Comparing manual consensus Ki67 PIs with VDS Ki67 PIs revealed substantial correlation (r = 0.90), while manual consensus versus DL Ki67 PIs demonstrated high correlation (r = 0.95). CONCLUSION Automated Ki67 hotspot detection and analysis correlated strongly with manual Ki67 assessment and provided higher PIs compared to manual assessment. The DL-based algorithm outperformed the VDS-based algorithm in clinical applicability, because it did not depend on virtual alignment of slides and correlated stronger with manual scores. Use of a DL-based algorithm may allow clearer Ki67 PI cutoff values, thereby improving the clinical usability of Ki67.
Collapse
Affiliation(s)
- Mieke C Zwager
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Shibo Yu
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Henk J Buikema
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Geertruida H de Bock
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | | | | | - Timco Koopman
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Pathologie FrieslandLeeuwardenThe Netherlands
| | - Bert van der Vegt
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
59
|
Lu Y, Zhu S, Wu C, Fei X, Shen K, Chen X. HER2-low status improves prognosis prediction in breast cancer patients receiving neoadjuvant treatment: A comparison of pathological stage, modified CPS+EG scoring system, and Neo-Bioscore. Chin J Cancer Res 2024; 36:729-741. [PMID: 39802892 PMCID: PMC11724176 DOI: 10.21147/j.issn.1000-9604.2024.06.10] [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: 07/17/2024] [Accepted: 11/23/2024] [Indexed: 01/16/2025] Open
Abstract
Objective To explore the prognosis-predictive influence of human epidermal growth factor receptor 2 (HER2)-low status in breast cancer patients after neoadjuvant therapy (NAT). Methods Consecutive patients with invasive breast cancer who underwent NAT and surgery from January 2009 to December 2020 at multiple centers were included. A modified CPS+EG scoring system that integrates HER2-low status, CPS+EGHlow was developed. Multiple scoring systems were compared via receiver operating characteristic curves with the area under curve (AUC), the Akaike information criterion, the C-index, and calibration curves. Results A total of 2,141 patients were included: 1,074, 640, and 427 patients in the training, internal validation, and external validation groups, respectively. HER2-low patients had a significantly better breast cancer-specific survival (BCSS, P=0.008) and recurrence-free interval (RFI, P=0.030) compared to HER2-zero patients (P=0.038) but inferior outcomes than HER2-amplified ones (BCSS, P=0.002; RFI, P<0.001). The CPS+EGHlow (AUC: 0.846, 0.817, 0.901) could stratify patients according to BCSS in training, internal validation, and external validation group, respectively, overperforming pathological stage (PS) (AUC: 0.746, 0.779, 0.754), CPS+EG (AUC: 0.771, 0.752, 0.748), and Neo-Bioscore (AUC: 0.783, 0.777, 0.786, all P<0.05). Conclusions HER2-low status showed a significant prognostic value in breast cancer patients after NAT. The CPS+EGHlow model significantly outperformed PS, CPS+EG, and Neo-Bioscore in clinical outcome prediction, which may guide further therapy targeting HER2-low.
Collapse
Affiliation(s)
- Yujie Lu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Siji Zhu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chenghui Wu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
60
|
Deckwirth V, Hundi S, Hytönen MK, Hannula S, Ellonen P, Björkenheim P, Sukura A, Lohi H. Differential somatic coding variant landscapes between laser microdissected luminal epithelial cells from canine mammary invasive ductal solid carcinoma and comedocarcinoma. BMC Cancer 2024; 24:1524. [PMID: 39696035 DOI: 10.1186/s12885-024-13239-w] [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: 01/03/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common cancer in women. Likewise, canine mammary tumors (CMT) represent the most common cancer in intact female dogs and develop in the majority spontaneously. Similarities exist in clinical presentation, histopathology, biomarkers, and treatment. However, CMT subtype-specific genomic background is less investigated. Here, we assess the genetic etiology of two histomorphological (HM) subtypes with BC counterparts, the CMT invasive ductal simple solid carcinoma (SC) and comedocarcinoma (CC), and compare the results with BC data. METHODS Groups of 11-13 transformed ductal luminal epithelial cells were laser-capture microdissected from snap-frozen invasive mammary SC and CC subtypes of one intact female dog. HM unaffected lobular luminal epithelial cells were controls. Single-cell whole genome libraries were generated using PicoPLEX and sequenced to compare the subtypes' somatic coding variant landscapes with each other and with BC data available in COSMIC-CGC and KEGG. Furthermore, HM and immunohistochemical (IHC) subtype characteristics were compared with the genomic results. RESULTS The CC had six times more variants than the SC. The SC showed variants in adherens junction genes and genes of the MAPK, mTOR and NF-kappa-B signaling pathways. In the CC, the extracellular matrix (ECM) receptor interaction, cell adhesion, PI3K-Akt and cGMP-PKG pathways were enriched, reflecting the higher cellular malignancy. Affected pathways in both CMT subtypes overlapped with BC pathways in KEGG. Additionally, we identified ATP6V1C2, GLYATL3, CARMIL3, GATAD2B, OBSCN, SIX2, CPEB3 and ZNF521 as potential new subtype-distinct driver genes. Furthermore, our results revealed biomarker alterations in IHC in the basal/myoepithelial cell layer without respective genetic mutations, suggesting changes to their complex signaling pathways, disturbed regulative feedback loops or other silencing mechanisms. CONCLUSIONS This study contributes to understanding the subtype-specific molecular mechanisms in the canine mammary invasive ductal simple SC and CC, and revealed subtype-specific molecular complexity for phenotypically similar characteristics. Several affected genes and signaling pathways overlapped with BC indicating the potential use of CMT as model for BC. Our findings emphasize the need for thorough characterization of cancer specimens with respect to translational cancer research, but also how insight into tumor heterogeneity will be crucial for the development of targeted prognostics and therapeutic interventions.
Collapse
Affiliation(s)
- Vivi Deckwirth
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sruthi Hundi
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Marjo K Hytönen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Sari Hannula
- Institute for Molecular Medicine Finland FIMM, Helsinki, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine Finland FIMM, Helsinki, Finland
| | - Pia Björkenheim
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Antti Sukura
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Hannes Lohi
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.
- Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Folkhälsan Research Center, Helsinki, Finland.
| |
Collapse
|
61
|
Shams A. Impact of prolactin treatment on enhancing the cellular responses of MCF7 breast cancer cells to tamoxifen treatment. Discov Oncol 2024; 15:797. [PMID: 39692941 DOI: 10.1007/s12672-024-01701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024] Open
Abstract
Breast cancer remains one of the most challenging diseases to treat due to its heterogeneity, propensity to recur, capacity to spread to distant vital organs, and, ultimately, patient death. Estrogen receptor-positive illness comprises the most common breast cancer subtype. Preclinical progress is hampered by the scarcity of medication-naïve estrogen receptor-positive tumour models that recapitulate metastatic development and treatment resistance. It is becoming increasingly clear that loss of differentiation and increased cellular stemness and plasticity are important causes of cancer evolution, heterogeneity, recurrence, metastasis, and treatment failure. Therefore, it has been suggested that reprogramming cancer cell differentiation could offer an effective method of reversing cancer through terminal differentiation and maturation. In this context, the hormone prolactin is well recognized for its pivotal involvement in the development of the mammary glands lobuloalveolar tissue and the terminal differentiation that drives the production of the milk protein gene and lactation. Additionally, numerous studies have examined the engagement of prolactin in breast cancer as a differentiation player that resulted in the ablation of tumour growth and progression. Here, we showed that a pre-treatment of the estrogen-positive breast cancer cell line with prolactin led to a considerable improvement in the sensitivity of this cancer cell to Tamoxifen endocrine therapy. We also showed a favourable prognostic value of prolactin receptors/estrogen receptors 1 (or alpha) co-expression on breast cancer patients outcomes, and this co-expression is highly correlated with the well-differentiated breast tumour type. Our results revealed a fruitful aspect of the effects of prolactin in improving the responses of breast cancer cells to conventional endocrine therapy. Moreover, these findings further validated the ability of prolactin as a persuader of a more differentiated and less aggressive breast cancer phenotype. Hence, it suggested a potential implication of prolactin as a therapeutic candidate.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Taif, Saudi Arabia.
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research,, Taif University, Taif 26432, Taif, Saudi Arabia.
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Taif, Saudi Arabia.
| |
Collapse
|
62
|
Wang Y, Liu L, Graff SL, Cheng L. Recent advancements in biomarkers and molecular diagnostics in hormonal receptor-positive breast cancer. Histopathology 2024. [PMID: 39687977 DOI: 10.1111/his.15395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Molecular applications have limited use in breast cancer compared to other cancer types. In recent years, with an improving appreciation of the molecular genetics of breast cancer and innovative novel targeted and immune-mediated therapeutics, opportunities have arisen for more biomarker analysis and molecular applications in the diagnosis and treatment of both locally advanced and metastatic breast cancers. In hormone receptor-positive, HER2-negative breast cancers, a growing number of revolutionized personalized therapies are in clinical use or on trials, such as CDK4/6 inhibitors and immune checkpoint inhibitors in adjuvant and neoadjuvant settings, and PIK3CA inhibitors in metastatic disease. In this review, we focus on biomarkers associated with those new therapeutic targets and molecular applications for genetic alterations associated with drug resistance or interaction from a pathology perspective for selecting and optimizing breast cancer treatment.
Collapse
Affiliation(s)
- Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liu Liu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Stephanie L Graff
- Division of Medical Oncology, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
63
|
Jørgensen JT, Egebjerg K, Lænkholm AV, Mau-Sørensen M. Human Epidermal Growth Factor Receptor 2 Pan Tumor Indication for Trastuzumab Deruxtecan. J Clin Oncol 2024; 42:4130-4133. [PMID: 39102639 DOI: 10.1200/jco.24.00884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/28/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Affiliation(s)
| | - Kristian Egebjerg
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anne-Vibeke Lænkholm
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Morten Mau-Sørensen
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
64
|
Alves da Quinta D, Rocha D, Retamales J, Giunta D, Artagaveytia N, Velazquez C, Daneri-Navarro A, Müller B, Abdelhay E, Bravo AI, Castro M, Rosales C, Alcoba E, Acosta Haab G, Carrizo F, Sorin I, Di Sibio A, Marques-Silveira M, Binato R, Caserta B, Greif G, Del Toro-Arreola A, Quintero-Ramos A, Gómez J, Podhajcer OL, Fernández EA, LACRN Investigators, Llera AS. Closing the gap: prognostic and predictive biomarker validation for personalized care in a Latin American hormone-dependent breast cancer cohort. Oncologist 2024; 29:e1701-e1713. [PMID: 39115892 PMCID: PMC11630759 DOI: 10.1093/oncolo/oyae191] [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: 12/21/2023] [Accepted: 06/22/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Several guidelines recommend the use of different classifiers to determine the risk of recurrence (ROR) and treatment decisions in patients with HR+HER2- breast cancer. However, data are still lacking for their usefulness in Latin American (LA) patients. Our aim was to evaluate the comparative prognostic and predictive performance of different ROR classifiers in a real-world LA cohort. METHODS The Molecular Profile of Breast Cancer Study (MPBCS) is an LA case-cohort study with 5-year follow-up. Stages I and II, clinically node-negative HR+HER2- patients (n = 340) who received adjuvant hormone therapy and/or chemotherapy, were analyzed. Time-dependent receiver-operator characteristic-area under the curve, univariate and multivariate Cox proportional hazards regression (CPHR) models were used to compare the prognostic performance of several risk biomarkers. Multivariate CPHR with interaction models tested the predictive ability of selected risk classifiers. RESULTS Within this cohort, transcriptomic-based classifiers such as the recurrence score (RS), EndoPredict (EP risk and EPClin), and PAM50-risk of recurrence scores (ROR-S and ROR-PC) presented better prognostic performances for node-negative patients (univariate C-index 0.61-0.68, adjusted C-index 0.77-0.80, adjusted hazard ratios [HR] between high and low risk: 4.06-9.97) than the traditional classifiers Ki67 and Nottingham Prognostic Index (univariate C-index 0.53-0.59, adjusted C-index 0.72-0.75, and adjusted HR 1.85-2.54). RS (and to some extent, EndoPredict) also showed predictive capacity for chemotherapy benefit in node-negative patients (interaction P = .0200 and .0510, respectively). CONCLUSION In summary, we could prove the clinical validity of most transcriptomic-based risk classifiers and their superiority over clinical and immunohistochemical-based methods in the heterogenous, real-world node-negative HR+HER2- MPBCS cohort.
Collapse
Affiliation(s)
- Daniela Alves da Quinta
- Laboratorio de Terapia Molecular y Celular, Fundación Instituto Leloir-CONICET, Ciudad de Buenos Aires, Argentina
- Universidad Argentina de la Empresa (UADE), Instituto de Tecnología (INTEC), Buenos Aires, Argentina
| | - Darío Rocha
- Universidad Nacional de Córdoba, Facultad de Ciencias Exactas, Físicas y Naturales, Córdoba, Argentina
| | - Javier Retamales
- Grupo Oncológico Cooperativo Chileno de Investigación, Santiago de Chile, Chile
| | - Diego Giunta
- Instituto Universitario Hospital Italiano de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Nora Artagaveytia
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | | | | | | | - Eliana Abdelhay
- Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Alicia I Bravo
- Hospital Regional de Agudos Eva Perón, San Martín, Provincia de Buenos Aires, Argentina
| | - Mónica Castro
- Instituto de Oncología Angel Roffo, Ciudad de Buenos Aires, Argentina
| | - Cristina Rosales
- Hospital Municipal de Oncología María Curie, Ciudad de Buenos Aires, Argentina
| | - Elsa Alcoba
- Hospital Municipal de Oncología María Curie, Ciudad de Buenos Aires, Argentina
| | | | - Fernando Carrizo
- Hospital Regional de Agudos Eva Perón, San Martín, Provincia de Buenos Aires, Argentina
| | - Irene Sorin
- Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | | | | | - Renata Binato
- Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Benedicta Caserta
- Department of Pathology, Centro Hospitalario Pereira Rossell, Montevideo, Uruguay
| | | | | | | | - Jorge Gómez
- Health Sciences Center, Texas A&M University, Bryan, TX 77807, United States
| | - Osvaldo L Podhajcer
- Laboratorio de Terapia Molecular y Celular, Fundación Instituto Leloir-CONICET, Ciudad de Buenos Aires, Argentina
| | - Elmer A Fernández
- Fundación para el Progreso de la Medicina, Laboratorio de Investigación en Cáncer, Córdoba, Argentina
- CONICET, Córdoba, Argentina
- FCEFyN, Depto. de Computación, Escuela de Ingeniería Biomédica, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Andrea S Llera
- Laboratorio de Terapia Molecular y Celular, Fundación Instituto Leloir-CONICET, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
65
|
Ansari N, Gaurav K, Anand A, Singh KR, Agarwal P, Agarwal A, Kumar S, Sonkar AA. Expression of cancer stem cell markers (CD24, CD44 & ALDH1A3 isoform) in Breast Cancer in Indian population considering clinicopathological characteristics and response to neoadjuvant chemotherapy - a prospective analysis from a university hospital. Indian J Surg Oncol 2024; 15:874-883. [PMID: 39555365 PMCID: PMC11564460 DOI: 10.1007/s13193-024-02013-w] [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: 05/07/2024] [Accepted: 06/26/2024] [Indexed: 11/19/2024] Open
Abstract
Cancer stem cells (CSC) are a small number of tumour propagating cells present in the bulk of tumour cells. Expression of biological markers for CSCs viz. cluster of differentiation 24 and 44 (CD24, CD44) and aldehyde dehyodeganse 1 (ALDH1) imply aggressive tumour cell behaviour and poorer outcome, particularly in breast cancer. N = 75 Tumor tissue samples from enrolled breast cancer patients were obtained and subjected to histopathological examination and immunohistochemistry (IHC) after informed consent from patients. In addition to the estrogen receptor, progesterone receptor, Her2 neu receptor, and Ki67 index; IHC was also performed for CD24, CD44 and ALDH1A3 expression. These markers' positivity was correlated with clinical parameters, therapy response and metastasis prognostication. Most patients had higher tumor stage and high nodal burden (81.3% with node positivity; N1-68%, N2 -6.6%, N3-6.6%). Study showed significant association of CD24-/CD44 + group (p = 0.021) with distant metastasis and significant association of CD24-/CD44 + /ALDH1A3 + (p = 0.008) with higher stage (T4 stage). Tumor tissues expressing CD24-/CD44 + /ALDH1A3 + group (p = 0.010) and CD24 + /CD44 + / ALDH1A3 + (p = 0.010) were significantly associated with poor response to treatment. The correlation of CD24-/CD44 + was also statistically significant (p = 0.009). Positive expression of either CD24 and/or CD44 presented with aggressive disease, larger tumours and heavy nodal burden at a younger age. Additionally, ALDH1A3 positivity signified higher metastasis rates. CD24-/CD44 + /ALDH1A3 + and CD24 + /CD44 + / ALDH1A3 + correlated with greater chances of distant metastasis, aggressive disease, limited response to chemotherapy and poorer prognosis. Thus, these observations establish the role of these CSC phenotypes as important factors for prognostic outcomes and predictors of adverse outcomes. CD24-/CD44 + /ALDH1A3 + expression could serve as an important prognostic marker and predictor of adverse outcomes in Indian breast cancer tumour biology.
Collapse
Affiliation(s)
- Nizamuddin Ansari
- Department of Surgery (General), King George’s Medical University, Lucknow, UP India
| | - Kushagra Gaurav
- Department of Surgery (General), King George’s Medical University, Lucknow, UP India
| | - Akshay Anand
- Department of Surgery (General), King George’s Medical University, Lucknow, UP India
| | - Kul Ranjan Singh
- Department of Endocrine Surgery, King George’s Medical University, Lucknow, UP India
| | - Preeti Agarwal
- Department of Pathology, King George’s Medical University, Lucknow, UP India
| | - Apoorva Agarwal
- Department of Pathology, King George’s Medical University, Lucknow, UP India
| | - Surender Kumar
- Department of Surgery (General), King George’s Medical University, Lucknow, UP India
| | - Abhinav Arun Sonkar
- Department of Surgery (General), King George’s Medical University, Lucknow, UP India
| |
Collapse
|
66
|
Luo HJ, Ren JL, Mei Guo L, Liang Niu J, Song XL. MRI-based machine learning radiomics for prediction of HER2 expression status in breast invasive ductal carcinoma. Eur J Radiol Open 2024; 13:100592. [PMID: 39149534 PMCID: PMC11324846 DOI: 10.1016/j.ejro.2024.100592] [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: 04/18/2024] [Revised: 07/08/2024] [Accepted: 07/14/2024] [Indexed: 08/17/2024] Open
Abstract
Background Human epidermal growth factor receptor 2 (HER2) is a tumor biomarker with significant prognostic and therapeutic implications for invasive ductal breast carcinoma (IDC). Objective This study aimed to explore the effectiveness of a multisequence magnetic resonance imaging (MRI)-based machine learning radiomics model in classifying the expression status of HER2, including HER2-positive, HER2-low, and HER2 completely negative (HER2-zero), among patients with IDC. Methods A total of 402 female patients with IDC confirmed through surgical pathology were enrolled and subsequently divided into a training group (n = 250, center I) and a validation group (n = 152, center II). Radiomics features were extracted from the preoperative MRI. A simulated annealing algorithm was used for key feature selection. Two classification tasks were performed: task 1, the classification of HER2-positive vs. HER2-negative (HER2-low and HER2-zero), and task 2, the classification of HER2-low vs. HER2-zero. Logistic regression, random forest (RF), and support vector machine were conducted to establish radiomics models. The performance of the models was evaluated using the area under the curve (AUC) of the operating characteristics (ROC). Results In total, 4506 radiomics features were extracted from multisequence MRI. A radiomics model for prediction of expression state of HER2 was successfully developed. Among the three classification algorithms, RF achieved the highest performance in classifying HER2-positive from HER2-negative and HER2-low from HER2-zero, with AUC values of 0.777 and 0.731, respectively. Conclusions Machine learning-based MRI radiomics may aid in the non-invasive prediction of the different expression status of HER2 in IDC.
Collapse
Affiliation(s)
- Hong-Jian Luo
- Department of Radiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zuiyi, Guizhou province, China
| | | | - Li Mei Guo
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| | - Jin Liang Niu
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| | - Xiao-Li Song
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| |
Collapse
|
67
|
Bhat-Nakshatri P, Gao H, Khatpe AS, Adebayo AK, McGuire PC, Erdogan C, Chen D, Jiang G, New F, German R, Emmert L, Sandusky G, Storniolo AM, Liu Y, Nakshatri H. Single-nucleus chromatin accessibility and transcriptomic map of breast tissues of women of diverse genetic ancestry. Nat Med 2024; 30:3482-3494. [PMID: 39122969 PMCID: PMC11976273 DOI: 10.1038/s41591-024-03011-9] [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: 10/04/2023] [Accepted: 04/22/2024] [Indexed: 08/12/2024]
Abstract
Single-nucleus analysis allows robust cell-type classification and helps to establish relationships between chromatin accessibility and cell-type-specific gene expression. Here, using samples from 92 women of several genetic ancestries, we developed a comprehensive chromatin accessibility and gene expression atlas of the breast tissue. Integrated analysis revealed ten distinct cell types, including three major epithelial subtypes (luminal hormone sensing, luminal adaptive secretory precursor (LASP) and basal-myoepithelial), two endothelial and adipocyte subtypes, fibroblasts, T cells, and macrophages. In addition to the known cell identity genes FOXA1 (luminal hormone sensing), EHF and ELF5 (LASP), TP63 and KRT14 (basal-myoepithelial), epithelial subtypes displayed several uncharacterized markers and inferred gene regulatory networks. By integrating breast epithelial cell gene expression signatures with spatial transcriptomics, we identified gene expression and signaling differences between lobular and ductal epithelial cells and age-associated changes in signaling networks. LASP cells and fibroblasts showed genetic ancestry-dependent variability. An estrogen receptor-positive subpopulation of LASP cells with alveolar progenitor cell state was enriched in women of Indigenous American ancestry. Fibroblasts from breast tissues of women of African and European ancestry clustered differently, with accompanying gene expression differences. Collectively, these data provide a vital resource for further exploring genetic ancestry-dependent variability in healthy breast biology.
Collapse
Affiliation(s)
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Aditi S Khatpe
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adedeji K Adebayo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Patrick C McGuire
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cihat Erdogan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Duojiao Chen
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Felicia New
- NanoString Technology Inc., Seattle, WA, USA
| | - Rana German
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lydia Emmert
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - George Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anna Maria Storniolo
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- VA Roudebush Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
68
|
Colomer R, González-Farré B, Ballesteros AI, Peg V, Bermejo B, Pérez-Mies B, de la Cruz S, Rojo F, Pernas S, Palacios J. Biomarkers in breast cancer 2024: an updated consensus statement by the Spanish Society of Medical Oncology and the Spanish Society of Pathology. Clin Transl Oncol 2024; 26:2935-2951. [PMID: 38869741 PMCID: PMC11564209 DOI: 10.1007/s12094-024-03541-1] [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: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024]
Abstract
This revised consensus statement of the Spanish Society of Medical Oncology (SEOM) and the Spanish Society of Pathological Anatomy (SEAP) updates the recommendations for biomarkers use in the diagnosis and treatment of breast cancer that we first published in 2018. The expert group recommends determining in early breast cancer the estrogen receptor (ER), progesterone receptor (PR), Ki-67, and Human Epidermal growth factor Receptor 2 (HER2), as well as BReast CAncer (BRCA) genes in high-risk HER2-negative breast cancer, to assist prognosis and help in indicating the therapeutic options, including hormone therapy, chemotherapy, anti-HER2 therapy, and other targeted therapies. One of the four available genetic prognostic platforms (Oncotype DX®, MammaPrint®, Prosigna®, or EndoPredict®) may be used in ER-positive patients with early breast cancer to establish a prognostic category and help decide with the patient whether adjuvant treatment may be limited to hormonal therapy. In second-line advanced breast cancer, in addition, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and estrogen receptor 1 (ESR1) should be tested in hormone-sensitive cases, BRCA gene mutations in HER2-negative cancers, and in triple-negative breast cancer (TNBC), programmed cell death-1 ligand (PD-L1). Newer biomarkers and technologies, including tumor-infiltrating lymphocytes (TILs), homologous recombination deficiency (HRD) testing, serine/threonine kinase (AKT) pathway activation, and next-generation sequencing (NGS), are at this point investigational.
Collapse
Affiliation(s)
- Ramon Colomer
- UAM Personalised Precision Medicine Chair & Medical Oncology Department, La Princesa University Hospital and Research Institute, C/Diego de León, 62, 28006, Madrid, Spain.
| | | | | | - Vicente Peg
- Pathological Anatomy Service, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Begoña Bermejo
- Medical Oncology Department, Biomedical Research Institute INCLIVA, Medicine Department of the University of Valencia and Clinic University Hospital, Valencia, Spain
| | - Belén Pérez-Mies
- Pathological Anatomy Service, Ramón y Cajal University Hospital, Faculty of Medicine, University of Alcalá, IRYCIS and CIBERONC, Madrid, Spain
| | - Susana de la Cruz
- Medical Oncology Department, Navarra University Hospital, Navarre, Spain
| | - Federico Rojo
- Anatomy Service, Fundación Jiménez Díaz University Hospital and CIBERONC, Madrid, Spain
| | - Sonia Pernas
- Oncology Department, Catalan Institute of Oncology (ICO)-IDIBELL, L'Hospitalet, Barcelona, Spain
| | - José Palacios
- Pathological Anatomy Service, Department of Pathology, Ramón y Cajal University Hospital, Faculty of Medicine, University of Alcalá, IRYCIS and CIBERONC, Ctra. Colmenar Viejo, Km 9,1, 28034, Madrid, Spain.
| |
Collapse
|
69
|
Zilenaite-Petrulaitiene D, Rasmusson A, Valkiuniene RB, Laurinaviciene A, Petkevicius L, Laurinavicius A. Spatial distributions of CD8 and Ki67 cells in the tumor microenvironment independently predict breast cancer-specific survival in patients with ER+HER2- and triple-negative breast carcinoma. PLoS One 2024; 19:e0314364. [PMID: 39576843 PMCID: PMC11584100 DOI: 10.1371/journal.pone.0314364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024] Open
Abstract
INTRODUCTION Breast cancer (BC) presents diverse malignancies with varying biological and clinical behaviors, driven by an interplay between cancer cells and tumor microenvironment. Deciphering these interactions is crucial for personalized diagnostics and treatment. This study explores the prognostic impact of tumor proliferation and immune response patterns, assessed by computational pathology indicators, on breast cancer-specific survival (BCSS) models in estrogen receptor-positive HER2-negative (ER+HER2-) and triple-negative BC (TNBC) patients. MATERIALS AND METHODS Whole-slide images of tumor surgical excision samples from 252 ER+HER2- patients and 63 TNBC patients stained for estrogen and progesterone receptors, Ki67, HER2, and CD8 were analyzed. Digital image analysis (DIA) was performed for tumor tissue segmentation and quantification of immunohistochemistry (IHC) markers; the DIA outputs were subsampled by hexagonal grids to assess the spatial distributions of Ki67-positive tumor cells and CD8-positive (CD8+) cell infiltrates, expressed as Ki67-entropy and CD8-immunogradient indicators, respectively. Prognostic models for BCSS were generated using multivariable Cox regression analysis, integrating clinicopathological and computational IHC indicators. RESULTS In the ER+HER2- BC, multivariable Cox regression revealed that high CD8+ density within the tumor interface zone (IZ) (HR: 0.26, p = 0.0056), low immunodrop indicator of CD8+ density (HR: 2.93, p = 0.0051), and low Ki67-entropy (HR: 5.95, p = 0.0.0061) were independent predictors of better BCSS, while lymph node involvement predicted worse BCSS (HR: 3.30, p = 0.0013). In TNBC, increased CD8+ density in the IZ stroma (HR: 0.19, p = 0.0119) and Ki67-entropy (HR: 3.31, p = 0.0250) were independent predictors of worse BCSS. Combining these independent indicators enhanced prognostic stratification in both BC subtypes. CONCLUSIONS Computational biomarkers, representing spatial properties of the tumor proliferation and immune cell infiltrates, provided independent prognostic information beyond conventional IHC markers in BC. Integrating Ki67-entropy and CD8-immunogradient indicators into prognostic models can improve patient stratification with regard to BCSS.
Collapse
Affiliation(s)
- Dovile Zilenaite-Petrulaitiene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Institute of Informatics, Faculty of Mathematics and Informatics, Vilnius University, Vilnius, Lithuania
| | - Allan Rasmusson
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Ruta Barbora Valkiuniene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Aida Laurinaviciene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Linas Petkevicius
- Institute of Informatics, Faculty of Mathematics and Informatics, Vilnius University, Vilnius, Lithuania
| | - Arvydas Laurinavicius
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
70
|
Turashvili G, Gao Y, Ai DA, Ewaz AM, Gjeorgjievski SG, Wang Q, Nguyen TTA, Zhang C, Li X. Low interobserver agreement among subspecialised breast pathologists in evaluating HER2-low breast cancer. J Clin Pathol 2024; 77:815-821. [PMID: 37714693 DOI: 10.1136/jcp-2023-209055] [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: 07/11/2023] [Accepted: 08/24/2023] [Indexed: 09/17/2023]
Abstract
AIMS Metastatic HER2-low breast cancer (HLBC) can be treated by trastuzumab deruxtecan. Assessment of low levels of HER2 protein expression suffers from poor interobserver reproducibility. The aim of the study was to evaluate the interobserver agreement among subspecialised breast pathologists and develop a practical algorithm for assessing HLBC. METHODS Six breast pathologists (4 juniors, 2 seniors) evaluated 106 HER2 immunostained slides with 0/1+expression. Two rounds (R1, R2) of ring study were performed before and after training with a modified Ki-67 algorithm, and concordance was assessed. RESULTS Agreement with 5% increments increased from substantial to almost perfect (R1: 0.796, R2: 0.804), and remained substantial for three categories (<1% vs 1%-10% vs >10%) (R1: 0.768, R2: 0.764). Seniors and juniors had almost perfect agreement with 5% increments (R1: 0.859 and 0.821, R2: 0.872 and 0.813). For the three categories, agreement remained almost perfect among seniors (R1: 0.837, R2: 0.860) and substantial among juniors (R1: 0.792, R2: 0.768). Binary analysis showed suboptimal agreement, decreasing for both juniors and seniors from substantial (R1: 0.650 and 0.620) to moderate (R2: 0.560 and 0.554) using the 1% cut-off, and increasing from moderate to substantial (R1: 0.478, R2: 0.712) among seniors but remaining moderate (R1: 0.576, R2: 0.465) among juniors using the 10% cut-off. The average scoring time per case was higher (72 vs 92 s). CONCLUSIONS Subspecialised breast pathologists have suboptimal agreement for immunohistochemical evaluation of HLBC using the modified Ki-67 methodology. An urgent need remains for a new assay/algorithm to reliably evaluate HLBC.
Collapse
Affiliation(s)
- Gulisa Turashvili
- Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| | - Yuan Gao
- Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| | - Di Andy Ai
- Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| | - Abdulwahab M Ewaz
- Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| | | | - Qun Wang
- Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| | - Thi T A Nguyen
- Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| | - Chao Zhang
- General Dynamics Information Technology Inc, Falls Church, Virginia, USA
| | - Xiaoxian Li
- Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| |
Collapse
|
71
|
Mueller C, Zimmermann JSM, Radosa MP, Hahn AK, Kaya AC, Huwer S, Stotz L, Wagenpfeil G, Radosa CG, Solomayer EF, Radosa JC. Correlation of preoperative sonographic staging and postoperative histopathologic staging in patients with invasive breast cancer. Arch Gynecol Obstet 2024; 310:2623-2630. [PMID: 39222087 PMCID: PMC11485201 DOI: 10.1007/s00404-024-07699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE To assess the accuracy of preoperative sonographic staging in patients with primary invasive breast cancer. METHODS We retrospectively analyzed a prospectively kept service database of patients with newly diagnosed, unifocal, cT1-3, invasive breast cancer. All patients were diagnosed at a single center institution between January 2013 and December 2021. Clinical T stage was assessed preoperatively by ultrasound and correlated with the definite postoperative pathologic T stage. Demographics, clinical and pathological characteristics were collected. Factors influencing accuracy, over- and underdiagnosis of sonographic staging were analyzed with multivariable regression analysis. RESULTS A total of 2478 patients were included in the analysis. Median patients' age was 65 years. 1577 patients (63.6%) had clinical T1 stage, 864 (34.9%) T2 and 37 (1.5%) T3 stage. The overall accuracy of sonography and histology was 76.5% (n = 1896), overestimation was observed in 9.1% (n = 225) of all cases, while underestimation occurred in 14.4% (n = 357) of all cases. Accuracy increased when clinical tumor stage cT was higher (OR 1.23; 95% CI 1.10-1.38, p ≤ 0.001). The highest accuracy was seen for patients with T2 stage (82.8%). The accuracy was lower in Luminal B tumors compared to Luminal A tumors (OR 0.71; 95% CI 0.59-0.87, p ≤ 0.001). We could not find any association between sonographic accuracy in HER2 positive patients, and demographic characteristics, or tumor-related factors. CONCLUSION Our unicentric study showed a high accuracy of sonography in predicting T stage, especially for tumors with clinical T2 stage. Tumor stage and biological tumor factors do affect the accuracy of sonographic staging.
Collapse
Affiliation(s)
- Carolin Mueller
- Department of Gynecology, Obstetrics, and Reproductive Medicine, Saarland University Medical Center, 66421, Homburg, Saar, Germany
- Outcomes Research Consortium, Department of Anesthesiology, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Julia Sarah Maria Zimmermann
- Department of Gynecology, Obstetrics, and Reproductive Medicine, Saarland University Medical Center, 66421, Homburg, Saar, Germany
| | - Marc Philipp Radosa
- Department of Gynecology and Obstetrics, Klinikum Bremen-Nord, Bremen, Germany
| | - Anna Katharina Hahn
- Department of Gynecology, Obstetrics, and Reproductive Medicine, Saarland University Medical Center, 66421, Homburg, Saar, Germany
| | - Askin Canguel Kaya
- Department of Gynecology, Obstetrics, and Reproductive Medicine, Saarland University Medical Center, 66421, Homburg, Saar, Germany
| | - Sarah Huwer
- Department of Obstetrics & Gynecology, Medical Center-University of Freiburg, 79106, Freiburg, Germany
| | - Lisa Stotz
- Department of Gynecology, Obstetrics, and Reproductive Medicine, Saarland University Medical Center, 66421, Homburg, Saar, Germany
| | - Gudrun Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical Informatics (IMBEI), Saarland University, Campus Homburg, 66421, Homburg, Germany
| | - Christoph Georg Radosa
- Institute and Polyclinic of Diagnostic and Interventional Radiology, Medical University, TU Dresden, Dresden, Germany
| | - Erich-Franz Solomayer
- Department of Gynecology, Obstetrics, and Reproductive Medicine, Saarland University Medical Center, 66421, Homburg, Saar, Germany
| | - Julia Caroline Radosa
- Department of Gynecology, Obstetrics, and Reproductive Medicine, Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| |
Collapse
|
72
|
Ju X, Chen Z, Yan H, Luo B, Zhao F, Huang A, Chen X, Yuan J. Correlation analysis of Ki67 changes with survival outcomes in breast cancer before and after neoadjuvant therapy based on residual cancer Burden grade. Pathol Res Pract 2024; 263:155650. [PMID: 39405801 DOI: 10.1016/j.prp.2024.155650] [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: 02/18/2024] [Revised: 09/19/2024] [Accepted: 10/10/2024] [Indexed: 11/10/2024]
Abstract
PURPOSE This study aims to investigate the change of Ki67 value pre- and post-neoadjuvant therapy (NAT) and evaluate its potential value in predicting survival outcomes in different molecular subtypes of breast cancer. METHODS A total of 257 breast cancer patients who underwent NAT at Renmin Hospital of Wuhan University from July 2019 to Sep 2023 were included in this study. The Ki67 index of the patients was re-interpreted by two attending physicians, and the changes of Ki67 value pre- and post-NAT were compared. Chi-square test (χ2) and logistic regression were conducted to examine the correlation between various characteristics and the efficacy of NAT. Disease-free survival (DFS) was calculated using the Kaplan-Meier curve and compared using the log-rank test. RESULTS Patients with higher histological grade, negative expression of estrogen receptor (ER) or progesterone receptor (PR), positive expression of human epidermal growth receptor 2 (HER2), higher pretreatment Ki67 index, absence of lymph node metastasis, and those with HER2 positive and triple-negative breast cancer were associated with improved efficacy of NAT. Our study identified that the optimal cut-off value for the changes in Ki67 index pre- and post-NAT related to the effectiveness of NAT was "-88.19 %" in whole chort, which was related to the aforementioned clinical characteristics. Besides, the optimal cut-off values for the luminal, HER2-enriched and triple-negative subtypes were "-91.83 %", "-46.12 %" and "-81.67 %", respectively. Survival analysis demonstrated that the changes in Ki67 value were significantly associated with DFS in the HER2-enriched and triple-negative subtype, but not in the luminal subtype. CONCLUSIONS Preoperative clinicopathological features and changes in Ki67 value pre-and post-NAT can contribute to providing patients with a more accurate prognosis.
Collapse
Affiliation(s)
- Xianli Ju
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhengzhuo Chen
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Bin Luo
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Fangrui Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Aoling Huang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xi Chen
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
73
|
Michaud O, Ahmad M, Hoda SA. Biomarker Testing in Microinvasive Carcinoma of the Breast. Am J Surg Pathol 2024; 48:1475-1479. [PMID: 38809275 DOI: 10.1097/pas.0000000000002252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Affiliation(s)
- Olivier Michaud
- Department of Pathology, Weill Cornell Medicine, New York, NY
| | | | | |
Collapse
|
74
|
Zhang L, Ning N, Liang H, Zhao S, Gao X, Liu A, Song Q, Duan X, Yang J, Xie L. The contrast-free diffusion MRI multiple index for the early prediction of pathological response to neoadjuvant chemotherapy in breast cancer. NMR IN BIOMEDICINE 2024; 37:e5176. [PMID: 38884131 DOI: 10.1002/nbm.5176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/21/2024] [Accepted: 04/21/2024] [Indexed: 06/18/2024]
Abstract
Early tumor response prediction can help avoid overtreatment with unnecessary chemotherapy sessions. It is important to determine whether multiple apparent diffusion coefficient indices (S index, ADC-diff) are effective in the early prediction of pathological response to neoadjuvant chemotherapy (NAC) in breast cancer (BC). Patients with stage II and III BCs who underwent T1WI, diffusion-weighted imaging (DWI), and dynamic contrast-enhanced MRI using a 3 T system were included. They were divided into two groups: major histological responders (MHRs, Miller-Payne G4/5) and nonmajor histological responders (nMHRs, Miller-Payne G1-3). Three b values were used for DWI to derive the S index; ADC-diff values were obtained using b = 0 and 1000 s/mm2. The different interquartile ranges of percentile S-index and ADC-diff values after treatment were calculated and compared. The assessment was performed at baseline and after two and four NAC cycles. A total of 59 patients were evaluated. There are some correlations of interquartile ranges of S-index parameters and ADC-diff values with histopathological prognostic factors (such as estrogen receptor and human epidermal growth factor receptor 2 expression, all p < 0.05), but no significant differences were found in some other interquartile ranges of S-index parameters or ADC-diff values between progesterone receptor positive and negative or for Ki-67 tumors (all P > 0.05). No differences were found in the dynamic contrast-enhanced MRI characteristics between the two groups. HER-2 expression and kurtosis of the S-index distribution were screened out as independent risk factors for predicting MHR group (p < 0.05, area under the curve (AUC) = 0.811) before NAC. After early NAC (two cycles), only the 10th percentile S index was statistically significant between the two groups (p < 0.05, AUC = 0.714). No significant differences were found in ADC-diff value at any time point of NAC between the two groups (P > 0.1). These findings demonstrate that the S-index value may be used as an early predictor of pathological response to NAC in BC; the value of ADC-diff as an imaging biomarker of NAC needs to be further confirmed by ongoing multicenter prospective trials.
Collapse
Affiliation(s)
- Lina Zhang
- PET-CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ning Ning
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hongbing Liang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Siqi Zhao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xue Gao
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ailian Liu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingwei Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaoyi Duan
- PET-CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jie Yang
- School of Public Health, Dalian Medical University, Dalian, China
| | - Lizhi Xie
- GE Healthcare, MR Research China, Beijing, China
| |
Collapse
|
75
|
Liu Y, Liu T, Zhu Z, Xie L, Bai D, Liu T, Gu W, Li W, Shu Y, Zhang J. An advanced hydrogel dressing system with progressive delivery and layer-to-layer response for diabetic wound healing. Acta Biomater 2024:S1742-7061(24)00638-X. [PMID: 39486779 DOI: 10.1016/j.actbio.2024.10.046] [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: 05/15/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Wound healing in diabetic patients presents a significant challenge due to delayed inflammatory responses, which obstruct subsequent healing stages. In response, we have developed a progressive, layer-by-layer responsive hydrogel, specifically designed to meet the dynamic requirements of diabetic wounds throughout different healing phases. This hydrogel initiates with a glucose-responsive layer formed by boronate ester bonds between 4-arm-poly (ethylene glycol) succinimidyl glutarate (4arm-PEG-SG) and 3-aminophenylboronic acid. This configuration ensures precise control over the physicochemical properties, facilitating accurate drug release during the healing process. Furthermore, we have incorporated an active pharmaceutical ingredient ionic liquid (API) composed of diclofenac and L-carnitine. This combination effectively tackles the solubility and stability issues commonly associated with anti-inflammatory drugs. To further refine drug release, we integrated matrix metalloproteinase-9 (MMP-9)-sensitive gelatin microcapsules, ensuring a controlled release and preventing the abrupt, uneven drug distribution often seen in other systems. Our hydrogel's rheological properties closely resemble human skin, offering a more harmonious approach to diabetic wound healing. Overall, this progressive layer-by-layer responsive wound management system, which is a safe, efficient, and intelligent approach, holds significant potential for the clinical treatment of diabetic wounds. STATEMENT OF SIGNIFICANCE: The two main problems of diabetic wounds are the long-term infiltration of inflammation and the delayed repair process. In this experiment, a glucose-responsive hierarchical drug delivery system was designed to intelligently adjust gel properties to meet the needs of inflammation and repair stage of wound healing, accelerate the transformation of inflammation and repair stage, and accelerate the process of repair stage. In addition, in order to achieve accurate drug release in anti-inflammatory layer hydrogels and avoid sudden drug release due to poor solubility of anti-inflammatory small molecule drugs, we constructed a ionic liquid of active pharmaceutical ingredients (API-ILs) using diclofenac and L-carnitine as raw materials. It was wrapped in MMP-9 enzyme active gelatin microcapsule to construct a double-reaction anti-inflammatory layer gel to achieve accurate drug release. These findings highlight the potential of our system in treating diabetic wounds, providing a significant advance in wound treatment.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China; Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, PR China; Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China
| | - Tianqi Liu
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China; Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China.
| | - Zhenye Zhu
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China
| | - Lin Xie
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China
| | - De Bai
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China
| | - Tonglin Liu
- Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
| | - Wenting Gu
- Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China
| | - Wei Li
- Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China
| | - Yang Shu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, PR China.
| | - Jiaheng Zhang
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China; Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China; Shenzhen Shinehigh Innovation Technology Co., Ltd., Shenzhen 518055, PR China.
| |
Collapse
|
76
|
Cebrecos I, Torras I, Castillo H, Pumarola C, Ganau S, Sitges C, Vidal-Sicart S, Schettini F, Sanfeliu E, Loinaz I, Garcia M, Oses G, Molla M, Vidal M, Mension E. Predicting Additional Metastases in Axillary Lymph Node Dissection After Neoadjuvant Chemotherapy: Ratio of Positive/Total Sentinel Nodes. Cancers (Basel) 2024; 16:3638. [PMID: 39518078 PMCID: PMC11545455 DOI: 10.3390/cancers16213638] [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: 09/15/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES The aim of the study was to determine the clinical value of the sentinel lymph node ratio (SLN-R) in predicting additional positive lymph nodes during axillary lymph node dissection (ALND) in breast cancer patients following neoadjuvant chemotherapy (NAC). METHODS A cross-sectional study was performed at a single institution evaluating data from 1521 BC patients. Inclusion criteria comprised cT1/cT4, cN0/cN1 status with positive post-NAC axillary staging by SLN/TAD, respectively, and subsequent ALND. RESULTS The study included 118 patients, divided into two groups based on the presence or absence of additional node metastasis at ALND: 39 in the residual disease group (RD) and 79 in the non-residual disease group (nRD). Univariate logistic regression analysis of SLN-R was conducted to assess its predictive value, yielding an odds ratio (OR) of 7.79 (CI 1.92-29.5, p = 0.003). An SLN-R cut-off point of <0.35 was identified using ROC curve analysis, with a false-negative rate of 10.2%, as a predictor for no additional metastasis at ALND following post-NAC SLN/TAD positivity. CONCLUSIONS The study concludes that SLN-R is a valuable predictor for determining the omission of ALND in cases where SLN/TAD is positive after NAC. This metric, in combination with other clinical variables, could help develop a nomogram to spare patients from ALND.
Collapse
Affiliation(s)
- Isaac Cebrecos
- Department of Obstetrics and Gynecology and Neonatology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (I.C.); (I.L.)
- Faculty of Medicine, University of Barcelona, 08007 Barcelona, Spain (M.M.); (M.V.)
| | - Ines Torras
- Department of Obstetrics and Gynecology and Neonatology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (I.C.); (I.L.)
- Faculty of Medicine, University of Barcelona, 08007 Barcelona, Spain (M.M.); (M.V.)
| | - Helena Castillo
- Department of Obstetrics and Gynecology and Neonatology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (I.C.); (I.L.)
| | - Claudia Pumarola
- Department of Obstetrics and Gynecology and Neonatology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (I.C.); (I.L.)
| | - Sergi Ganau
- Department of Radiology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (S.G.)
| | - Carla Sitges
- Department of Radiology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (S.G.)
| | - Sergi Vidal-Sicart
- Department of Nuclear Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain;
- Diagnosis and Therapy in Oncology Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Francesco Schettini
- Faculty of Medicine, University of Barcelona, 08007 Barcelona, Spain (M.M.); (M.V.)
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Esther Sanfeliu
- Faculty of Medicine, University of Barcelona, 08007 Barcelona, Spain (M.M.); (M.V.)
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Pathology, Biomedical Diagnostic Center, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Ignacio Loinaz
- Department of Obstetrics and Gynecology and Neonatology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (I.C.); (I.L.)
| | - Marta Garcia
- Department of Obstetrics and Gynecology and Neonatology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (I.C.); (I.L.)
| | - Gabriela Oses
- Department of Radiation Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Meritxell Molla
- Faculty of Medicine, University of Barcelona, 08007 Barcelona, Spain (M.M.); (M.V.)
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Radiation Oncology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Maria Vidal
- Faculty of Medicine, University of Barcelona, 08007 Barcelona, Spain (M.M.); (M.V.)
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Eduard Mension
- Department of Obstetrics and Gynecology and Neonatology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (I.C.); (I.L.)
- Faculty of Medicine, University of Barcelona, 08007 Barcelona, Spain (M.M.); (M.V.)
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
77
|
Leigh J, McGee SF, Vandermeer L, Williams P, Rushton M. A Multicenter Physician Survey Evaluating the Use of Ki-67 in Breast Cancer Management in Canada. Biomedicines 2024; 12:2471. [PMID: 39595037 PMCID: PMC11592389 DOI: 10.3390/biomedicines12112471] [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: 10/04/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Ki-67's response to pre-operative endocrine therapy (ET) in early breast cancer is an evidence-based tool to guide adjuvant treatment decisions. Physicians across Canada were surveyed to explore current practice patterns and perceived barriers to the use of Ki-67 in practice. METHODS Physicians were invited to participate in an anonymous survey and were eligible if they prescribed systemic therapy for breast cancer in Canada. Respondents were asked to describe their usage of Ki-67, perceptions of the evidence surrounding Ki-67 ET response, and interest in future trials using this approach. RESULTS The survey received 48/163 responses (29.4%). The majority of respondents (97.6%) reported access to Ki-67 testing upon request. Treatment decisions for adjuvant Abemaciclib was the most common reason (97.6%), followed by adjuvant chemotherapy decisions (16.7%). Only 19.0% had used Ki-67's response to pre-operative ET in practice. Common barriers to this approach that were identified included a lack of awareness from other providers (54.8%), an increased resource requirement (54.8%), and a lack of timely medical oncology consultation (52.4%). The majority of physicians (85.3%) reported that they would participate in future trials using the Ki-67 endocrine response, and that rate of treatment decision change (95.2%) and cost analysis (42.3%) were important endpoints. CONCLUSIONS Despite the widespread availability of Ki-67 testing, few physicians in Canada currently use it to assess endocrine response, predominantly due to logistical and resource constraints. There is a high level of interest in participating in future trials using this strategy, which should focus on disease related outcomes, feasibility, and the financial impact on the public healthcare system.
Collapse
Affiliation(s)
- Jennifer Leigh
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada (M.R.)
| | - Sharon F. McGee
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada (M.R.)
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Lisa Vandermeer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Phillip Williams
- Department of Pathology, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
| | - Moira Rushton
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada (M.R.)
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
78
|
Zhao Y, Gong J, Liu H, Huang H, Tan WS, Cai H. A chemically defined, mechanically tunable, and bioactive hyaluronic acid/alginate double-network hydrogel for liver cancer organoid construction. Int J Biol Macromol 2024; 282:136707. [PMID: 39442832 DOI: 10.1016/j.ijbiomac.2024.136707] [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: 06/04/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Liver cancer organoids replicate the pathophysiology of primary tumors, making them ideal for drug screening and efficacy evaluation. However, their growth in complex, variable, animal-derived matrices hinders practical application. Here, we designed an easily accessible, chemically defined, biocompatible double-network hydrogel (HADR) using methacrylated hyaluronic acid (HAMA), sodium alginate (SA), methacrylamide dopamine (DMA), and c(RGDFC) for liver cancer organoid culture. By optimizing critical extracellular matrix (ECM) parameters, the HADR hydrogel achieves compatibility with the physiological mechanics of the human liver and fosters the adhesion and proliferation of multiple cell types. In vitro drug efficacy tests showed that HepG2 cell line-derived liver cancer organoids exhibited higher IC50 values than 2D cultures, indicating greater drug resistance. Subcutaneous tumor models in nude mice revealed that HADR hydrogels created a microenvironment for HepG2 cells mirroring the natural tumor ECM, leading to increased tumor volume, denser cell arrangement, and concurrent microvascular development. In vivo drug efficacy evaluations indicated that DOX treatment downregulated Ki-67 and MMP-9 expression, inhibiting HepG2 cell proliferation, invasion, and metastasis. These findings demonstrate the potential of HADR hydrogels for liver cancer organoid culture, offering new strategies for personalized drug screening and efficacy evaluation.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Junjie Gong
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Hanwen Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Huimin Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| |
Collapse
|
79
|
Chang H, Chen J, Wang D, Li H, Ming L, Li Y, Yu D, Yang YX, Kong P, Jia W, Yan Q, Liu X, Zeng Q. Multimodal apparent diffusion MRI model in noninvasive evaluation of breast cancer and Ki-67 expression. Cancer Imaging 2024; 24:137. [PMID: 39394171 PMCID: PMC11470582 DOI: 10.1186/s40644-024-00780-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: 03/11/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND To assess the capability of multimodal apparent diffusion (MAD) weighted magnetic resonance imaging (MRI) to distinguish between malignant and benign breast lesions, and to predict Ki-67 expression level in breast cancer. METHODS This retrospective study was conducted with 93 patients who had postoperative pathology-confirmed breast cancer or benign breast lesions. MAD images were acquired using a 3.0 T MRI scanner with 16 b values. The MAD parameters, as flow (fF, DF), unimpeded (fluid) (fUI), hindered (fH, DH, and αH), and restricted (fR, DR), were calculated. The differences of the parameters were compared by Mann-Whitney U test between the benign/malignant lesions and high/low Ki-67 expression level. The diagnostic performance was assessed by the area under the receiver operating characteristic curve (AUC). RESULTS The fR in the malignant lesions was significantly higher than in the benign lesions (P = 0.001), whereas the fUI and DH were found to be significantly lower (P = 0.007 and P < 0.001, respectively). Compared with individual parameter in differentiating malignant from benign breast lesions, the combination parameters of MAD (fR, DH, and fUI) provided the highest AUC (0.851). Of the 73 malignant lesions, 42 (57.5%) were assessed as Ki-67 low expression and 31 (42.5%) were Ki-67 high expression. The Ki-67 high status showed lower DH, higher DF and higher αH (P < 0.05). The combination parameters of DH, DF, and αH provided the highest AUC (0.691) for evaluating Ki-67 expression level. CONCLUSIONS MAD weighted MRI is a useful method for the breast lesions diagnostics and the preoperative prediction of Ki-67 expression level.
Collapse
Affiliation(s)
- Huan Chang
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766 Jingshi Road, Jinan, Shandong, China
| | - Jinming Chen
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766 Jingshi Road, Jinan, Shandong, China
| | - Dawei Wang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Hongxia Li
- Department of Radiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Ming
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yuting Li
- Department of Radiology, The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Dan Yu
- United Imaging Research Institute of Intelligent Imaging, Beijing, People's Republic of China
| | - Yu Xin Yang
- United Imaging Research Institute of Intelligent Imaging, Beijing, People's Republic of China
| | - Peng Kong
- Department of Breast Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Wenjing Jia
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical Universityand, Shandong Academy of Medical Sciences , Jinan, Shandong, China
| | - Qingqing Yan
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical Universityand, Shandong Academy of Medical Sciences , Jinan, Shandong, China
| | - Xinhui Liu
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical Universityand, Shandong Academy of Medical Sciences , Jinan, Shandong, China
| | - Qingshi Zeng
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766 Jingshi Road, Jinan, Shandong, China.
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China.
| |
Collapse
|
80
|
Shen Y, Guan X, Li S, Hou X, Yu J, Yin H, Shan X, Han X, Wang L, Zhou B, Li X, Sun L, Zhang Y, Xu H, Yue W. Exploiting a tumor softening targeted bomb for mechanical gene therapy of chemoresistant Triple-Negative breast cancer. CHEMICAL ENGINEERING JOURNAL 2024; 498:155217. [DOI: 10.1016/j.cej.2024.155217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
81
|
Hanna MG, Olson NH, Zarella M, Dash RC, Herrmann MD, Furtado LV, Stram MN, Raciti PM, Hassell L, Mays A, Pantanowitz L, Sirintrapun JS, Krishnamurthy S, Parwani A, Lujan G, Evans A, Glassy EF, Bui MM, Singh R, Souers RJ, de Baca ME, Seheult JN. Recommendations for Performance Evaluation of Machine Learning in Pathology: A Concept Paper From the College of American Pathologists. Arch Pathol Lab Med 2024; 148:e335-e361. [PMID: 38041522 DOI: 10.5858/arpa.2023-0042-cp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 12/03/2023]
Abstract
CONTEXT.— Machine learning applications in the pathology clinical domain are emerging rapidly. As decision support systems continue to mature, laboratories will increasingly need guidance to evaluate their performance in clinical practice. Currently there are no formal guidelines to assist pathology laboratories in verification and/or validation of such systems. These recommendations are being proposed for the evaluation of machine learning systems in the clinical practice of pathology. OBJECTIVE.— To propose recommendations for performance evaluation of in vitro diagnostic tests on patient samples that incorporate machine learning as part of the preanalytical, analytical, or postanalytical phases of the laboratory workflow. Topics described include considerations for machine learning model evaluation including risk assessment, predeployment requirements, data sourcing and curation, verification and validation, change control management, human-computer interaction, practitioner training, and competency evaluation. DATA SOURCES.— An expert panel performed a review of the literature, Clinical and Laboratory Standards Institute guidance, and laboratory and government regulatory frameworks. CONCLUSIONS.— Review of the literature and existing documents enabled the development of proposed recommendations. This white paper pertains to performance evaluation of machine learning systems intended to be implemented for clinical patient testing. Further studies with real-world clinical data are encouraged to support these proposed recommendations. Performance evaluation of machine learning models is critical to verification and/or validation of in vitro diagnostic tests using machine learning intended for clinical practice.
Collapse
Affiliation(s)
- Matthew G Hanna
- From the Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York (Hanna, Sirintrapun)
| | - Niels H Olson
- The Defense Innovation Unit, Mountain View, California (Olson)
- The Department of Pathology, Uniformed Services University, Bethesda, Maryland (Olson)
| | - Mark Zarella
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Zarella, Seheult)
| | - Rajesh C Dash
- Department of Pathology, Duke University Health System, Durham, North Carolina (Dash)
| | - Markus D Herrmann
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston (Herrmann)
| | - Larissa V Furtado
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee (Furtado)
| | - Michelle N Stram
- The Department of Forensic Medicine, New York University, and Office of Chief Medical Examiner, New York (Stram)
| | | | - Lewis Hassell
- Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City (Hassell)
| | - Alex Mays
- The MITRE Corporation, McLean, Virginia (Mays)
| | - Liron Pantanowitz
- Department of Pathology & Clinical Labs, University of Michigan, Ann Arbor (Pantanowitz)
| | - Joseph S Sirintrapun
- From the Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York (Hanna, Sirintrapun)
| | | | - Anil Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus (Parwani, Lujan)
| | - Giovanni Lujan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus (Parwani, Lujan)
| | - Andrew Evans
- Laboratory Medicine, Mackenzie Health, Toronto, Ontario, Canada (Evans)
| | - Eric F Glassy
- Affiliated Pathologists Medical Group, Rancho Dominguez, California (Glassy)
| | - Marilyn M Bui
- Departments of Pathology and Machine Learning, Moffitt Cancer Center, Tampa, Florida (Bui)
| | - Rajendra Singh
- Department of Dermatopathology, Summit Health, Summit Woodland Park, New Jersey (Singh)
| | - Rhona J Souers
- Department of Biostatistics, College of American Pathologists, Northfield, Illinois (Souers)
| | | | - Jansen N Seheult
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Zarella, Seheult)
| |
Collapse
|
82
|
Hsu E, Arezo SM, Graff SL. Updates in Systemic Treatment of Hormone Receptor-Positive Early-Stage Breast Cancer. Curr Treat Options Oncol 2024; 25:1323-1334. [PMID: 39361142 DOI: 10.1007/s11864-024-01258-5] [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] [Accepted: 08/21/2024] [Indexed: 10/17/2024]
Abstract
OPINION STATEMENT Hormone-receptor positive (HR +) and human epidermal growth factor receptor 2 (HER2) negative early breast cancer (eBC) is a heterogeneous disease with several contributing factors for increased risk of recurrence, including tumor features, individual biomarkers, and genomic risk. The current standard approach in the management of HR + /HER2neg eBC includes chemotherapy and endocrine therapy (ET), and additional therapies based on risk profile, menopausal status, and genetics are sometimes appropriate. The risk of recurrence is more pronounced in patients with high-risk eBC including large tumor size, nodal involvement, high proliferative index, and genetic predisposition. In premenopausal patients with high-risk eBC, ovarian function suppression in combination with adjuvant ET improves survival. In postmenopausal patients, extended aromatase inhibitor (AI) therapy can be considered. Recent trials have identified novel treatment approaches to reduce the risk of recurrence in high-risk HR + /HER2neg eBC including the addition of cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors to adjuvant ET. For patients with germline BRCA1/BRCA2 mutations, adjuvant poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors have been shown to improve overall survival (OS). However, despite these recent advances, the risk of recurrence remains substantial, highlighting an area of unmet need. There are several ongoing clinical trials further investigating the role of CDK 4/6 inhibitors and immunotherapy in high-risk HR + /HER2neg eBC.
Collapse
Affiliation(s)
- Emily Hsu
- Legorreta Cancer Center at Brown University, Providence, RI, USA
- Lifespan Cancer Institute, Providence, RI, USA
| | - Sabrina M Arezo
- Warren Alpert School of Medicine, Brown University, Providence, RI, USA
| | - Stephanie L Graff
- Legorreta Cancer Center at Brown University, Providence, RI, USA.
- Lifespan Cancer Institute, Providence, RI, USA.
| |
Collapse
|
83
|
Elghazaly H, Azim HA, Rugo HS, Cameron D, Swain SM, Curigliano G, Harbeck N, Tripathy D, Arun B, Aapro M, Piccart M, Cardoso F, Gligorov J, Elghazawy H, El Saghir NS, Penault-Llorca F, Perez EA, Poortmans P, Abdelaziz H, El-Zawahry HM, Kassem L, Sabry M, Viale G, Al-Sukhun S, Gado N, Leung JWT, Ezz Elarab L, Cardoso MJ, Abdel Karim K, Foheidi M, Elmaadawy MM, Conte P, Selim ASM, Kandil A, Kamal RM, Paltuev RM, Guarneri V, Abulkhair O, Zakaria O, Golshan M, Orecchia R, ElMahdy M, Abdel-Aziz AM, Eldin NB. Tailoring neoadjuvant systemic therapy in breast cancer: "The advent of a personalized approach"-The Breast-Gynecological and Immuno-Oncology International Cancer Conference (BGICC) consensus and recommendations. Cancer 2024; 130:3251-3271. [PMID: 38985794 DOI: 10.1002/cncr.35389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The management of early breast cancer (BC) has witnessed an uprise in the use of neoadjuvant therapy and a remarkable reshaping of the systemic therapy postneoadjuvant treatment in the last few years, with the evolution of many controversial clinical situations that require consensus. METHODS During the 14th Breast-Gynecological and Immuno-Oncology International Cancer Conference held in Egypt in 2022, a panel of 44 BC experts from 13 countries voted on statements concerning debatable challenges in the neo/adjuvant treatment setting. The recommendations were subsequently updated based on the most recent data emerging. A modified Delphi approach was used to develop this consensus. A consensus was achieved when ≥75% of voters selected an answer. RESULTS AND CONCLUSIONS The consensus recommendations addressed different escalation and de-escalation strategies in the setting of neoadjuvant therapy for early BC. The recommendations recapitulate the available clinical evidence and expert opinion to individualize patient management and optimize therapy outcomes. Consensus was reached in 63% of the statements (52/83), and the rationale behind each statement was clarified.
Collapse
Affiliation(s)
- Hesham Elghazaly
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hamdy A Azim
- Department of Clinical Oncology, Kasr Alainy School of Medicine, Cairo University, Giza, Egypt
| | - Hope S Rugo
- Department of Medicine, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - David Cameron
- Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh and National Health Service Lothian, Edinburgh, UK
| | - Sandra M Swain
- Georgetown Lombardi Comprehensive Cancer Center, MedStar Health, Washington, District of Columbia, USA
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, European Institute of Oncology, IRCCS, University of Milano, Milan, Italy
| | - Nadia Harbeck
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center Munich, Breast Center, Ludwig Maximilian University Hospital, Munich, Germany
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Matti Aapro
- Breast Center, Clinique de Genolier, Genolier, Switzerland
| | - Martine Piccart
- Institut Jules Bordet and L'Université Libre de Bruxelles, Brussels, Belgium
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal
| | - Joseph Gligorov
- Medical Oncology Department, L'Assistance Publique-Hôpitaux de Paris, Institute Universitaire de Cancérologie, Sorbonne Université, Paris, France
| | - Hagar Elghazawy
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nagi S El Saghir
- Division of Hematology Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Frederique Penault-Llorca
- National Institute of Health and Medical Research Unit 1240 "Molecular Imaging and Theranostic Strategies", Department of Pathology, Clermont Auvergne University, Center Jean Perrin, Clermont-Ferrand, France
| | - Edith A Perez
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Philip Poortmans
- Iridium Network and Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk-Antwerp, Belgium
| | - Hany Abdelaziz
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Heba M El-Zawahry
- Department of Clinical Oncology, Kasr Alainy School of Medicine, Cairo University, Giza, Egypt
| | - Loay Kassem
- Department of Clinical Oncology, Kasr Alainy School of Medicine, Cairo University, Giza, Egypt
| | - Mohamed Sabry
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Neven Gado
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Jessica W T Leung
- Department of Breast Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lobna Ezz Elarab
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maria João Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Faculdade de Medicina, Lisbon, Portugal
| | - Khaled Abdel Karim
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Meteb Foheidi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Adult Medical Oncology, Princess Noorah Oncology Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs-Western Region, Jeddah, Saudi Arabia
| | - Merit M Elmaadawy
- Diagnostic Radiology Department, Mansoura University, Mansoura, Egypt
| | - Pierfranco Conte
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, Italy
- Division of Oncology, Istituto Oncologico Veneto-IRCCS, Padova, Italy
| | - Ashraf S M Selim
- Diagnostic and Interventional Radiology Department, Cairo University, Giza, Egypt
| | - Alaa Kandil
- Department of Clinical Oncology, Alexandria School of Medicine, Alexandria, Egypt
| | - Rasha M Kamal
- Diagnostic and Interventional Radiology Department, Cairo University, Giza, Egypt
| | - Ruslan M Paltuev
- Department of Breast Tumours of Federal State Budgetary Institution "Petrov Research Institute of Oncology", Russian Association of Oncological Mammology, St Petersburg, Russia
| | - Valentina Guarneri
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, Italy
- Division of Oncology, Istituto Oncologico Veneto-IRCCS, Padova, Italy
| | - Omalkhair Abulkhair
- Medical Oncology Department, Oncology Services, Alhabib Hospital, Riyad, Saudi Arabia
| | - Omar Zakaria
- Department of Surgery, Cairo University, Cairo, Egypt
| | - Mehra Golshan
- Department of Surgery, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Roberto Orecchia
- Scientific Directorate, IRCCS European Institute of Oncology, University of Milan, Milan, Italy
| | - Manal ElMahdy
- Department of Pathology, Ain Shams University, Cairo, Egypt
| | - Ahmed M Abdel-Aziz
- Department of Pathology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nermean Bahie Eldin
- Department of Clinical Oncology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
84
|
Keerthana Devi D, Pavithra V, Joseph LD, Challa CB. Correlation of Androgen Receptor Expression With Ki67 Proliferative Index and Other Clinicopathological Characteristics in Invasive Mammary Carcinomas. Cureus 2024; 16:e70867. [PMID: 39497884 PMCID: PMC11534436 DOI: 10.7759/cureus.70867] [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] [Accepted: 10/04/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction The clinical importance of androgen receptor (AR) status in breast cancer is uncertain. The existing knowledge regarding the association of androgen receptor expression with clinicopathological factors of breast cancer is also limited. The main aim of this study is to evaluate the AR expression in breast cancer and to correlate it with the Ki67 proliferative index and other clinicopathological prognostic factors. Methods The expression of AR was evaluated in 192 invasive mammary carcinoma cases and the expression patterns were correlated with various clinicopathological prognostic factors such as age, tumor size, pathological primary tumor (pT) stage, nodal status, histological grade, estrogen receptor (ER) expression, progesterone receptor (PgR) expression, human epidermal growth factor receptor 2 (Her2) status, molecular subtype, and Ki67 labeling index. Immunohistochemistry was performed to assess AR, ER, PgR, Her2, and Ki67 expression. The clinicopathological data required for the analysis were obtained from the laboratory information system. Results Out of the 192 breast carcinoma cases, 139 (72.4%) showed AR-positive expression. The average age of the AR-positive cases was slightly higher than the AR-negative cases. AR-positive tumors tended to have a lower histological grade and positive ER and PgR expression. The expression of AR did not correlate with tumor size, pT stage, nodal status, Her2 status, and Ki67 labeling index. Conclusion The expression of AR is noted in a significant proportion of breast carcinoma cases. AR expression may be related to good prognostic factors such as ER expression, PgR expression, and lower histologic grade. We also observed that AR expression did not have any association with the Ki67 proliferative index.
Collapse
Affiliation(s)
- D Keerthana Devi
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - V Pavithra
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Leena D Joseph
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Chithra Bhanu Challa
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| |
Collapse
|
85
|
Rassy E, Mosele MF, Di Meglio A, Pistilli B, Andre F. Precision oncology in patients with breast cancer: towards a 'screen and characterize' approach. ESMO Open 2024; 9:103716. [PMID: 39303452 PMCID: PMC11439525 DOI: 10.1016/j.esmoop.2024.103716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- E Rassy
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Oncostat U1018, Inserm, Université Paris-Saclay, Equipe labellisée Ligue Contre le Cancer, Villejuif
| | - M F Mosele
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif
| | - A Di Meglio
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif
| | - B Pistilli
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; INSERM U1279, Gustave Roussy, Villejuif, France
| | - F Andre
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif.
| |
Collapse
|
86
|
Bernhardt M, Weinhold L, Sanders C, Hommerding O, Lau JF, Toma M, Tischler V, Schmid M, Zienkiewicz T, Hildenbrand R, Gerlach P, Zhou H, Braun M, Müller G, Sieber E, Marko C, Kristiansen G. Peer-to-peer validation of Ki-67 scoring in a pathology quality circle as a tool to assess interobserver variability: are we better than we thought? APMIS 2024; 132:718-727. [PMID: 38951722 DOI: 10.1111/apm.13451] [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: 02/27/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
Ki-67, a nuclear protein expressed in all stages of cellular proliferation, is a valuable tool to assess tumor proliferation and has been linked to more aggressive tumor behavior. However, interlaboratory staining heterogeneity and inter-observer variability challenge its reproducibility. Round Robin tests are a suitable tool to standardize and harmonize immunohistochemical and molecular analyses in histopathology. The study investigates the interrater and interlaboratory reproducibility of Ki-67-scoring using both manual and automated approaches. Unstained TMA slides comprising diverse tumor types (breast cancer, neuroendocrine tumors, lymphomas, and head and neck squamous cell carcinoma) were distributed to six pathology laboratories, each employing their routine staining protocols. Manual and automated scoring methods were applied, and interrater and interlaboratory agreement assessed using intraclass correlation coefficients (ICC). The results highlight good-to-excellent reliability overall, with automated scoring demonstrating higher consistency (ICC 0.955) than manual scoring (ICC 0.871). Results were more variable when looking at the individual entities. Reliability remained good for lymphomas (ICC 0.878) and breast cancer (ICC 0.784) and was poor in well-differentiated neuroendocrine tumors (ICC 0.354). This study clearly advocates standardized practices and training to ensure consistency in Ki-67-assessment, and it demonstrates that this can be achieved in a peer-to-peer approach in local quality-circles.
Collapse
Affiliation(s)
- Marit Bernhardt
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Leonie Weinhold
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | | | | | | | - Marieta Toma
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Verena Tischler
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Matthias Schmid
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | | | | | | | - Hui Zhou
- Pathologie24 Pathology Practice Bonn City Centre, Bonn, Germany
| | - Martin Braun
- Institute of Pathology and Cytology, Rhein-Sieg, Troisdorf, Germany
| | - Gunnar Müller
- Department of Pathology, Federal Armed Forces Hospital, Koblenz, Germany
| | - Erich Sieber
- Department of Pathology, Federal Armed Forces Hospital, Koblenz, Germany
| | | | | |
Collapse
|
87
|
Tau S, Chamberlin MD, Yang H, Marotti JD, Roberts AM, Carmichael MM, Cressey L, Dragnev C, Demidenko E, Hampsch RA, Soucy SM, Kolling F, Samkoe KS, Alvarez JV, Kettenbach AN, Miller TW. Endocrine persistence in ER+ breast cancer is accompanied by metabolic vulnerability in oxidative phosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615177. [PMID: 39386444 PMCID: PMC11463551 DOI: 10.1101/2024.09.26.615177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Despite adjuvant treatment with endocrine therapies, estrogen receptor-positive (ER+) breast cancers recur in a significant proportion of patients. Recurrences are attributable to clinically undetectable endocrine-tolerant persister cancer cells that retain tumor-forming potential. Therefore, strategies targeting such persister cells may prevent recurrent disease. Using CRISPR-Cas9 genome-wide knockout screening in ER+ breast cancer cells, we identified a survival mechanism involving metabolic reprogramming with reliance upon mitochondrial respiration in endocrine-tolerant persister cells. Quantitative proteomic profiling showed reduced levels of glycolytic proteins in persisters. Metabolic tracing of glucose revealed an energy-depleted state in persisters where oxidative phosphorylation was required to generate ATP. A phase II clinical trial was conducted to evaluate changes in mitochondrial markers in primary ER+/HER2-breast tumors induced by neoadjuvant endocrine therapy ( NCT04568616 ). In an analysis of tumor specimens from 32 patients, tumors exhibiting residual cell proliferation after aromatase inhibitor-induced estrogen deprivation with letrozole showed increased mitochondrial content. Genetic profiling and barcode lineage tracing showed that endocrine-tolerant persistence occurred stochastically without genetic predisposition. Mice bearing cell line- and patient-derived xenografts were used to measure the anti-tumor effects of mitochondrial complex I inhibition in the context of endocrine therapy. Pharmacological inhibition of complex I suppressed the tumor-forming potential of persisters and synergized with the anti-estrogen fulvestrant to induce regression of patient-derived xenografts. These findings indicate that mitochondrial metabolism is essential in endocrine-tolerant persister ER+ breast cancer cells and warrant the development of treatment strategies to leverage this vulnerability in the context of endocrine-sensitive disease. Statement of Significance Endocrine-tolerant persister cancer cells that survive endocrine therapy can cause recurrent disease. Persister cells exhibit increased energetic dependence upon mitochondria for survival and tumor re-growth potential.
Collapse
|
88
|
Takeda M, Ito H, Kitahata K, Sato S, Nishide A, Gamo K, Managi S, Tezuka T, Yoshizawa A, Kim M. α-Parvin Expression in Breast Cancer Tissues: Correlation with Clinical Parameters and Prognostic Significance. Cells 2024; 13:1572. [PMID: 39329755 PMCID: PMC11430769 DOI: 10.3390/cells13181572] [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: 08/13/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Stromal cells play a critical role in the tumor microenvironment of breast cancer (BC), as they are recruited by tumor cells and regulate the metastatic spread. Though high expression of α-parvin, a member of the parvin family of actin-binding proteins, is reported to be associated with a poor prognosis and metastasis in several cancers, its role in carcinogenesis has not been thoroughly explored. Therefore, we aimed to examine the expression of α-parvin in BC patients by compartmentalizing and quantifying tissues to determine whether α-parvin can be a potential therapeutic target. We performed immunohistochemical (IHC) staining of α-parvin in BC tissues, and the IHC scores were calculated in the overall tissue, stroma, and epithelium using image analysis software. The expression of α-parvin was significantly higher in BC tissues (p = 0.0002) and BC stroma (p < 0.0001) than in normal tissues. Furthermore, all α-parvin scores were significantly positively correlated with the proliferation marker Ki67. The overall and stroma scores are associated with the tumor, (lymph) node, and metastasis (TNM) classification, stage, and grade. These results suggest that high expression of α-parvin in stroma is associated with BCs and might be a new predictive marker for diagnosing BC.
Collapse
Affiliation(s)
- Midori Takeda
- Laboratory of Integrative Molecular Medicine, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto-shi 606-8501, Kyoto, Japan; (M.T.)
- Urban Institute & Department of Civil Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka-shi 819-0395, Fukuoka, Japan
| | - Hiroaki Ito
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto-shi 606-8507, Kyoto, Japan
| | - Keisuke Kitahata
- Laboratory of Integrative Molecular Medicine, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto-shi 606-8501, Kyoto, Japan; (M.T.)
| | - Sota Sato
- Laboratory of Integrative Molecular Medicine, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto-shi 606-8501, Kyoto, Japan; (M.T.)
| | - Akira Nishide
- Laboratory of Integrative Molecular Medicine, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto-shi 606-8501, Kyoto, Japan; (M.T.)
| | - Kanae Gamo
- FIMECS, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa-shi 251-0012, Kanagawa, Japan
| | - Shunsuke Managi
- Urban Institute & Department of Civil Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka-shi 819-0395, Fukuoka, Japan
| | - Tohru Tezuka
- Laboratory of Integrative Molecular Medicine, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto-shi 606-8501, Kyoto, Japan; (M.T.)
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo-cho, Kashihara-shi 634-8521, Nara, Japan
| | - Minsoo Kim
- Laboratory of Integrative Molecular Medicine, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto-shi 606-8501, Kyoto, Japan; (M.T.)
| |
Collapse
|
89
|
Huang X, Wu L, Liu Y, Xu Z, Liu C, Liu Z, Liang C. Development and validation of machine learning models for predicting HER2-zero and HER2-low breast cancers. Br J Radiol 2024; 97:1568-1576. [PMID: 38991838 PMCID: PMC11332671 DOI: 10.1093/bjr/tqae124] [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: 01/15/2024] [Revised: 02/03/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVES To develop and validate machine learning models for human epidermal growth factor receptor 2 (HER2)-zero and HER2-low using MRI features pre-neoadjuvant therapy (NAT). METHODS Five hundred and sixteen breast cancer patients post-NAT surgery were randomly divided into training (n = 362) and internal validation sets (n = 154) for model building and evaluation. MRI features (tumour diameter, enhancement type, background parenchymal enhancement, enhancement pattern, percentage of enhancement, signal enhancement ratio, breast oedema, and apparent diffusion coefficient) were reviewed. Logistic regression (LR), support vector machine (SVM), k-nearest neighbour (KNN), and extreme gradient boosting (XGBoost) models utilized MRI characteristics for HER2 status assessment in training and validation datasets. The best-performing model generated a HER2 score, which was subsequently correlated with pathological complete response (pCR) and disease-free survival (DFS). RESULTS The XGBoost model outperformed LR, SVM, and KNN, achieving an area under the receiver operating characteristic curve (AUC) of 0.783 (95% CI, 0.733-0.833) and 0.787 (95% CI, 0.709-0.865) in the validation dataset. Its HER2 score for predicting pCR had an AUC of 0.708 in the training datasets and 0.695 in the validation dataset. Additionally, the low HER2 score was significantly associated with shorter DFS in the validation dataset (hazard ratio: 2.748, 95% CI, 1.016-7.432, P = .037). CONCLUSIONS The XGBoost model could help distinguish HER2-zero and HER2-low breast cancers and has the potential to predict pCR and prognosis in breast cancer patients undergoing NAT. ADVANCES IN KNOWLEDGE HER2-low-expressing breast cancer can benefit from the HER2-targeted therapy. Prediction of HER2-low expression is crucial for appropriate management. MRI features offer a solution to this clinical issue.
Collapse
Affiliation(s)
- Xu Huang
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Lei Wu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Yu Liu
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
- Department of Ultrasound, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zeyan Xu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Chunling Liu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou 510080, China
| |
Collapse
|
90
|
Jackisch C, Anastasiadou L, Aulmann S, Argyriadis A, Möbus V, Solbach C, Baier P, Giesecke D, Ackermann S, Schulmeyer E, Gabriel B, Mosch D, Buchen S, Krapfl E, Hurst U, Vescia M, Tesch H, Thill M. The REMAR (Rhein-Main-Registry) real-world study: prospective evaluation of the 21-gene breast recurrence score® assay in addition to Ki-67 for adjuvant treatment decisions in early-stage breast cancer. Breast Cancer Res Treat 2024; 207:263-274. [PMID: 38874685 PMCID: PMC11297120 DOI: 10.1007/s10549-024-07390-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE Ki-67 is recommended by international/national guidelines for risk stratification in early breast cancer (EBC), particularly for defining "intermediate risk," despite inter-laboratory/inter-observer variability and cutoff uncertainty. We investigated Ki-67 (> 10%- < 40%, determined locally) as a prognostic marker for intermediate/high risk in EBC, pN0-1 patients. METHODS This prospective, non-interventional, real-world study included females ≥ 18 years, with pN0/pN1mi/pN1, HR+ , HER2-negative EBC, and locally determined Ki-67 ranging 10%-40%. The primary outcome was changes in treatment recommendations after disclosing the Oncotype DX Breast Recurrence Score®(RS) assay result. RESULTS The analysis included 567 patients (median age, 57 [range, 29-83] years; 70%/1%/29%/ with pN0/pN1mi/pN1 disease; 81% and 19% with RS results 0-25 and 26-100, respectively). The correlations between local and central Ki-67, local Ki-67, and the RS, and central Ki-67 and the RS results were weak (r = 0.35, r = 0.3, and r = 0.46, respectively), and discrepancies were noted in both directions (e.g., local Ki-67 was lower or higher than central Ki-67). After disclosing the RS, treatment recommendations changed for 190 patients (34%). Changes were observed in pN0 and pN1mi/pN1 patients and in patients with centrally determined Ki-67 ≤ 10% and > 10%. Treatment changes were aligned with RS results (adding chemotherapy for patients with higher RS results, omitting it for lower RS results), and their net result was 8% reduction in adjuvant chemotherapy use (from 32% pre-RS results to 24% post-RS results). CONCLUSION The Oncotype DX® assay is a tool for individualizing treatments that adds to classic treatment decision factors. The RS result and Ki-67 are not interchangeable, and Ki-67, as well as nodal status, should not be used as gatekeepers for testing eligibility, to avoid under and overtreatment.
Collapse
Affiliation(s)
- Christian Jackisch
- Department of Gynecology and Obstetrics, Sana Klinikum Offenbach GmbH, Offenbach, Germany.
- OncoNet Rhein Main e. v., Frankfurt, Germany.
- KEM, Evang. Kliniken Essen-Mitte gGmbH, Henricistr. 92, 45136, Essen, Germany.
| | - Louiza Anastasiadou
- Department of Palliative Medicine, Agaplesion Markus Hospital, Frankfurt, Germany
| | | | - Athanasios Argyriadis
- Department of Gynecology and Obstetrics, Sana Klinikum Offenbach GmbH, Offenbach, Germany
| | - Volker Möbus
- OncoNet Rhein Main e. v., Frankfurt, Germany
- Department of Gynecology and Obstetrics, Städtische Kliniken Frankfurt Hoechst, Frankfurt, Germany
| | - Christine Solbach
- OncoNet Rhein Main e. v., Frankfurt, Germany
- Department of Gynecology and Obstetrics, Universitaetsklinikum Frankfurt, Frankfurt, Germany
| | - Peter Baier
- Department of Gynecology and Obstetrics, Ketteler Krankenhaus Offenbach, Offenbach, Germany
| | - Dagmar Giesecke
- Department of Gynecology and Obstetrics, Hochtaunus Kliniken, Bad Homburg, Germany
| | - Sven Ackermann
- Department of Gynecology and Obstetrics, Städtische Kliniken Darmstadt, Darmstadt, Germany
| | - Elke Schulmeyer
- Department of Gynecology and Obstetrics, Main Kinzig Kliniken, Gelnhausen, Germany
| | - Boris Gabriel
- Department of Gynecology and Obstetrics, St. Josefs Hospital, Wiesbaden, Germany
| | - Dietrich Mosch
- Department of Gynecology and Obstetrics, Varisano Kliniken Frankfurt-Main Taunus, Bad Soden I.T., Germany
| | - Stephanie Buchen
- OncoNet Rhein Main e. v., Frankfurt, Germany
- Department of Obsetrics and Gynecology, Agaplesion Kliniken Wiesbaden, Wiesbaden, Germany
| | - Eckart Krapfl
- OncoNet Rhein Main e. v., Frankfurt, Germany
- Department of Obsterics and Gynecology, Agaplesion Klliniken Langen, Langen, Germany
| | - Ursula Hurst
- Department of Gynecology and Obstetrics, Kreiskrankenhaus Bergstrasse, Heppenheim, Germany
| | - Mario Vescia
- Department of Obsetrics and Gynecology, GPR Klinikum Ruesselsheim, Rüsselsheim, Germany
| | - Hans Tesch
- OncoNet Rhein Main e. v., Frankfurt, Germany
- Center for Oncology and Hematology, Onkologie Bethanien, Frankfurt, Germany
| | - Marc Thill
- OncoNet Rhein Main e. v., Frankfurt, Germany
- Department of Gynecology and Gynecological Oncology, Agaplesion Markus Hospital, Frankfurt, Germany
| |
Collapse
|
91
|
Badr NM, Zaakouk M, Zhang Q, Kearns D, Kong A, Shaaban AM. Concordance between ER, PR, Ki67, and HER2-low expression in breast cancer by MammaTyper RT-qPCR and immunohistochemistry: implications for the practising pathologist. Histopathology 2024; 85:437-450. [PMID: 38651302 DOI: 10.1111/his.15193] [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: 11/25/2023] [Revised: 03/21/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND There are limited data on the role of multigene tests and their correlation with immunohistochemistry (IHC), especially on core biopsy. MammaTyper is a quantitative conformite Europeeanne (CE) marked, National Institute for Health and Care excellence (NICE) approved, in in vitro diagnostic quantitative real-time polymerase chain reaction (RT-qPCR) test for assessment of mRNA expression of four biomarkers (ESR1, PGR, ERBB2, MKI67). METHODS We evaluated the concordance of MammaTyper with oestrogen receptor (ER), progesterone receptor (PR), HER2, and Ki67 by IHC on 133 core needle biopsies of breast cancer. HER2 was positive if IHC 3+ or 2+ and fluorescence in situ hybridization (FISH)-amplified. Global and hotspot Ki67 expression was analysed using a cutoff of ≥20% assessed manually and by digital image analysis. Agreements were expressed as overall percent agreement (OPA), positive percent agreement (PPA), negative percent agreement (NPA), and Cohen's kappa. RESULTS RT-qPCR results of ESR1 were highly concordant with IHC with OPA of 94.7% using 1% cutoff and 91.7% when the low ER-positive category was included. The PPA and NPA between RT-qPCR and IHC for PR was 91.5% and 88.0%, respectively, when using the 1% cutoff. For ERBB2/HER2, the OPA was 95% and the PPA was 84.6%. 40 of 72 HER2 IHC score 0 tumours were classified as ERBB2 low. Best concordance between MKI67 by MammaTyper and Ki67 IHC was achieved using hotspot digital image analysis (OPA: 87.2%, PPA: 90.6%, NPA: 80%). CONCLUSION RT-qPCR-based assessment of the mRNA expression of ESR1, PGR, ERBB2, and MKI67 showed high concordance with IHC, suggesting that the MammaTyper test on core needle biopsies represents a reliable, efficient, and reproducible alternative for breast cancer classification and refining HER2 low categorisation.
Collapse
Affiliation(s)
- Nahla M Badr
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Mohamed Zaakouk
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Cancer Pathology Department, National Cancer Institute, Cairo University, Giza, Egypt
| | - Qi Zhang
- Shuwen Biotech Co. Ltd., Hangzhou, Zhejiang Province, China
| | | | - Anthony Kong
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- King's College London, London, UK
| | - Abeer M Shaaban
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Queen Elizabeth Hospital Birmingham, Birmingham, UK
| |
Collapse
|
92
|
Ribeiro R, Carvalho FM, Baiocchi G, Guindalini RSC, da Cunha JR, Anjos CHD, de Nadai Costa C, Gifoni ACLVC, Neto RC, Cagnacci AQC, Carneiro VCG, Calabrich A, Moretti-Marques R, Pinheiro RN, de Castro Ribeiro HS. Guidelines of the Brazilian Society of Surgical Oncology for anatomopathological, immunohistochemical, and molecular testing in female tumors. J Surg Oncol 2024; 130:882-895. [PMID: 39038206 DOI: 10.1002/jso.27717] [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: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Precision medicine has revolutionized oncology, providing more personalized diagnosis, treatment, and monitoring for patients with cancer. In the context of female-specific tumors, such as breast, ovarian, endometrial, and cervical cancer, proper tissue collection and handling are essential for obtaining tissue, immunohistochemical (IHC), and molecular data to guide therapeutic decisions. OBJECTIVES To establish guidelines for the collection and handling of tumor tissue, to enhance the quality of samples for histopathological, IHC, genomic, and molecular analyses. These guidelines are fundamental in informing therapeutic decisions in cancer treatment. METHOD The guidelines were developed by a multidisciplinary panel of renowned specialists between June 12, 2013 and February 12, 2024. Initially, the panel deliberated on critical and controversial topics related to conducting precision medicine studies focusing on female tumors. Subsequently, 22 pivotal topics were identified within the framework and assigned to groups. These groups reviewed relevant literature and drafted preliminary recommendations. Following this, the recommendations were reviewed by the coordinators and received unanimous approval. Finally, the groups made the final adjustments, classified the level of evidence, and ranked the recommendations. CONCLUSION The collection of surgical samples requires minimum quality standards to enable histopathological, IHC, genomic, and molecular analyses. These analyses provide crucial data for informing therapeutic decisions, significantly impacting potential survival gains for patients with female tumors.
Collapse
Affiliation(s)
- Reitan Ribeiro
- Department of Gynecology Oncology, Erasto Gaertner Hospital, Curitiba, Paraná, Brazil
| | - Filomena Marino Carvalho
- Department of Pathology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Glauco Baiocchi
- Department of Gynecologic Oncology, AC Camargo Cancer Center , São Paulo, São Paulo, Brazil
| | | | | | | | | | | | - Renato Cagnacci Neto
- Department of Mastology, Breast Cancer Reference Center, AC Camargo Cancer, CenterSão Paulo, São Paulo, Brazil
| | - Allyne Queiroz Carneiro Cagnacci
- Department of Oncology, Oncology Center, Hospital Alemão Oswaldo Cruz, São Paulo, São Paulo, Brazil
- Hereditary Cancer Department, Instituto do Câncer do Estado de São Paulo (ICESPSP), São Paulo, São Paulo, Brazil
| | - Vandré Cabral Gomes Carneiro
- Department of Gynecology Oncology, Instituto de Medicina Integral Professor Fernando Figueira (IMIP), Recife, Pernambuco, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Oncogenetic, Oncologia D'OR, Recife, Pernambuco, Brazil
| | - Aknar Calabrich
- Department of Oncology, Clínica AMO/DASA, Salvador, Bahia, Brazil
| | - Renato Moretti-Marques
- Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
93
|
Lashen AG, Toss M, Miligy I, Rewcastle E, Kiraz U, Janssen EAM, Green AR, Quinn C, Ellis I, Rakha EA. Nottingham prognostic x (NPx): a risk stratification tool in ER-positive HER2-negative breast cancer: a validation study. Histopathology 2024; 85:468-477. [PMID: 38867570 DOI: 10.1111/his.15234] [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: 12/28/2023] [Revised: 04/21/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024]
Abstract
AIMS In this study, we validate the use of Nottingham Prognostic x (NPx), consisting of tumour size, tumour grade, progesterone receptor (PR) and Ki67 in luminal BC. MATERIALS AND METHODS Two large cohorts of luminal early-stage BC (n = 2864) were included. PR and Ki67 expression were assessed using full-face resection samples using immunohistochemistry. NPx was calculated and correlated with clinical variables and outcome, together with Oncotype DX recurrence score (RS), that is frequently used as a risk stratifier in luminal BC. RESULTS In the whole cohort, 38% of patients were classified as high risk using NPx which showed significant association with parameters characteristics of aggressive tumour behaviour and shorter survival (P < 0.0001). NPx classified the moderate Nottingham Prognostic Index (NPI) risk group (n = 1812) into two distinct prognostic subgroups. Of the 82% low-risk group, only 3.8% developed events. Contrasting this, 14% of the high-risk patients developed events during follow-up. A strong association was observed between NPx and Oncotype Dx RS (P < 0.0001), where 66% of patients with intermediate risk RS who had subsequent distant metastases also had a high-risk NPx. CONCLUSION NPx is a reliable prognostic index in patients with luminal early-stage BC, and in selected patients may be used to guide adjuvant chemotherapy recommendations.
Collapse
Affiliation(s)
- Ayat G Lashen
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham, UK
| | - Michael Toss
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Histopathology, Sheffield Teaching Hospitals NHS Foundation Trust Sheffield, Sheffield, UK
| | - Islam Miligy
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| | - Emma Rewcastle
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, Stavanger University, Stavanger, Norway
| | - Umay Kiraz
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, Stavanger University, Stavanger, Norway
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, Stavanger University, Stavanger, Norway
- Menzies Health Institute Queensland and Griffith University, Gold Coast, Queensland, Australia
| | - Andrew R Green
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham, UK
| | - Cecily Quinn
- Department of Pathology, Vincent's University Hospital, Dublin, Ireland
| | - Ian Ellis
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Pathology, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
94
|
Miligy IM, Awasthi R, Mir Y, Khurana A, Sharma V, Chandaran U, Rakha E, Maurice Y, Kearns D, Oweis R, Asar A, Ironside A, Shaaban AM. Morphological and molecular changes of oestrogen receptor-positive breast cancer following bridging endocrine therapy: a United Kingdom multicentre study. Histopathology 2024; 85:405-417. [PMID: 38845397 DOI: 10.1111/his.15238] [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: 01/15/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 08/09/2024]
Abstract
AIMS Standard neoadjuvant endocrine therapy (NAET) is used for 6-9 months to downstage hormone-receptor-positive breast cancer. Bridging ET was introduced during the COVID-19 pandemic to delay surgical intervention. There are no data in the literature on the effect of short course therapy on tumour response. We aimed to analyse the effect of bridging ET and validate the previously proposed neoadjuvant ET pathological reporting criteria. METHODS AND RESULTS This was a multicentre cohort of 256 patients who received bridging ET between March and October 2020. Assessment of paired pre- and post-NAET hormone receptors and HER2 and posttherapy Ki67 expression was done. The median duration of NAET was 45 days. In all, 86% of cases achieved partial pathological response and 9% showed minimal residual disease. Histological response to ET was observed from as early as day 6 posttherapy. Central scarring was noted in 32.8% of cases and lymphocytic infiltrate was seen in 43.4% of cases. Significant changes associated with the duration of ET were observed in tumour grade (21%), with downgrading identified in 12% of tumours (P < 0.001), progesterone receptor (PR) expression with switch to PR-negative status in 26% of cases (P < 0.001), and HER2 status with a switch from HER2-low to HER2-negative status in 32% of cases (P < 0.001). The median patient survival was 475 days, with an overall survival rate of 99.6%. CONCLUSIONS Changes characteristic of tumour regression and significant changes in PR and HER2 occurred following a short course of NAET. The findings support biomarker testing on pretreatment core biopsies and retesting following therapy.
Collapse
Affiliation(s)
- Islam M Miligy
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
- Histopathology Department, Menoufia University, Shebin El Kom, Egypt
| | - Rachna Awasthi
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
| | - Yasmeen Mir
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Anuj Khurana
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Vijay Sharma
- Pathology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Usha Chandaran
- Histopathology Department, Salford Royal Hospital, Salford, UK
| | - Emad Rakha
- Histopathology Department, Nottingham City Hospital, Nottingham, UK
| | - Yasmine Maurice
- Histopathology Department, Heartlands General Hospital, Birmingham, UK
| | - Daniel Kearns
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
| | - Rami Oweis
- Histopathology Department, Rotherham Foundation Trust, Rotherham, UK
| | - Amal Asar
- Histopathology Department, Rotherham Foundation Trust, Rotherham, UK
| | | | - Abeer M Shaaban
- Cellular Pathology, Queen Elizabeth Hospital, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
95
|
O’Keefe K, Desai NV, Tan AR. Practical Guidance on Abemaciclib in Combination with Adjuvant Endocrine Therapy for Treating Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative High-Risk Early Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:517-527. [PMID: 39224861 PMCID: PMC11368096 DOI: 10.2147/bctt.s271441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
The most common subtype of breast cancer is hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer, accounting for 65-70% of all breast cancer cases diagnosed in the United States. Until 2015, single-agent endocrine therapy (ET) was the recommended first-line treatment for metastatic HR-positive, HER2-negative breast cancer. However, the paradigm has since shifted, as targeted therapy is now recommended in combination with ET. The cyclin-dependent kinase (CDK) 4/6 inhibitors have revolutionized the treatment of this breast cancer subtype, and combining either palbociclib, ribociclib, or abemaciclib with ET is now the standard first-line treatment for metastatic disease. Results of clinical trials in the metastatic setting have demonstrated that treatment with the combination of a CDK4/6 inhibitor and ET rather than ET alone is associated with longer overall survival, longer progression-free survival, and better objective response rates. Each of the CDK4/6 inhibitors has been investigated in combination with ET in patients with early-stage HR-positive, HER2-negative breast cancer who are at high risk of relapse. In October 2021, abemaciclib was the first CDK4/6 inhibitor approved in combination with ET by the US Food and Drug Administration for adjuvant treatment of patients with HR-positive, HER2-negative, high-risk early breast cancer. Herein, we provide practical guidance on the use of abemaciclib in combination with ET for HR-positive, HER2-negative, high-risk early breast cancer to assist clinicians in their day-to-day practice, and we review clinically relevant topics of dosing, side effect management, sequencing and optimal timing for initiation, and patient selection.
Collapse
Affiliation(s)
- Kaitlyn O’Keefe
- Department of Solid Tumor and Investigational Therapeutics, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC, USA
| | - Neelam V Desai
- Department of Solid Tumor and Investigational Therapeutics, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC, USA
| | - Antoinette R Tan
- Department of Solid Tumor and Investigational Therapeutics, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC, USA
| |
Collapse
|
96
|
Sato N, Tsujimoto M, Nakatsuji M, Tsuji H, Sugama Y, Shimazu K, Shimoda M, Ishihara H. Flow cytometric analysis for Ki67 assessment in formalin-fixed paraffin-embedded breast cancer tissue. BMC Biol 2024; 22:181. [PMID: 39183273 PMCID: PMC11346000 DOI: 10.1186/s12915-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Pathologists commonly employ the Ki67 immunohistochemistry labelling index (LI) when deciding appropriate therapeutic strategies for patients with breast cancer. However, despite several attempts at standardizing the Ki67 LI, inter-observer and inter-laboratory bias remain problematic. We developed a flow cytometric assay that employed tissue dissociation, enzymatic treatment and a gating process to analyse Ki67 in formalin-fixed paraffin-embedded (FFPE) breast cancer tissue. RESULTS We demonstrated that mechanical homogenizations combined with thrombin treatment can be used to recover efficiently intact single-cell nuclei from FFPE breast cancer tissue. Ki67 in the recovered cell nuclei retained reactivity against the MIB-1 antibody, which has been widely used in clinical settings. Additionally, since the method did not alter the nucleoskeletal structure of tissues, the nuclei of cancer cells can be enriched in data analysis based on differences in size and complexity of nuclei of lymphocytes and normal mammary cells. In a clinical study using the developed protocol, Ki67 positivity was correlated with the Ki67 LI obtained by hot spot analysis by a pathologist in Japan (rho = 0.756, P < 0.0001). The number of cancer cell nuclei subjected to the analysis in our assay was more than twice the number routinely checked by pathologists in clinical settings. CONCLUSIONS The findings of this study showed the application of this new flow cytometry method could potentially be used to standardize Ki67 assessments in breast cancer.
Collapse
Affiliation(s)
- Natsuki Sato
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
| | - Masahiko Tsujimoto
- Department of Diagnostic Pathology, Daini Osaka Police Hospital, 2-6-40 Karasugatsuji, Tennoji-Ku, Osaka, 543-8922, Japan
- Present Address: Osaka Pathology and Cytology Laboratory, 2-2-26 Kunijima, Higashiyodogawa-Ku, Osaka, 533-0024, Japan
| | - Masatoshi Nakatsuji
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Hiromi Tsuji
- Department of Diagnostic Pathology, Osaka Police Hospital, 10-31 Kitayamacho, Tennoji-Ku, Osaka, Japan
| | - Yuji Sugama
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hideki Ishihara
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan.
- Department of Research Support, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka, 567-0085, Japan.
| |
Collapse
|
97
|
Huang H, Xie J, Wang F, Jiao S, Li X, Wang L, Liu D, Wang C, Wei X, Tan P, Tu P, Li J, Hu Z. Commiphora myrrha n-hexane extract suppressed breast cancer progression through induction of G0/G1 phase arrest and apoptotic cell death by inhibiting the Cyclin D1/CDK4-Rb signaling pathway. Front Pharmacol 2024; 15:1425157. [PMID: 39161904 PMCID: PMC11330881 DOI: 10.3389/fphar.2024.1425157] [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: 04/29/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Background Breast cancer (BC) is one of the most frequently observed malignancies globally, yet drug development for BC has been encountering escalating challenges. Commiphora myrrha is derived from the dried resin of C. myrrha (T. Nees) Engl., and is widely adopted in China for treating BC. However, the anti-BC effect and underlying mechanism of C. myrrha remain largely unclear. Methods MTT assay, EdU assay, and colony formation were used to determine the effect of C. myrrha n-hexane extract (CMHE) on the proliferation of human BC cells. Cell cycle distribution and apoptosis were assessed via flow cytometry analysis. Moreover, metastatic potential was evaluated using wound-scratch assay and matrigel invasion assay. The 4T1 breast cancer-bearing mouse model was established to evaluate the anti-BC efficacy of CMHE in vivo. RNA-sequencing analysis, quantitative real-time PCR, immunoblotting, immunohistochemical analysis, RNA interference assay, and database analysis were conducted to uncover the underlying mechanism of the anti-BC effect of CMHE. Results We demonstrated the significant inhibition in the proliferative capability of BC cell lines MDA-MB-231 and MCF-7 by CMHE. Moreover, CMHE-induced G0/G1 phase arrest and apoptosis of the above two BC cell lines were also observed. CMHE dramatically repressed the metastatic potential of these two cells in vitro. Additionally, the administration of CMHE remarkably suppressed tumor growth in 4T1 tumor-bearing mice. No obvious toxic or side effects of CMHE administration in mice were noted. Furthermore, immunohistochemical (IHC) analysis demonstrated that CMHE treatment inhibited the proliferative and metastatic abilities of cancer cells, while also promoting apoptosis in the tumor tissues of mice. Based on RNA sequencing analysis, quantitative real-time PCR, immunoblotting, and IHC assay, the administration of CMHE downregulated Cyclin D1/CDK4-Rb signaling pathway in BC. Furthermore, RNA interference assay and database analysis showed that downregulated Cyclin D1/CDK4 signaling cascade participated in the anti-BC activity of CMHE. Conclusion CMHE treatment resulted in the suppression of BC cell growth through the stimulation of cell cycle arrest at the G0/G1 phase and the induction of apoptotic cell death via the inhibition of the Cyclin D1/CDK4-Rb pathway, thereby enhancing the anti-BC effect of CMHE. CMHE has potential anti-BC effects, particularly in those harboring aberrant activation of Cyclin D1/CDK4-Rb signaling.
Collapse
Affiliation(s)
- Huiming Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jinxin Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fei Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shungang Jiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xingxing Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Longyan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dongxiao Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chaochao Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xuejiao Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Peng Tan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
98
|
Hong CE, Lyu SY. Modulation of Breast Cancer Cell Apoptosis and Macrophage Polarization by Mistletoe Lectin in 2D and 3D Models. Int J Mol Sci 2024; 25:8459. [PMID: 39126027 PMCID: PMC11313472 DOI: 10.3390/ijms25158459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Korean mistletoe (Viscum album L. var. coloratum) is renowned for its medicinal properties, including anti-cancer and immunoadjuvant effects. This study aimed to elucidate the mechanisms by which Korean mistletoe lectin (V. album L. var. coloratum agglutinin; VCA) modulates breast cancer cell apoptosis and macrophage polarization. The specific objectives were to (1) investigate the direct effects of VCA on MCF-7 breast cancer cells and THP-1-derived M1/M2 macrophages; (2) analyze the impact of VCA on the paracrine interactions between these cell types; and (3) compare the efficacy of VCA in 2D vs. 3D co-culture models to bridge the gap between in vitro and in vivo studies. We employed both 2D and 3D models, co-culturing human M1/M2 macrophages with human MCF-7 breast cancer cells in a Transwell system. Our research demonstrated that M1 and M2 macrophages significantly influenced the immune and apoptotic responses of breast cancer cells when exposed to VCA. M1 macrophages exhibited cytotoxic characteristics and enhanced VCA-induced apoptosis in both 2D and 3D co-culture models. Conversely, M2 macrophages initially displayed a protective effect by reducing apoptosis in breast cancer cells, but this protective effect was reversed upon exposure to VCA. Furthermore, our findings illustrate VCA's ability to modulate M1 and M2 polarization in breast cancer cells. Finally, the use of magnetic 3D cell cultures suggests their potential to yield results comparable to conventional 2D cultures, bridging the gap between in vitro and in vivo studies.
Collapse
Affiliation(s)
- Chang-Eui Hong
- College of Pharmacy, Sunchon National University, Suncheon 57922, Republic of Korea;
- Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Su-Yun Lyu
- College of Pharmacy, Sunchon National University, Suncheon 57922, Republic of Korea;
- Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| |
Collapse
|
99
|
Sun S, Zhou J, Bai Y, Gao W, Lin L, Jiang T, You C, Gu Y. Role of oedema and shrinkage patterns for prediction of response to neoadjuvant chemotherapy and survival outcomes in luminal breast cancer. Clin Radiol 2024; 79:e1010-e1020. [PMID: 38830784 DOI: 10.1016/j.crad.2024.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024]
Abstract
AIMS To explore the independent and additional value of oedema and shrinkage patterns for predicting the disease-free survival (DFS) and neoadjuvant chemotherapy (NAC) response in luminal breast cancer (BC). MATERIALS AND METHODS Patients with luminal BC who underwent NAC were enrolled in this study from 2017 to 2022. Traditional MRI features include BI-RADS-based MRI descriptors, tumor size, and ADC values, while emerging MRI features include oedema and shrinkage patterns, all of which were evaluated before, early, and after NAC. The changes in features during NAC were also evaluated. The value of features was evaluated through univariate, multivariate analyses. RESULTS A total of 258 patients were enrolled in this study, of which 77 responded to NAC. Diffuse oedema, stable or increased oedema during early NAC were adverse predictors for treatment response, while a greater reduction in tumor size and increase in ADC value were favorable predictors (all P<0.05). Furthermore, 20 of 60 patients who were followed up experienced recurrence. Diffuse oedema, pre-pectoral or subcutaneous oedema, and non-concentric shrinkage patterns after NAC were risk factors for DFS, whereas a greater increase in ADC value was a protective factor. Incorporating oedema and shrinkage patterns into traditional MRI features improved the predictive performance for treatment response (AUC from 0.76-0.78 to 0.80-0.83) and DFS (C-index from 0.67-69 to 0.75-0.80). CONCLUSIONS Oedema is an unfavorable predictor for treatment response and survival outcomes, while shrinkage patterns contribute more to the prognostic value, both of which could offer supplementary benefits for clinical outcomes in luminal BC.
Collapse
Affiliation(s)
- S Sun
- Shanghai Institute of Medical Imaging, Shanghai 200032, China; Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - J Zhou
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Y Bai
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
| | - W Gao
- Department of Radiology, The First People's Hospital of Honghe State, Mengzi, Yunnan 661100, China
| | - L Lin
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - T Jiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - C You
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Y Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
100
|
Rewcastle E, Skaland I, Gudlaugsson E, Fykse SK, Baak JPA, Janssen EAM. The Ki67 dilemma: investigating prognostic cut-offs and reproducibility for automated Ki67 scoring in breast cancer. Breast Cancer Res Treat 2024; 207:1-12. [PMID: 38797793 PMCID: PMC11231004 DOI: 10.1007/s10549-024-07352-4] [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: 03/09/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Quantification of Ki67 in breast cancer is a well-established prognostic and predictive marker, but inter-laboratory variability has hampered its clinical usefulness. This study compares the prognostic value and reproducibility of Ki67 scoring using four automated, digital image analysis (DIA) methods and two manual methods. METHODS The study cohort consisted of 367 patients diagnosed between 1990 and 2004, with hormone receptor positive, HER2 negative, lymph node negative breast cancer. Manual scoring of Ki67 was performed using predefined criteria. DIA Ki67 scoring was performed using QuPath and Visiopharm® platforms. Reproducibility was assessed by the intraclass correlation coefficient (ICC). ROC curve survival analysis identified optimal cutoff values in addition to recommendations by the International Ki67 Working Group and Norwegian Guidelines. Kaplan-Meier curves, log-rank test and Cox regression analysis assessed the association between Ki67 scoring and distant metastasis (DM) free survival. RESULTS The manual hotspot and global scoring methods showed good agreement when compared to their counterpart DIA methods (ICC > 0.780), and good to excellent agreement between different DIA hotspot scoring platforms (ICC 0.781-0.906). Different Ki67 cutoffs demonstrate significant DM-free survival (p < 0.05). DIA scoring had greater prognostic value for DM-free survival using a 14% cutoff (HR 3.054-4.077) than manual scoring (HR 2.012-2.056). The use of a single cutoff for all scoring methods affected the distribution of prediction outcomes (e.g. false positives and negatives). CONCLUSION This study demonstrates that DIA scoring of Ki67 is superior to manual methods, but further study is required to standardize automated, DIA scoring and definition of a clinical cut-off.
Collapse
Affiliation(s)
- Emma Rewcastle
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway.
| | - Ivar Skaland
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Einar Gudlaugsson
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Silja Kavlie Fykse
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Jan P A Baak
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Emiel A M Janssen
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|