1
|
Li Y, Ma J, Ma X, Chen C, Ruan M, Yang W, Shui R. PD-L1 expression and tumor-infiltrating lymphocytes: Correlations and prognostic values in Chinese triple-negative breast cancer patients with different molecular subtyping. Pathol Res Pract 2024; 262:155556. [PMID: 39216323 DOI: 10.1016/j.prp.2024.155556] [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: 04/09/2024] [Revised: 07/26/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To investigate the correlation between programmed death ligand-1 (PD-L1) expression and tumor-infiltrating lymphocytes (TILs) and evaluate the prognostic value of PD-L1 and TILs in Chinese triple-negative breast cancer (TNBC) patients with different molecular subtype METHODS: This retrospective study was conducted at 2020. Specifically, the pre-chemotherapy clinical data and non-stained tissue blocks of 465 TNBC patients visited the Fudan University Shanghai Cancer Center (FUSCC) between 2008 and 2014 were collected, with their blocks sliced and stained using PD-L1(SP142), and the outcome of subsequent chemotherapy obtained in 2020. The relapse-free survival (RFS) of the study population was calculated. The baseline PD-L1 expression status correlations with TILs and molecular subtypes were assessed using Spearman's rank correlation analysis and the Kruskal-Wallis test. Kaplan-Meier survival analyses were undertaken to evaluate the prognosis value of TILs and PD-L1 expression. RESULTS PD-L1 expression status on IC was moderately and positively correlated with stromal tumor-infiltrating lymphocytes (sTILs) (rs = 0.502, P <0.001) and iTILs (rs = 0.410, P < 0.001), respectively. PD-L1 expression status and TILs showed significant differences among molecular subtypes (P < 0.001), with the highest proportion of PD-L1+ and high TILs patients observed in the immunomodulatory (IM) subtype. TILs were significantly associated with RFS. Moreover, sTILs could act as an independent predictor of RFS (RR 0.953, 95 % CI 0.920 ∼ 0.987, P = 0.007), while PD-L1 expression status did not show the same prognostic significance. CONCLUSIONS The incorporation of pre-treatment TILs and PD-L1 expression status as valuable tools for optimizing patient selection for immunotherapy and managing the risks associated with chemotherapy in Chinese TNBC patients. DATA AVAILABILITY The data sets generated and analyzed during the current study are available from the corresponding author.
Collapse
Affiliation(s)
- Yanping Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jing Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoxi Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Chen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Miao Ruan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Ruohong Shui
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
2
|
de Moraes FCA, Souza MEC, Sano VKT, Moraes RA, Melo AC. Association of tumor-infiltrating lymphocytes with clinical outcomes in patients with triple-negative breast cancer receiving neoadjuvant chemotherapy: a systematic review and meta-analysis. Clin Transl Oncol 2024:10.1007/s12094-024-03661-8. [PMID: 39154313 DOI: 10.1007/s12094-024-03661-8] [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: 06/29/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVE Triple-negative breast cancer (TNBC) presents a clinical challenge as an aggressive tumor, correlated with unfavorable prognosis. Tumor-infiltrating lymphocytes (TILs) have garnered interest as a potential prognostic biomarker. However, the disparity in outcomes between varying TILs rates remains inadequately explored. METHODS PubMed, Scopus, Web of Science, and Cochrane databases were searched for studies about the prognostic value of TILs in patients with TNBC receiving neoadjuvant chemotherapy. The hazard ratios (HRs) or odds ratios (ORs) were computed for binary endpoints, with 95% confidence intervals (CIs). RESULTS Twenty-nine studies were included, involving a population of six thousand one hundred sixty-one (80.41%) with TNBC. The cut-off TILs value ranged from 10 to 60%, with 50% being the most related value. Compared with the low-TIL expression group, the disease-free survival (DFS) (HR 0.71; 95% CI 0.61-0.82; p < 0.00001) and overall survival (OS) (HR 0.76; 95% CI 0.63-0.90; p = 0.002) rates showed significant improvement with higher TIL infiltrations. In the subgroup analyses of the lymphocyte subtypes CD4 + and CD8 + , there was statistical significance favoring higher TILs rates in both subtypes, each associated with improved DFS (HR 0.48; 95% CI 0.33-0.71; p = 0.0002) and OS (HR 0.53; 95% CI 0.36-0.78; p = 0.001), regardless of which cell subtype was predominantly infiltrated. The complete pathological response analysis showed better rates for the higher TIL group than the control for both the TIL (OR 1.29; 95% CI 1.13-1.48; p = 0.0003) and Ki-67 (OR 2.74; 95% CI 2.01-3.73; p < 0.00001) analyses. CONCLUSION Higher expressions of TILs in patients with TNBC were associated with improved significantly DFS, OS, and pCR outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Ana C Melo
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
3
|
Hu L, Gu Y, Xu W, Wang C. Association of clinicopathologic and sonographic features with stromal tumor-infiltrating lymphocytes in triple-negative breast cancer. BMC Cancer 2024; 24:997. [PMID: 39135184 PMCID: PMC11320771 DOI: 10.1186/s12885-024-12778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Increased level of stromal tumor-infiltrating lymphocytes (sTILs) are associated with therapeutic outcomes and prognosis in triple-negative breast cancer (TNBC). This study aimed to investigate the associations of clinicopathologic and sonographic features with sTILs level in TNBC. METHODS This study included invasive TNBC patients with postoperative evaluation of sTILs after surgical resection. Tumor shape, margin, orientation, echo pattern, posterior features, calcification, and vascularity were retrospectively evaluated. The patients were categorized into high-sTILs (≥ 20%) and low-sTILs (< 20%) level groups. Chi-square or Fisher's exact tests were used to assess the association of clinicopathologic and sonographic features with sTILs level. RESULTS The 171 patients (mean ± SD age, 54.7 ± 10.3 years [range, 22‒87 years]) included 58.5% (100/171) with low-sTILs level and 41.5% (71/171) with high-sTILs level. The TNBC tumors with high-sTILs level were more likely to be no special type invasive carcinoma (p = 0.008), higher histologic grade (p = 0.029), higher Ki-67 proliferation rate (all p < 0.05), and lower frequency of associated DCIS component (p = 0.026). In addition, the TNBC tumors with high-sTILs level were more likely to be an oval or round shape (p = 0.001), parallel orientation (p = 0.011), circumscribed or micro-lobulated margins (p < 0.001), complex cystic and solid echo patterns (p = 0.001), posterior enhancement (p = 0.002), and less likely to have a heterogeneous pattern (p = 0.001) and no posterior features (p = 0.002). CONCLUSIONS This preliminary study showed that preoperative sonographic characteristics could be helpful in distinguishing high-sTILs from low-sTILs in TNBC patients.
Collapse
Affiliation(s)
- Ling Hu
- Department of Ultrasound in Medicine, Hangzhou Women's Hospital, Hangzhou, Zhejiang, China
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yunxia Gu
- Department of Ultrasound in Medicine, Hangzhou Women's Hospital, Hangzhou, Zhejiang, China
| | - Wen Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chao Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Khan AA, Ahuja S, Kiruthikasri K, Zaheer S. Assessment of stromal tumor-infiltrating lymphocytes in neoadjuvant chemotherapy for invasive breast carcinoma: Predictive insights across molecular subtypes. Pathol Res Pract 2024; 260:155382. [PMID: 38850879 DOI: 10.1016/j.prp.2024.155382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND AND AIMS Breast cancer, a leading cause of female mortality, has prompted the widespread adoption of Neoadjuvant chemotherapy (NAC) for its potential to minimize metastasis risk and downstaging tumors. Tumor Infiltrating Lymphocytes (TILs) have emerged as key immunological biomarkers, particularly in breast cancer research. This study focuses on evaluating Stromal TILs (sTILs) in pre-NAC core needle biopsies of Invasive Breast Carcinoma, No Special Type (IBC, NST) and correlating it with NAC response. MATERIALS AND METHODS A retrospective study spanning three years (October 2020 to September 2023) was conducted in a tertiary care hospital, involving 73 patients meeting specific inclusion criteria. Pathological assessments, including hormone receptor status, molecular subtyping, and TILs evaluation, were performed. Logistic regression and statistical analyses were conducted to determine associations between TILs, clinicopathological parameters, and complete response. RESULTS The study demonstrated excellent discriminatory power of TILs (>10 %) in predicting complete response. Univariate and multivariate logistic regression underscored the independent predictive value of TILs, emphasizing their significance across diverse molecular subtypes. CONCLUSION This study provides crucial insights into immune response assessment, particularly sTILs, in optimizing breast cancer treatment strategies and patient outcomes during NAC, contributing to the evolving landscape of personalized emphasising oncology.
Collapse
Affiliation(s)
- Adil Aziz Khan
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Kiruthikasri Kiruthikasri
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
5
|
Rustamadji P, Wiyarta E, Pramono M, Maulanisa SC. Response to Neoadjuvant Chemotherapy in Invasive Breast Cancer Predicted by CD4+, CD8+, and FOXP3+ Tumor-Infiltrating Lymphocytes. Asian Pac J Cancer Prev 2024; 25:1607-1613. [PMID: 38809632 PMCID: PMC11318808 DOI: 10.31557/apjcp.2024.25.5.1607] [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: 11/22/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Response to neoadjuvant chemotherapy (NC) in individuals with invasive breast cancer (IBC) must be monitored, and biomarkers are needed. NC can activate an anti-tumour immune response in its microenvironment, known as Tumor-infiltrating Lymphocytes (TIL). TIL components believed to have great potential as predictors are CD4+, CD8+, and FOXP3+ TIL. This study aims to explore TIL components that can potentially be predictive biomarkers of NC pathological responses. METHODS A sample size of 40 were analyzed based on the relationship between CD4+, CD8+, and FOXP3+ TIL expression with the Miller-Payne (MP) grading system. Age, tumour grade, PR, ER, Ki-67, and HER2 were also evaluated. CD4+, CD8+, and FOXP3+ TIL expressions were analayzed by IHC staining, while other data were collected from archives. Data was analyzed using univariate and multivariate analysis. RESULTS Univariate analysis showed a significant relationship between CD4+ TIL and MP (p<0.001), CD8+ and MP (p=0.004), and FOXP3 with MP (p<0.001). The simultaneous integration of the three biomarkers in one model was not good enough to be a predictive model. Therefore, an exploratory analysis was conducted by testing several alternative models that combined two of the three existing biomarkers. It turned out that CD4+ TIL in model 2 (CD4+CD8+) and FOXP3+ TIL in model 4 (CD8+FOXP3+) showed significant coefficient values. Moreover, all of the threshold coefficients in model 4 are significant. CONCLUSION This study shows that CD4+, CD8+, and FOXP3+ TIL have promising potential as predictive biomarkers. In particular, FOXP3+ is dominant in predictive models of pathological response in patients with IBC.
Collapse
MESH Headings
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Female
- Forkhead Transcription Factors/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Neoadjuvant Therapy/methods
- Middle Aged
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Biomarkers, Tumor/metabolism
- Prognosis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Adult
- Follow-Up Studies
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Tumor Microenvironment/immunology
- Neoplasm Invasiveness
- Aged
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/metabolism
Collapse
Affiliation(s)
- Primariadewi Rustamadji
- Department of Anatomic Pathology, Faculty of Medicine Universitas Indonesia-Dr. Cipto Mangunkusumo National Hospital, Jakarta, Indonesia.
| | | | | | | |
Collapse
|
6
|
Hu L, Jin P, Xu W, Wang C, Huang P. Clinical and radiomics integrated nomogram for preoperative prediction of tumor-infiltrating lymphocytes in patients with triple-negative breast cancer. Front Oncol 2024; 14:1370466. [PMID: 38567151 PMCID: PMC10985173 DOI: 10.3389/fonc.2024.1370466] [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: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Objectives The present study aimed to develop a radiomics nomogram based on conventional ultrasound (CUS) to preoperatively distinguish high tumor-infiltrating lymphocytes (TILs) and low TILs in triple-negative breast cancer (TNBC) patients. Methods In the present study, 145 TNBC patients were retrospectively included. Pathological evaluation of TILs in the hematoxylin and eosin sections was set as the gold standard. The patients were randomly allocated into training dataset and validation dataset with a ratio of 7:3. Clinical features (age and CUS features) and radiomics features were collected. Then, the Rad-score model was constructed after the radiomics feature selection. The clinical features model and clinical features plus Rad-score (Clin+RS) model were built using logistic regression analysis. Furthermore, the performance of the models was evaluated by analyzing the receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA). Results Univariate analysis and LASSO regression were employed to identify a subset of 25 radiomics features from a pool of 837 radiomics features, followed by the calculation of Rad-score. The Clin+RS integrated model, which combined posterior echo and Rad-score, demonstrated better predictive performance compared to both the Rad-score model and clinical model, achieving AUC values of 0.848 in the training dataset and 0.847 in the validation dataset. Conclusion The Clin+RS integrated model, incorporating posterior echo and Rad-score, demonstrated an acceptable preoperative evaluation of the TIL level. The Clin+RS integrated nomogram holds tremendous potential for preoperative individualized prediction of the TIL level in TNBC.
Collapse
Affiliation(s)
- Ling Hu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Ultrasound in Medicine, Hangzhou Women’s Hospital, Hangzhou, Zhejiang, China
| | - Peile Jin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wen Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chao Wang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Huertas-Caro CA, Ramírez MA, Rey-Vargas L, Bejarano-Rivera LM, Ballen DF, Nuñez M, Mejía JC, Sua-Villegas LF, Cock-Rada A, Zabaleta J, Fejerman L, Sanabria-Salas MC, Serrano-Gomez SJ. Tumor infiltrating lymphocytes (TILs) are a prognosis biomarker in Colombian patients with triple negative breast cancer. Sci Rep 2023; 13:21324. [PMID: 38044375 PMCID: PMC10694133 DOI: 10.1038/s41598-023-48300-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023] Open
Abstract
Triple negative breast cancer (TNBC) is highly immunogenic and high levels of tumor infiltrating lymphocytes (TILs) have been associated with a better prognosis and higher probability to achieve pathological complete response. Here, we explore the potential role of stromal TILs level and composition as a prognostic and predictive biomarker in TNBC. 195 Tumor biospecimens from patients diagnosed with TNBC were included. Stromal TILs (sTILs), positive CD4/CD8 cells were evaluated. Differences in clinic-pathological characteristics according to immune infiltration were assessed. The predictive and prognostic value of immune infiltration was analyzed by multivariate models. Higher immune infiltration was observed in patients with favorable clinical-pathological features. Survival analysis showed that longer overall survival times were observed in patients with a higher infiltration of sTILs (p = 0.00043), CD4 + (p = 0.0074) and CD8 + (p = 0.008). In the multivariate analysis, low levels of sTILs were found to be associated with a higher mortality hazard (HR: 1.59, 95% CI 1.01-2.48). CD4 and CD8 immune infiltration were associated with higher odds for pathological complete response (OR: 1.20, 95% CI 1.00-1.46, OR: 1.28, 1.02-1.65, respectively). Our results suggest that immune infiltration could be used as a prognostic marker for overall survival in TNBC patients.
Collapse
Affiliation(s)
- Carlos A Huertas-Caro
- Cancer Biology Research Group, National Cancer Institute of Colombia, Bogotá, Colombia
| | - Mayra A Ramírez
- Cancer Biology Research Group, National Cancer Institute of Colombia, Bogotá, Colombia
| | - Laura Rey-Vargas
- Cancer Biology Research Group, National Cancer Institute of Colombia, Bogotá, Colombia
| | | | - Diego Felipe Ballen
- Clinical Oncology Unit. Instituto Nacional de Cancerología and Adjunct Clinical Professor, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Marcela Nuñez
- Research Support and Follow-Up Group, National Cancer Institute of Colombia, Calle 1 No. 9 -85, Bogotá, DC, Colombia
| | - Juan Carlos Mejía
- Grupo de Patología, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Luz Fernanda Sua-Villegas
- Department of Pathology and Laboratory Medicine, Fundación Valle del Lili, and Faculty of Health Sciences, Universidad ICESI, Cali, Colombia
| | - Alicia Cock-Rada
- Department of Oncological Breast Surgery and Mastology, Instituto de Cancerología Las Américas, Medellín, Colombia
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Laura Fejerman
- Department of Public Health Sciences, University of California Davis, Davis, CA, USA
- UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | | | - Silvia J Serrano-Gomez
- Cancer Biology Research Group, National Cancer Institute of Colombia, Bogotá, Colombia.
- Research Support and Follow-Up Group, National Cancer Institute of Colombia, Calle 1 No. 9 -85, Bogotá, DC, Colombia.
| |
Collapse
|
8
|
Landén AH, Chin K, Kovács A, Holmberg E, Molnar E, Stenmark Tullberg A, Wärnberg F, Karlsson P. Evaluation of tumor-infiltrating lymphocytes and mammographic density as predictors of response to neoadjuvant systemic therapy in breast cancer. Acta Oncol 2023; 62:1862-1872. [PMID: 37934084 DOI: 10.1080/0284186x.2023.2274483] [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: 06/16/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Response rates vary among breast cancer patients treated with neoadjuvant systemic therapy (NAST). Thus, there is a need for reliable treatment predictors. Evidence suggests tumor-infiltrating lymphocytes (TILs) predict NAST response. Still, TILs are seldom used clinically as a treatment determinant. Mammographic density (MD) is another potential marker for NAST benefit and its relationship with TILs is unknown. Our aims were to investigate TILs and MD as predictors of NAST response and to study the unexplored relationship between TILs and MD. MATERIAL AND METHODS We studied 315 invasive breast carcinomas treated with NAST between 2013 and 2020. Clinicopathological data were retrieved from medical records. The endpoint was defined as pathological complete response (pCR) in the breast. TILs were evaluated in pre-treatment core biopsies and categorized as high (≥10%) or low (<10%). MD was scored (a-d) according to the breast imaging reporting and data system (BI-RADS) fifth edition. Binary logistic regression and Spearman's test of correlation were performed using SPSS. RESULTS Out of 315 carcinomas, 136 achieved pCR. 94 carcinomas had high TILs and 215 had low TILs. Six carcinomas had no available TIL data. The number of carcinomas in each BI-RADS category were 37, 122, 112, and 44 for a, b, c, and d, respectively. High TILs were independently associated with pCR (OR: 2.95; 95% CI: 1.59-5.46) compared to low TILs. In the univariable analysis, MD (BI-RADS d vs. a) showed a tendency of higher likelihood for pCR (OR: 2.43; 95% CI: 0.99-5.98). However, the association was non-significant, which is consistent with the result of the multivariable analysis (OR: 2.51; 95% CI: 0.78-8.04). We found no correlation between TILs and MD (0.02; p = .80). CONCLUSION TILs significantly predicted NAST response. We could not define MD as a significant predictor of NAST response. These findings should be further replicated.
Collapse
Affiliation(s)
- Amalia H Landén
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kian Chin
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Erik Holmberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eva Molnar
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Axel Stenmark Tullberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Wärnberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Kusama H, Kittaka N, Soma A, Taniguchi A, Kanaoka H, Nakajima S, Oyama Y, Seto Y, Okuno J, Watanabe N, Matsui S, Nishio M, Fujisawa F, Honma K, Tamaki Y, Nakayama T. Predictive factors for response to neoadjuvant chemotherapy: inflammatory and immune markers in triple-negative breast cancer. Breast Cancer 2023; 30:1085-1093. [PMID: 37782377 DOI: 10.1007/s12282-023-01504-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/09/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) predict response to neoadjuvant chemotherapy (NAC) in triple-negative breast cancer (TNBC) patients. However, the TIL level can be determined at a few facilities. By contrast, neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) are easily and objectively determined from the results of full blood counts. We conducted a retrospective study to investigate whether TILs, NLR, and PLR predict NAC efficacy and whether NLR and PLR could be surrogate markers for TILs in TNBC. METHODS Of the 266 patients diagnosed with TNBC between 2013 and 2019, 66 who underwent radical surgery after sequential administration of anthracycline and taxane as NAC were included in the study. TILs, NLR, and PLR were evaluated as predictors of pathologic complete response (pCR) using cutoff values determined from receiver operating characteristic curves. RESULTS The cutoff values of TILs, NLR, and PLR were 20%, 2.6, and 180, respectively. High TIL level was associated with low NLR (P = 0.01) and low PLR (P = 0.01). High TIL level (odds ratio [OR] 4.28 [95% CI 1.40-13.1]; P = 0.01), low NLR (OR 5.51 [95% CI 1.60-18.9]; P = 0.01), and low PLR (OR 3.29 [95% CI 1.13-9.57]; P = 0.03) were associated with pCR. Low NLR predicted pCR independently (OR 6.59 [95% CI 1.45-30.0]; P = 0.01). CONCLUSIONS TILs, NLR, and PLR predicted NAC efficacy against TNBC. TIL level was associated with NLR and PLR. NLR was an independent predictive factor and may be a useful surrogate marker for TILs when predicting pCR.
Collapse
Affiliation(s)
- Hiroki Kusama
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Nobuyoshi Kittaka
- Department of Breast Surgery, Osaka Rosai Hospital, 1179-3 Nagasone-Cho Kita-Ku Sakai-Shi, Osaka, 541-8567, Japan
| | - Ai Soma
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Azusa Taniguchi
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Haruka Kanaoka
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Satomi Nakajima
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Yuri Oyama
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Yukiko Seto
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Jun Okuno
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Noriyuki Watanabe
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Saki Matsui
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan
| | - Minako Nishio
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Fumie Fujisawa
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Keiichiro Honma
- Department of Pathology, Osaka International Cancer Institute, Osaka, Japan
| | - Yasuhiro Tamaki
- Department of Breast and Endocrine Surgery, Kaizuka City Hospital, 3-10-20 Hori Kaizuka-Shi, Osaka, 597-0015, Japan
| | - Takahiro Nakayama
- Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, 3-1-69 Otemae Chuo-Ku, Osaka, 541-8567, Japan.
| |
Collapse
|
10
|
Li S, Liu Y, Zhang P, Wang M, Sun L. Cost-effectiveness analysis of tumor-infiltrating lymphocytes biomarkers guiding chemotherapy de-escalation in early triple-negative breast cancer. Cancer Med 2023; 12:21001-21012. [PMID: 37964682 PMCID: PMC10709734 DOI: 10.1002/cam4.6656] [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: 04/26/2023] [Revised: 09/22/2023] [Accepted: 10/01/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND To accelerate the clinical translation of tumor-infiltrating lymphocytes (TILs) biomarkers for guiding chemotherapy de-escalation in early-stage triple-negative breast cancer (TNBC), cost-effectiveness evidence is essential but has not been investigated. We intend to evaluate the cost-effectiveness of using TILs to guiding chemotherapy de-escalation in patients with early-stage TNBC from the perspective of the Chinese health service system. METHODS The hybrid decision-tree-Markov model was designed to compare the cost-effectiveness of cytotoxic chemotherapy guided by whether TILs assay was performed in 50-year-old female patients with early-stage TNBC over a lifetime horizon. In Strategy (1), if TILs testing was performed, patients with TILs values exceeding 30% could be spared from chemotherapy. In Strategy (2), where no TILs testing was performed, all patients were administered chemotherapy following China's clinical practices. Based on the algorithm built by Guyot, the individual patient data were reconstructed from the published Kaplan-Meier curves, and the survival functions were calculated by parametric methods. Cost estimates were valued in Chinese yuan (as per rates in 2022). RESULTS In 50-year-old female patients with early-stage TNBC, Strategy (1), which employs TILs testing to guide cytotoxic chemotherapy yielded an additional 0.47 quality-adjusted life years (QALYs) and saved 40,976 yuan, with an incremental cost-effectiveness ratio (ICER) of -87,182.98 yuan per QALY gained compared with Strategy (2). This indicates that compared with Strategy (2), Strategy (1) is the dominant scheme. The results were sensitive to utility parameters, discount rates, and treatment costs after relapse. At a willingness-to-pay threshold of 85,700 yuan (based on GDP per capita) per QALY, the probability of TILs being cost-effective was almost 100%. CONCLUSIONS The application of biomarkers (TILs) to guide decisions for chemotherapy de-escalation is a cost-effective strategy for early-stage TNBC patients and deserves to be widely promoted in clinical practice.
Collapse
Affiliation(s)
- Shiqi Li
- Department of Pharmacy Administration, School of Business AdministrationShenyang Pharmaceutical UniversityShenyangChina
| | - Yuhan Liu
- Shanghai Health Development Research Centre (Shanghai Medical Information Centre)ShanghaiChina
| | - Peigen Zhang
- Department of Pharmacy Administration, School of Business AdministrationShenyang Pharmaceutical UniversityShenyangChina
| | - Mengmeng Wang
- The Department of CardiologyGeneral Hospital of Northern Theater CommandShenyangChina
| | - Lihua Sun
- Department of Pharmacy Administration, School of Business AdministrationShenyang Pharmaceutical UniversityShenyangChina
| |
Collapse
|
11
|
Johnson KCC, Goldstein D, Tharakan J, Quiroga D, Kassem M, Grimm M, Miah A, Vargo C, Berger M, Sudheendra P, Pariser A, Gatti-Mays ME, Williams N, Stover D, Sardesai S, Wesolowski R, Ramaswamy B, Tozbikian G, Schnell PM, Cherian MA. The Immunomodulatory Effects of Dexamethasone on Neoadjuvant Chemotherapy for Triple-Negative Breast Cancer. Oncol Ther 2023; 11:361-374. [PMID: 37354381 PMCID: PMC10447758 DOI: 10.1007/s40487-023-00235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 05/26/2023] [Indexed: 06/26/2023] Open
Abstract
INTRODUCTION The immunomodulatory impact of corticosteroids and concurrent chemotherapy is poorly understood within triple-negative breast cancer (TNBC). On a biochemical level, steroids have been linked to the signaling of chemotherapy-resistant pathways. However, on a clinical level, steroids play an essential role in chemotherapy tolerance through the prevention of chemotherapy-induced nausea and vomiting (CINV) and hypersensitivity reactions. Given these conflicting roles, we wanted to evaluate this interplay more rigorously in the context of early-stage TNBC. METHODS We performed a retrospective analysis of patients with operable TNBC who received neoadjuvant chemotherapy (NAC) between January 2012 and November 2018, with the primary goal of examining the dose-dependent relationship between pathological complete response (pCR) rates and corticosteroid use. Secondary endpoints included the impact of steroid dosing on overall survival (OS) and recurrence-free survival (RFS), along with a breakdown in pCR rates based on steroid doses provided during each chemotherapy phase. Further adjusted analyses were performed based on patient age, diabetic status, and anatomical stage. Finally, we explored the relationship between tumor-infiltrating lymphocytes (TILs) seen on tissue samples at baseline and dexamethasone doses in terms of pCR rates. RESULTS In total, of the 174 patients screened within this study period, 116 met full eligibility criteria. Of these eligible patients, all were female, with a median age of 51.5 years (27.0 to 74.0) and a mean body mass index (BMI) of 29.7 [standard deviation (SD) 7.04]. The majority were nondiabetic (80.2%). For cancer stage, 69.8% (n = 81) had stage 2 breast cancer. We found no statistically significant association between pCR rates and dexamethasone use, both in terms of the total dose (p = 0.55) and mean dose per NAC cycle (p = 0.74). Similarly, no difference was noted when adjusting for diabetic status, metformin use, or age at diagnosis, regardless of the total steroid dose provided (p = 0.72) or mean dose per cycle (p = 0.49). No meaningful changes to pCR rate were seen with higher mean or higher total steroid doses during the paclitaxel (T) phase (adjusted p = 0.16 and p = 0.76, respectively) or doxorubicin and cyclophosphamide (AC) phase (adjusted p = 0.83 and p = 0.77, respectively). Furthermore, we found no clinically significant association between dexamethasone dose and either RFS (p = 0.45) or OS (p = 0.89). Of the 56 patients who had available pre-treatment biopsy tissue samples, 27 achieved pCR, with higher TILs at baseline being associated with higher pCR rates, regardless of the mean dexamethasone dose used. CONCLUSION Our findings demonstrate that dexamethasone has no clinically significant impact on pCR, RFS, or OS when given concurrently with NAC in patients with curative TNBC, regardless of diabetic status.
Collapse
Affiliation(s)
- Kai Conrad Cecil Johnson
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | | | - Jasmin Tharakan
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Dionisia Quiroga
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Mahmoud Kassem
- Department of Surgery, Mercy Health West Hospital, Cincinnati, OH, USA
| | - Michael Grimm
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Abdul Miah
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Craig Vargo
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Michael Berger
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Preeti Sudheendra
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Ashley Pariser
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Margaret E Gatti-Mays
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Nicole Williams
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Daniel Stover
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Sagar Sardesai
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Gary Tozbikian
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Patrick M Schnell
- Division of Biostatistics, The Ohio State University College of Public Health, Columbus, OH, USA
| | - Mathew A Cherian
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Abuhadra N, Sun R, Yam C, Rauch GM, Ding Q, Lim B, Thompson AM, Mittendorf EA, Adrada BE, Damodaran S, Virani K, White J, Ravenberg E, Sun J, Choi J, Candelaria R, Arun B, Ueno NT, Santiago L, Saleem S, Abouharb S, Murthy RK, Ibrahim N, Sahin A, Valero V, Symmans WF, Litton JK, Tripathy D, Moulder S, Huo L. Predictive Roles of Baseline Stromal Tumor-Infiltrating Lymphocytes and Ki-67 in Pathologic Complete Response in an Early-Stage Triple-Negative Breast Cancer Prospective Trial. Cancers (Basel) 2023; 15:3275. [PMID: 37444385 DOI: 10.3390/cancers15133275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
High stromal tumor-infiltrating lymphocytes (sTILs) are associated with improved pathologic complete response (pCR) in triple-negative breast cancer (TNBC). We hypothesize that integrating high sTILs and additional clinicopathologic features associated with pCR could enhance our ability to predict the group of patients on whom treatment de-escalation strategies could be tested. In this prospective early-stage TNBC neoadjuvant chemotherapy study, pretreatment biopsies from 408 patients were evaluated for their clinical and demographic features, as well as biomarkers including sTILs, Ki-67, PD-L1 and androgen receptor. Multivariate logistic regression models were developed to generate a computed response score to predict pCR. The pCR rate for the entire cohort was 41%. Recursive partitioning analysis identified ≥20% as the optimal cutoff for sTILs to denote 35% (143/408) of patients as having high sTILs, with a pCR rate of 59%, and 65% (265/408) of patients as having low sTILs, with a pCR rate of 31%. High Ki-67 (cutoff > 35%) was identified as the only predictor of pCR in addition to sTILs in the training set. This finding was verified in the testing set, where the highest computed response score encompassing both high sTILa and high Ki-67 predicted a pCR rate of 65%. Integrating Ki67 and sTIL may refine the selection of early stage TNBC patients for neoadjuvant clinical trials evaluating de-escalation strategies.
Collapse
Affiliation(s)
- Nour Abuhadra
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gaiane M Rauch
- Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qingqing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bora Lim
- Department of Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alastair M Thompson
- Division of Surgical Oncology, Section of Breast Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Beatriz E Adrada
- Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kiran Virani
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jason White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jia Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jaihee Choi
- Department of Statistics, Rice University, Houston, TX 77005, USA
| | - Rosalind Candelaria
- Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lumarie Santiago
- Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sadia Saleem
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sausan Abouharb
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rashmi K Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nuhad Ibrahim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aysegul Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - William Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stacy Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
13
|
van den Ende NS, Nguyen AH, Jager A, Kok M, Debets R, van Deurzen CHM. Triple-Negative Breast Cancer and Predictive Markers of Response to Neoadjuvant Chemotherapy: A Systematic Review. Int J Mol Sci 2023; 24:ijms24032969. [PMID: 36769287 PMCID: PMC9918290 DOI: 10.3390/ijms24032969] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Around 40-50% of all triple-negative breast cancer (TNBC) patients achieve a pathological complete response (pCR) after treatment with neoadjuvant chemotherapy (NAC). The identification of biomarkers predicting the response to NAC could be helpful for personalized treatment. This systematic review provides an overview of putative biomarkers at baseline that are predictive for a pCR following NAC. Embase, Medline and Web of Science were searched for articles published between January 2010 and August 2022. The articles had to meet the following criteria: patients with primary invasive TNBC without distant metastases and patients must have received NAC. In total, 2045 articles were screened by two reviewers resulting in the inclusion of 92 articles. Overall, the most frequently reported biomarkers associated with a pCR were a high expression of Ki-67, an expression of PD-L1 and the abundance of tumor-infiltrating lymphocytes, particularly CD8+ T cells, and corresponding immune gene signatures. In addition, our review reveals proteomic, genomic and transcriptomic markers that relate to cancer cells, the tumor microenvironment and the peripheral blood, which also affect chemo-sensitivity. We conclude that a prediction model based on a combination of tumor and immune markers is likely to better stratify TNBC patients with respect to NAC response.
Collapse
Affiliation(s)
- Nadine S. van den Ende
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
- Correspondence: ; Tel.: +31-640213383
| | - Anh H. Nguyen
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Marleen Kok
- Department of Medical Oncology, Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Carolien H. M. van Deurzen
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
14
|
Deep learning with biopsy whole slide images for pretreatment prediction of pathological complete response to neoadjuvant chemotherapy in breast cancer:A multicenter study. Breast 2022; 66:183-190. [PMID: 36308926 PMCID: PMC9619175 DOI: 10.1016/j.breast.2022.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 09/18/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
INTRODUCTION Predicting pathological complete response (pCR) for patients receiving neoadjuvant chemotherapy (NAC) is crucial in establishing individualized treatment. Whole-slide images (WSIs) of tumor tissues reflect the histopathologic information of the tumor, which is important for therapeutic response effectiveness. In this study, we aimed to investigate whether predictive information for pCR could be detected from WSIs. MATERIALS AND METHODS We retrospectively collected data from four cohorts of 874 patients diagnosed with biopsy-proven breast cancer. A deep learning pathological model (DLPM) was constructed to predict pCR using biopsy WSIs in the primary cohort, and it was then validated in three external cohorts. The DLPM could generate a deep learning pathological score (DLPs) for each patient; stromal tumor-infiltrating lymphocytes (TILs) were selected for comparison with DLPs. RESULTS The WSI feature-based DLPM showed good predictive performance with the highest area under the curve (AUC) of 0.72 among the cohorts. Alternatively, the combination of the DLPM and clinical characteristics offered a better prediction performance (AUC >0.70) in all cohorts. We also evaluated the performance of DLPM in three different breast subtypes with the best prediction for the triple-negative breast cancer (TNBC) subtype (AUC: 0.73). Moreover, DLPM combined with clinical characteristics and stromal TILs achieved the highest AUC in the primary cohort (AUC: 0.82) and validation cohort 1 (AUC: 0.80). CONCLUSION Our study suggested that WSIs integrated with deep learning could potentially predict pCR to NAC in breast cancer. The predictive performance will be improved by combining clinical characteristics. DLPs from DLPM can provide more information compared to stromal TILs for pCR prediction.
Collapse
|
15
|
Li S, Zhang Y, Zhang P, Xue S, Chen Y, Sun L, Yang R. Predictive and prognostic values of tumor infiltrating lymphocytes in breast cancers treated with neoadjuvant chemotherapy: A meta-analysis. Breast 2022; 66:97-109. [PMID: 36219945 PMCID: PMC9550538 DOI: 10.1016/j.breast.2022.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/11/2022] [Accepted: 10/03/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND This meta-analysis assessed the predictive and prognostic value of tumor infiltrating lymphocytes (TILs) in neoadjuvant chemotherapy (NACT) treated breast cancer and an optimal threshold for predicting pathologic complete response (pCR). METHODS A systematic search of PubMed, EMBASE and Web of Science electronic databases was conducted to identify eligible studies published before April 2022. Either a fixed or random effects model was applied to estimate the pooled hazard ratio (HR) and odds ratio (OR) for prognosis and predictive values of TILs in breast cancer patients treated with NACT. The study is registered with PROSPERO (CRD42020221521). RESULTS A total of 29 published studies were eligible. Increased levels of TILs predicted response to NACT in HER2 positive breast cancer (OR = 2.54 95%CI, 1.50-4.29) and triple negative breast cancer (TNBC) (OR = 3.67, 95%CI, 1.93-6.97), but not for hormone receptor (HR) positive breast cancer (OR = 1.68, 95 %CI, 0.67-4.25). A threshold of 20% of H & E-stained TILs was associated with prediction of pCR in both HER2 positive breast cancer (P = 0.035) and TNBC (P = 0.001). Moreover, increased levels of TILs (either iTILs or sTILs) were associated with survival benefit in HER2-positive breast cancer and TNBC. However, an increased level of TILs was not a prognostic factor for survival in HR positive breast cancer (pooled HR = 0.64, 95%CI: 0.03-14.1, P = 0.78). CONCLUSIONS Increased levels of TILs were associated with increased rates of response to NACT and improved prognosis for the molecular subtypes of TNBC and HER2-positive breast cancer, but not for patients with HR positive breast cancer. A threshold of 20% TILs was the most powerful outcome prognosticator of pCR.
Collapse
Affiliation(s)
- Shiqi Li
- Department of Pharmacy Administration, School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Ying Zhang
- Department of Clinical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, China
| | - Peigen Zhang
- Department of Pharmacy Administration, School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Shuijing Xue
- Department of Pharmacy Administration, School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Yu Chen
- Department of Pharmacy Administration, School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Lihua Sun
- Department of Pharmacy Administration, School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China,Corresponding author. Department of pharmacy administration, School of Business Administration, Shenyang Pharmaceutical University, 103 Wen hua Road, Shenyang, 110016, Liaoning Province, PR China.
| | - Rui Yang
- Clinical Pharmacology Laboratory, The Second Affiliated Hospital, Liaoning University of Traditional Chinese Medicine, Shenyang, 110034, China
| |
Collapse
|
16
|
Ahmad MZ, Alasiri AS, Alasmary MY, Abdullah MM, Ahmad J, Abdel Wahab BA, M Alqahtani SA, Pathak K, Mustafa G, Khan MA, Saikia R, Gogoi U. Emerging advances in nanomedicine for breast cancer immunotherapy: opportunities and challenges. Immunotherapy 2022; 14:957-983. [PMID: 35852105 DOI: 10.2217/imt-2021-0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is one of the most common causes of cancer-related morbidity and mortality in women worldwide. Early diagnosis and an appropriate therapeutic approach for all cancers are climacterics for a favorable prognosis. Targeting the immune system in breast cancer is already a clinical reality with notable successes, specifically with checkpoint blockade antibodies and chimeric antigen receptor T-cell therapy. However, there have been inevitable setbacks in the clinical application of cancer immunotherapy, including inadequate immune responses due to insufficient delivery of immunostimulants to immune cells and uncontrolled immune system modulation. Rapid advancements and new evidence have suggested that nanomedicine-based immunotherapy may be a viable option for treating breast cancer.
Collapse
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Ali S Alasiri
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Mohammed Yahia Alasmary
- Medical Department, College of Medicine, Najran University, Najran, 11001, Kingdom of Saudi Arabia
| | - M M Abdullah
- Advanced Materials & Nano-Research Centre, Department of Physics, Faculty of Science & Arts, Najran University, Najran, 11001, Kingdom Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Basel A Abdel Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, 11001, Kingdom of Saudi Arabia
- Department of Pharmacology, College of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Saif Aboud M Alqahtani
- Internal Medicine Department, College of Medicine, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
| | - Kalyani Pathak
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Gulam Mustafa
- College of Pharmacy, Shaqra University, Ad-Dawadmi Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Ahmad Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Riya Saikia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Urvashee Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| |
Collapse
|
17
|
Huertas-Caro CA, Ramirez MA, Gonzalez-Torres HJ, Sanabria-Salas MC, Serrano-Gómez SJ. Immune Lymphocyte Infiltrate and its Prognostic Value in Triple-Negative Breast Cancer. Front Oncol 2022; 12:910976. [PMID: 35924147 PMCID: PMC9342669 DOI: 10.3389/fonc.2022.910976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) occurs more frequently in young (<50 years) non-Hispanic black and Hispanic/Latina women. It is considered the most aggressive subtype of breast cancer, although, recently, immune infiltrate has been associated with long-term survival, lower risk of death and recurrence, and response to neoadjuvant chemotherapy. The aim of this review was to evaluate the clinical impact of the immune infiltrate in TNBC by discussing whether its prognostic value varies across different populations. A comprehensive systematic search in databases such as PubMed and Web of Science was conducted to include papers focused on tumor-infiltrating lymphocytes (TILs) in TNBC in different population groups and that were published before January 2021. TNBC patients with higher levels of TILs had longer overall survival and disease-free survival times compared with TNBC patients with low TIL levels. Similar results were observed for CD4+, CD8+ TIL populations. On the other hand, patients with high TIL levels showed a higher rate of pathological complete response regardless of the population group (Asian, European, and American). These results altogether suggest that TIL subpopulations might have a prognostic role in TNBC, but the underlying mechanism needs to be elucidated. Although the prognosis value of TILs was not found different between the population groups analyzed in the revised literature, further studies including underrepresented populations with different genetic ancestries are still necessary to conclude in this regard.
Collapse
Affiliation(s)
| | - Mayra Alejandra Ramirez
- Grupo de investigación en biología del cáncer, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Henry J. Gonzalez-Torres
- Doctorado en Ciencias Biomédicas, Universidad del Valle, Cali, Colombia
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | | | - Silvia J. Serrano-Gómez
- Grupo de apoyo y seguimiento para la investigación, Instituto Nacional de Cancerología, Bogotá, Colombia
| |
Collapse
|
18
|
Candelaria RP, Spak DA, Rauch GM, Huo L, Bassett RL, Santiago L, Scoggins ME, Guirguis MS, Patel MM, Whitman GJ, Moulder SL, Thompson AM, Ravenberg EE, White JB, Abuhadra NK, Valero V, Litton J, Adrada BE, Yang WT. BI-RADS Ultrasound Lexicon Descriptors and Stromal Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer. Acad Radiol 2022; 29 Suppl 1:S35-S41. [PMID: 34272161 PMCID: PMC8755852 DOI: 10.1016/j.acra.2021.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Increased levels of stromal tumor-infiltrating lymphocytes (sTILs) have recently been considered a favorable independent prognostic and predictive biomarker in triple-negative breast cancer (TNBC). The purpose of this study was to determine the relationship between BI-RADS (Breast Imaging Reporting and Data System) ultrasound lexicon descriptors and sTILs in TNBC. MATERIALS AND METHODS Patients with stage I-III TNBC were evaluated within a single-institution neoadjuvant clinical trial. Two fellowship-trained breast radiologists used the BI-RADS ultrasound lexicon to assess pretreatment tumor shape, margin, echo pattern, orientation, posterior features, and vascularity. sTILs were defined as low <20 or high ≥20 on the pretreatment biopsy. Fisher's exact tests were used to assess the association between lexicon descriptors and sTIL levels. RESULTS The 284 patients (mean age 52 years, range 24-79 years) were comprised of 68% (193/284) with low-sTIL tumors and 32% (91/284) with high-sTIL tumors. TNBC tumors with high sTILs were more likely to have the following features: (1) oval/round shape than irregular shape (p = 0.003), (2) circumscribed or microlobulated margins than spiculated, indistinct, or angular margins (p = 0.0005); (3) complex cystic and solid pattern than heterogeneous pattern (p = 0.006); and (4) posterior enhancement than shadowing (p = 0.002). There was no significant association between sTILs and descriptors for orientation and vascularity (p = 0.06 and p = 0.49, respectively). CONCLUSION BI-RADS ultrasound descriptors of the pretreatment appearance of a TNBC tumor can be useful in discriminating between tumors with low and high sTIL levels. Therefore, there is a potential use of ultrasound tumor characteristics to complement sTILs when used as stratification factors in treatment algorithms for TNBC.
Collapse
|
19
|
Van Bockstal MR, François A, Altinay S, Arnould L, Balkenhol M, Broeckx G, Burguès O, Colpaert C, Dedeurwaerdere F, Dessauvagie B, Duwel V, Floris G, Fox S, Gerosa C, Hastir D, Jaffer S, Kurpershoek E, Lacroix-Triki M, Laka A, Lambein K, MacGrogan GM, Marchio C, Martinez MDM, Nofech-Mozes S, Peeters D, Ravarino A, Reisenbichler E, Resetkova E, Sanati S, Schelfhout AM, Schelfhout V, Shaaban A, Sinke R, Stanciu-Pop CM, van Deurzen CHM, Van de Vijver KK, Van Rompuy AS, Vincent-Salomon A, Wen H, Wong S, Bouzin C, Galant C. Interobserver variability in the assessment of stromal tumor-infiltrating lymphocytes (sTILs) in triple-negative invasive breast carcinoma influences the association with pathological complete response: the IVITA study. Mod Pathol 2021; 34:2130-2140. [PMID: 34218258 PMCID: PMC8595512 DOI: 10.1038/s41379-021-00865-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
High stromal tumor-infiltrating lymphocytes (sTILs) in triple-negative breast cancer (TNBC) are associated with pathological complete response (pCR) after neoadjuvant chemotherapy (NAC). Histopathological assessment of sTILs in TNBC biopsies is characterized by substantial interobserver variability, but it is unknown whether this affects its association with pCR. Here, we aimed to investigate the degree of interobserver variability in an international study, and its impact on the relationship between sTILs and pCR. Forty pathologists assessed sTILs as a percentage in digitalized biopsy slides, originating from 41 TNBC patients who were treated with NAC followed by surgery. Pathological response was quantified by the MD Anderson Residual Cancer Burden (RCB) score. Intraclass correlation coefficients (ICCs) were calculated per pathologist duo and Bland-Altman plots were constructed. The relation between sTILs and pCR or RCB class was investigated. The ICCs ranged from -0.376 to 0.947 (mean: 0.659), indicating substantial interobserver variability. Nevertheless, high sTILs scores were significantly associated with pCR for 36 participants (90%), and with RCB class for eight participants (20%). Post hoc sTILs cutoffs at 20% and 40% resulted in variable associations with pCR. The sTILs in TNBC with RCB-II and RCB-III were intermediate to those of RCB-0 and RCB-I, with lowest sTILs observed in RCB-I. However, the limited number of RCB-I cases precludes any definite conclusions due to lack of power, and this observation therefore requires further investigation. In conclusion, sTILs are a robust marker for pCR at the group level. However, if sTILs are to be used to guide the NAC scheme for individual patients, the observed interobserver variability might substantially affect the chance of obtaining a pCR. Future studies should determine the 'ideal' sTILs threshold, and attempt to fine-tune the patient selection for sTILs-based de-escalation of NAC regimens. At present, there is insufficient evidence for robust and reproducible sTILs-guided therapeutic decisions.
Collapse
Affiliation(s)
- Mieke R. Van Bockstal
- Department of pathology, Cliniques universitaires Saint-Luc Bruxelles, Avenue Hippocrate 10, Woluwé-Saint-Lambert 1200, Belgium
| | - Aline François
- Department of pathology, Cliniques universitaires Saint-Luc Bruxelles, Avenue Hippocrate 10, Woluwé-Saint-Lambert 1200, Belgium
| | - Serdar Altinay
- Department of Pathology, University of Health Sciences, Bakirköy Dr. Sadi Konuk Health Application and Research Center, 34147 Istanbul, Turkey
| | - Laurent Arnould
- Département de Biologie et de Pathologie des Tumeurs, Centre George-François Leclerc, 1 Rue Pr. Marion, 21000 Dijon, France
| | - Maschenka Balkenhol
- Department of Pathology, Radboud University Medical Center, PO Box 9100, 6500, HB Nijmegen, The Netherlands
| | - Glenn Broeckx
- Department of Pathology, University Hospital Antwerp, Drie Eikenstraat 655, 2650 Edegem, Belgium
| | - Octavio Burguès
- Department of Pathology, Hospital Clínico Universitario de Valencia, Av. De Blasco Ibáñez 17, 46010 València, Valencia, Spain
| | - Cecile Colpaert
- Department of Pathology, AZ Turnhout Campus Sint-Jozef, Steenweg op Merksplas 44, 2300 Turnhout, Belgium
| | | | - Benjamin Dessauvagie
- Division of Pathology and Laboratory Medicine, Medical School, The University of Western Australia, Crawley, WA 6009, Australia,Anatomical Pathology, PathWest Laboratory Medicine WA, Perth, Australia
| | - Valérie Duwel
- Department of pathology, AZ Klina Brasschaat, Augustijnslei 100, 2930 Brasschaat, Belgium
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, KU Leuven – University of Leuven, Herestraat 49, 3000 Leuven, Belgium,Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven – University of Leuven, Leuven, Belgium
| | - Stephen Fox
- Department of Pathology, Peter MacCallum Cancer Center and the University of Melbourne, Melbourne, Vic 3000, Australia
| | - Clara Gerosa
- Department of Pathology, University of Cagliari, AOU San Giovanni di Dio, Via Ospedale 54, 09124 Cagliari, Italy
| | - Delfyne Hastir
- Institute of Pathology, Lausanne University Hospital, Rue du Bugnon 25, CH-1011 Lausanne, Switzerland
| | - Shabnam Jaffer
- Department of Pathology, Mount Sinai Hospital and Icahn School of Medicine, New York, New York, NY10029 USA
| | | | - Magali Lacroix-Triki
- Department of Pathology, Gustave-Roussy Cancer Campus, 114 Rue Edouard-Vaillant, 94805 Villejuif, France
| | - Andoni Laka
- Department of Pathology, Clinique Notre-Dame de Grâce (CNDG), Chaussée de Nivelles 212, 6041 Gosselies, Belgium
| | - Kathleen Lambein
- Department of Pathology, AZ St Lucas Hospital, Groenebriel 1, 9000 Ghent, Belgium
| | - Gaëtan Marie MacGrogan
- Surgical Pathology Unit, Department of Pathobiology, Institut Bergonié, F-33076 Bordeaux, France
| | - Caterina Marchio
- Department of Medical Sciences, University of Turin, 10126 Torino, Italy,Pathology Unit, FPO-IRCCS, Candiolo Cancer Institute, Candiolo, Italy
| | | | - Sharon Nofech-Mozes
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Ontario, ON M4N 3M5, Canada
| | - Dieter Peeters
- Department of Pathology, AZ St Maarten, Liersesteenweg 435, 2800 Mechelen, Belgium,Histopathology, Imaging and Quantification Unit, HistoGeneX, Sint-Bavostraat 78, 2610 Antwerp, Belgium
| | - Alberto Ravarino
- Department of Pathology, University of Cagliari, AOU San Giovanni di Dio, Via Ospedale 54, 09124 Cagliari, Italy
| | - Emily Reisenbichler
- Department of Pathology, Yale School of Medicine, Yale New Haven Hospital, 310 Cedar Street, New Haven, CT06510, United States
| | - Erika Resetkova
- The University of Texas MD Anderson Cancer Center, Houston TX77030, Texas, USA
| | - Souzan Sanati
- Department of Pathology and Lab Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Rm8612, Los Angeles, CA90048, United States
| | - Anne-Marie Schelfhout
- Department of Pathology, Onze-Lieve-Vrouwziekenhuis Aalst, Moorselbaan 164, 9300 Aalst, Belgium
| | - Vera Schelfhout
- Department of Pathology, AZ St Maarten, Liersesteenweg 435, 2800 Mechelen, Belgium
| | - Abeer Shaaban
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, University of Birmingham, Birmingham B15 2GW, United Kingdom
| | - Renata Sinke
- Pathan BV, Kleiweg 500, 3045 PM Rotterdam, The Netherlands
| | - Claudia M Stanciu-Pop
- Department of Pathology, CHU UCL Namur, Site Godinne, Avenue Docteur G. Thérasse 1, 5530 Yvoir, Belgium
| | - Carolien HM van Deurzen
- Department of Pathology, Erasmus Medical Center Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Koen K Van de Vijver
- Department of Pathology, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Anne-Sophie Van Rompuy
- Department of Pathology, University Hospitals Leuven, KU Leuven – University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Anne Vincent-Salomon
- Pôle de Médicine Diagnostique & Théranostique, INSERM U934, Institut Curie, 26 Rue d’Ulm, 75248 Paris Cedex 05, France
| | - Hannah Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Serena Wong
- Department of Pathology, Yale School of Medicine, Yale New Haven Hospital, 310 Cedar Street, New Haven, CT06510, United States
| | - Caroline Bouzin
- 2IP IREC Imaging Platform, Institute of Clinical and Experimental Research (IREC), Université catholique de Louvain, Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Christine Galant
- Department of pathology, Cliniques universitaires Saint-Luc Bruxelles, Avenue Hippocrate 10, Woluwé-Saint-Lambert 1200, Belgium,Institute of Clinical and Experimental Research (IREC), Université catholique de Louvain, Avenue Hippocrate 55, 1200 Brussels, Belgium
| |
Collapse
|
20
|
Lusho S, Durando X, Mouret-Reynier MA, Kossai M, Lacrampe N, Molnar I, Penault-Llorca F, Radosevic-Robin N, Abrial C. Platelet-to-Lymphocyte Ratio Is Associated With Favorable Response to Neoadjuvant Chemotherapy in Triple Negative Breast Cancer: A Study on 120 Patients. Front Oncol 2021; 11:678315. [PMID: 34367964 PMCID: PMC8331686 DOI: 10.3389/fonc.2021.678315] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/29/2021] [Indexed: 12/31/2022] Open
Abstract
Introduction Triple negative breast cancer (TNBC) is highly heterogeneous, but still most of the patients are treated by the anthracycline/taxane-based neoadjuvant therapy (NACT). Tumor-infiltrating lymphocytes (TILs) are a strong predictive and prognostic biomarker in TNBC, however are not always available. Peripheral blood counts, which reflect the systemic inflammatory/immune status, are easier to obtain than TILs. We investigated whether baseline white cell or platelet counts, as well as, Neutrophil-to-Lymphocyte Ratio (NLR) or Platelet-to-Lymphocyte Ratio (PLR) could replace baseline TILs as predictive or prognostic biomarkers in a series of TNBC treated by standard NACT. Patients and Methods One hundred twenty patients uniformly treated by FEC/taxane NACT in a tertiary cancer care center were retrospectively analyzed. The presence of pathological complete response (pCR: ypT0/Tis, ypN0) or the presence of pCR and/small residual disease (ypT0/Tis/T1ab, ypN0) were considered as good responses in data analysis. Baseline/pre-NACT blood count, NLR, PLR and TILs were evaluated as predictors of response, distant recurrence rate and distant recurrence-free survival (DRFS). Results TILs ≥30% and ≥1.5% were best predictors of pCR and distant recurrence risk, respectively (p = 0.007, p = 0.012). However, in this cohort, pCR status was not significantly associated with recurrence. Only the ensemble of patients with pCR and small residual disease had lower recurrence risk and longer survival DRFS (p = 0.042, p = 0.024, respectively) than the rest of the cohort (larger residual disease). The only parameter which could predict the pCR/small residual disease status was PLR: patients with values lower than 133.25 had significantly higher chance of reaching that status after NACT (p = 0.045). However, no direct correlation could be established between baseline PLR and metastatic recurrence. No correlation either was found between TIL and individual blood counts, or between TILs and NLR or PLR. Conclusion In this cohort, TILs retained their pCR predictive value; however PLR was a better predictor of the ensemble of responses which had good outcome in terms of less distant recurrences or longer DRFS (pCR or small residual disease). Thus, baseline PLR is worth further, prospective investigation together with baseline TILs, as it might indicate a good TNBC response to NACT when TILs are unavailable.
Collapse
Affiliation(s)
- Sejdi Lusho
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Delegation for Clinical Research and Innovation, Centre Jean Perrin, Clermont-Ferrand, France.,Centre for Clinical Investigation, INSERM U501, Clermont-Ferrand, France
| | - Xavier Durando
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Delegation for Clinical Research and Innovation, Centre Jean Perrin, Clermont-Ferrand, France.,Centre for Clinical Investigation, INSERM U501, Clermont-Ferrand, France.,Department of Medical Oncology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Marie-Ange Mouret-Reynier
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Delegation for Clinical Research and Innovation, Centre Jean Perrin, Clermont-Ferrand, France.,Centre for Clinical Investigation, INSERM U501, Clermont-Ferrand, France.,Department of Medical Oncology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Myriam Kossai
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Department of Pathology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Nathalie Lacrampe
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Department of Pathology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Ioana Molnar
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Delegation for Clinical Research and Innovation, Centre Jean Perrin, Clermont-Ferrand, France.,Centre for Clinical Investigation, INSERM U501, Clermont-Ferrand, France
| | - Frederique Penault-Llorca
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France
| | - Nina Radosevic-Robin
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Department of Pathology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Catherine Abrial
- Clermont Auvergne University, INSERM U1240 "Molecular Imaging and Theranostic Strategies", Centre Jean Perrin, Clermont-Ferrand, France.,Delegation for Clinical Research and Innovation, Centre Jean Perrin, Clermont-Ferrand, France.,Centre for Clinical Investigation, INSERM U501, Clermont-Ferrand, France
| |
Collapse
|
21
|
Zormpas-Petridis K, Noguera R, Ivankovic DK, Roxanis I, Jamin Y, Yuan Y. SuperHistopath: A Deep Learning Pipeline for Mapping Tumor Heterogeneity on Low-Resolution Whole-Slide Digital Histopathology Images. Front Oncol 2021; 10:586292. [PMID: 33552964 PMCID: PMC7855703 DOI: 10.3389/fonc.2020.586292] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/30/2020] [Indexed: 12/27/2022] Open
Abstract
High computational cost associated with digital pathology image analysis approaches is a challenge towards their translation in routine pathology clinic. Here, we propose a computationally efficient framework (SuperHistopath), designed to map global context features reflecting the rich tumor morphological heterogeneity. SuperHistopath efficiently combines i) a segmentation approach using the linear iterative clustering (SLIC) superpixels algorithm applied directly on the whole-slide images at low resolution (5x magnification) to adhere to region boundaries and form homogeneous spatial units at tissue-level, followed by ii) classification of superpixels using a convolution neural network (CNN). To demonstrate how versatile SuperHistopath was in accomplishing histopathology tasks, we classified tumor tissue, stroma, necrosis, lymphocytes clusters, differentiating regions, fat, hemorrhage and normal tissue, in 127 melanomas, 23 triple-negative breast cancers, and 73 samples from transgenic mouse models of high-risk childhood neuroblastoma with high accuracy (98.8%, 93.1% and 98.3% respectively). Furthermore, SuperHistopath enabled discovery of significant differences in tumor phenotype of neuroblastoma mouse models emulating genomic variants of high-risk disease, and stratification of melanoma patients (high ratio of lymphocyte-to-tumor superpixels (p = 0.015) and low stroma-to-tumor ratio (p = 0.028) were associated with a favorable prognosis). Finally, SuperHistopath is efficient for annotation of ground-truth datasets (as there is no need of boundary delineation), training and application (~5 min for classifying a whole-slide image and as low as ~30 min for network training). These attributes make SuperHistopath particularly attractive for research in rich datasets and could also facilitate its adoption in the clinic to accelerate pathologist workflow with the quantification of phenotypes, predictive/prognosis markers.
Collapse
Affiliation(s)
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia-INCLIVA Biomedical Health Research Institute, Valencia, Spain.,Low Prevalence Tumors, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Ioannis Roxanis
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Yann Jamin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Yinyin Yuan
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
22
|
Gao ZH, Li CX, Liu M, Jiang JY. Predictive and prognostic role of tumour-infiltrating lymphocytes in breast cancer patients with different molecular subtypes: a meta-analysis. BMC Cancer 2020; 20:1150. [PMID: 33238978 PMCID: PMC7690150 DOI: 10.1186/s12885-020-07654-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Whether tumour-infiltrating lymphocytes (TILs) play different roles in different molecular subtypes of breast cancer remains unknown. Additionally, their prognostic and predictive value in different molecular subtypes of breast cancer is still controversial. The aim of our meta-analysis was to assess the prognostic and predictive value of TILs in different molecular subtypes of breast cancer by summarizing all relevant studies performing multivariate analysis. METHODS PubMed, Embase, EBSCO, ScienceDirect, the Cochrane Database and Web of Science were comprehensively searched (until March 2020). Hazard ratios (HRs), odds ratios (ORs) and their 95% confidence intervals (CIs) were used as effect measures to perform our meta-analysis. A random effect model was used. Stata software, version 15 (2017) (StataCorp, College Station, TX, USA) was used to perform the statistical analysis. RESULTS Thirty-three studies including 18,170 eligible breast cancer patients were analysed. The meta-analysis showed that high TIL expression was significantly associated with increased pathological complete response (pCR) rates after neoadjuvant chemotherapy in patients with the HER2-enriched molecular subtype (OR = 1.137, 95% CI [1.061 ~ 1.218], p < 0.001) and triple-negative breast cancer (TNBC) subtype (OR = 1.120, 95% CI [1.061 ~ 1.182], p < 0.001). However, high TIL expression was not significantly associated with high pCR rates after neoadjuvant chemotherapy in patients with the luminal molecular subtype of breast cancer (OR = 1.154, 95% CI [0.789 ~ 1.690], p = 0.460). We carried out a meta-analysis on the HRs of overall survival (OS) and disease-free survival (DFS) to assess the prognostic value of TILs in breast cancer with different molecular subtypes more deeply. Our meta-analysis confirmed that high TILs were associated with significantly improved DFS in patients with the HER2-enriched molecular subtype [HR = 0.940, 95% CI (0.903 ~ 0.979), p = 0.003] and TNBC molecular subtype [HR = 0.907, 95% CI (0.862 ~ 0.954), p < 0.001]. However, high TILs were not associated with significantly better DFS in patients with the luminal molecular subtype of breast cancer [HR = 0.998, 95% CI (0.977 ~ 1.019), p = 0.840]. Furthermore, the results confirmed that high TILs were significantly related to better OS in patients with the HER2-enriched molecular subtype [HR = 0.910, 95% CI (0.866 ~ 0.957), p < 0.001] and TNBC molecular subtype [HR = 0.869, 95% CI (0.836 ~ 0.904), p < 0.001]. Conversely, the summarized results indicated that high TILs were significantly associated with poor OS in patients with the luminal molecular subtype of breast cancer [HR = 1.077, 95% CI (1.016 ~ 1.141), p = 0.012]. CONCLUSIONS Our meta-analysis confirms that high TILs are associated with favourable survival and predicts pCR in breast cancer patients with the TNBC and HER2-enriched molecular subtypes.
Collapse
Affiliation(s)
- Zhao-Hua Gao
- Department of Breast Surgery, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, P.R. China. .,Department of Breast Surgery, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, P.R. China. .,Department of Breast Surgery, Dalian Medical University Clinical Oncology College, Shenyang, Liaoning, 110042, P.R. China.
| | - Cun-Xin Li
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, P.R. China
| | - Ming Liu
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, P.R. China
| | - Jia-Yuan Jiang
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, P.R. China
| |
Collapse
|
23
|
Tinholt M, Stavik B, Tekpli X, Garred Ø, Borgen E, Kristensen V, Sahlberg KK, Sandset PM, Iversen N. Coagulation factor V is a marker of tumor-infiltrating immune cells in breast cancer. Oncoimmunology 2020; 9:1824644. [PMID: 33457104 PMCID: PMC7781787 DOI: 10.1080/2162402x.2020.1824644] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/14/2020] [Indexed: 01/06/2023] Open
Abstract
Background Factor (F) V is an essential cofactor in blood coagulation, however, F5 expression in breast tumors has also been linked to tumor aggressiveness and overall survival. The specific role of FV in breast cancer is yet unknown. We therefore aimed at dissecting the biological relevance of FV in breast cancer. Methods Gene expression data from a Scandinavian breast cancer cohort (n = 363) and the cancer genome atlas (TCGA) (n = 981) and 12 replication cohorts were used to search for F5 co-expressed genes, followed by gene ontology analysis. Pathological and bioinformatic tools were used to evaluate immune cell infiltration and tumor purity. T cell activation, proliferation and migration were studied in FV treated Jurkat T cells. Results F5 co-expressed genes were mainly associated with immune system processes and cell activation. Tumors with high expression of F5 were more infiltrated with both lymphoid (T cells, NK cells, and B cells) and myeloid cells (macrophages and dendritic cells), and F5 expression was negatively correlated with tumor purity (ρ = -0.32). Confirming a prognostic role, data from the Kaplan-Meier plotter showed that high F5 expression was associated with improved relapse-free survival. The strongest association was observed in basal-like breast cancer (HR = 0.55; 95% CI, 0.42-0.71). Exogenous FV did not substantially affect activation, proliferation or migration of human T cells. Conclusions F5 was identified as a novel marker of immune cell infiltration in breast cancer, and the prognostic role of F5 was verified. FV emerge as an interesting immunological biomarker with potential therapeutic relevance for the cancer-inflammation-thrombosis circuit.
Collapse
Affiliation(s)
- Mari Tinholt
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Benedicte Stavik
- Department of Haematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Norway
| | - Xavier Tekpli
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Vessela Kristensen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology (Epigen), Akershus University Hospital, Lillestrøm, Norway
| | - Kristine Kleivi Sahlberg
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Department of Research, Vestre Viken, Drammen, Norway
| | - Per Morten Sandset
- Department of Haematology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nina Iversen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
24
|
Piña-Sánchez P, Valdez-Salazar HA, Ruiz-Tachiquín ME. Circulating microRNAs and their role in the immune response in triple-negative breast cancer. Oncol Lett 2020; 20:224. [PMID: 32968446 PMCID: PMC7499949 DOI: 10.3892/ol.2020.12087] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 07/29/2020] [Indexed: 01/01/2023] Open
Abstract
Breast cancer (BC) is the most common type of cancer in women worldwide, and despite advances in treatments, its incidence and mortality are increasing. Therefore, it is necessary to develop new, non-invasive tests that provide more accurate diagnosis and prognosis in a timely manner. A promising approach is measuring the presence of biomarkers to detect tumors at various stages and determine their specific characteristics, thus allowing for more personalized treatment. MicroRNAs (miRNAs) serve a role in gene expression, primarily by interacting with messenger RNAs, and may be potential biomarkers for detecting cancer. They are detectable in tissues and blood, including plasma and/or serum, are stable and often tumor specific. Also, different miRNAs are associated with specific BC molecular subtypes. Triple-negative BC (TNBC) is a type of BC in which the primary targets for hormonal therapy are absent. It is an aggressive phenotype, which frequently metastasizes and is associated with an unfavorable prognosis. The present review focuses on circulating miRNAs in patients with TNBC, with an emphasis on their interaction with the immune response checkpoint genes PD-1, PD-L1 and CTLA4. Modulation and response of the immune system are of interest in cancer treatment due to the success of immunotherapy in the treatment of various neoplasms. Based on the findings of this literature review and the in silico analysis performed as part of this review, it is concluded that circulating hsa-miR-195 and hsa-miR-155 in TNBC interact with checkpoint genes involved in the immune response. Further analysis of the expression of these circulating miRNAs and their association with prognosis in patients with TNBC treated with immunotherapy should be assessed to evaluate their possible use as non-invasive predictive biomarkers. In addition, functional studies to analyze biologically relevant targets in the development and prognosis of TNBC, which could be therapeutic targets, are also recommended.
Collapse
Affiliation(s)
- Patricia Piña-Sánchez
- Oncological Diseases Medical Research Unit, Oncology Hospital, XXI Century National Medical Center, Mexican Institute of Social Security (IMSS), Mexico City 06720, Mexico
| | - Hilda-Alicia Valdez-Salazar
- Infectious and Parasitic Diseases Medical Research Unit, Pediatrics Hospital 'Dr. Silvestre Frenk Freund', XXI Century National Medical Center, Mexican Institute of Social Security (IMSS), Mexico City 06720, Mexico
| | - Martha-Eugenia Ruiz-Tachiquín
- Oncological Diseases Medical Research Unit, Oncology Hospital, XXI Century National Medical Center, Mexican Institute of Social Security (IMSS), Mexico City 06720, Mexico
| |
Collapse
|
25
|
Dediu M, Zielinski C. Reply to the Correspondence by Untch et al. "Concerning Dediu M, Zielinski C: A Proposal to Redefine Pathologic Complete Remission as Endpoint following Neoadjuvant Chemotherapy in Early Breast Cancer" [Breast Care 2019; DOI 10.1159/000500624]. Breast Care (Basel) 2020; 15:314-316. [PMID: 32774227 DOI: 10.1159/000502508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/05/2019] [Indexed: 11/19/2022] Open
Affiliation(s)
| | - Christoph Zielinski
- Vienna Cancer Center, Vienna Hospital Association and Medical University Vienna, Vienna, Austria
| |
Collapse
|
26
|
Wei L, Wu N, Wei F, Li F, Zhang Y, Liu J, Ren X. Prognosis significance of indoleamine 2, 3-dioxygenase, programmed death ligand-1 and tumor-infiltrating immune cells in microenvironment of breast cancer. Int Immunopharmacol 2020; 84:106506. [PMID: 32330866 DOI: 10.1016/j.intimp.2020.106506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The immune microenvironment plays an increasingly important role in predicting the prognosis of multiple tumors and selecting patients for immunotherapy trials. We studied the expression of indoleamine 2, 3-dioxygenase (IDO) and programmed death ligand-1 (PD-L1), detected the proportion of tumor-infiltrating immune cells (TIIs), and further analyzed the association of these immunological characteristics with the clinicopathological parameters and prognosis of breast cancer patients. METHODS Immunohistochemical staining for IDO, PD-L1, CD4, CD8, Foxp3, CD20, CD56 and CD68 expression in breast cancer tissues was carried out. IDO and PD-L1 expression were scored by extent in tumor cells. TIIs expressing CD4, CD8, Foxp3, CD20, CD56 or CD68 were evaluated by positive count. Clinicopathological characteristics and follow-up were recorded. RESULTS The frequencies of IDO-high-expressing and PD-L1-expressing tissue were 33.77% and 24.68%, respectively. The co-expression of IDO and PD-L1 was identified in 16/77 (20.78%) of cases. IDO high expression, CD4+ T cells and CD56+ cells were most frequently observed in patients with tumor-draining lymph nodes(TDLNs) metastasis. Immune cells were more common in non-luminal breast cancer than in luminal breast cancer. In survival analysis, PFS were not associated with high levels of IDO and PD-L1, nor were TIIs. However, CD20 and CD68 were significant risk factors for prognostic after adjusting covariates by COX regression. IDOhighFoxp3highT patients had a tendency with shorter progression-free survival. CONCLUSIONS Although we found a limited prognostic effect of TIIs on survival in breast cancer patients, IDO combined with TIIs can help to evaluate the prognosis of patients.
Collapse
Affiliation(s)
- Lijuan Wei
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin Clinical Research Center for Cancer, China
| | - Nan Wu
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin Clinical Research Center for Cancer, China
| | - Feng Wei
- National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin Clinical Research Center for Cancer, China; Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fangxuan Li
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin Clinical Research Center for Cancer, China
| | - Yanhui Zhang
- National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin Clinical Research Center for Cancer, China; Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Juntian Liu
- Department of Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin Clinical Research Center for Cancer, China.
| | - Xiubao Ren
- National Clinical Research Center for Cancer, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin Clinical Research Center for Cancer, China; Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
27
|
Goldsberry WN, Meza-Perez S, Londoño AI, Katre AA, Mott BT, Roane BM, Goel N, Wall JA, Cooper SJ, Norian LA, Randall TD, Birrer MJ, Arend RC. Inhibiting WNT Ligand Production for Improved Immune Recognition in the Ovarian Tumor Microenvironment. Cancers (Basel) 2020; 12:cancers12030766. [PMID: 32213921 PMCID: PMC7140065 DOI: 10.3390/cancers12030766] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/21/2022] Open
Abstract
In ovarian cancer, upregulation of the Wnt/β–catenin pathway leads to chemoresistance and correlates with T cell exclusion from the tumor microenvironment (TME). Our objectives were to validate these findings in an independent cohort of ovarian cancer subjects and determine whether inhibiting the Wnt pathway in a syngeneic ovarian cancer murine model could create a more T-cell-inflamed TME, which would lead to decreased tumor growth and improved survival. We preformed RNA sequencing in a cohort of human high grade serous ovarian carcinoma subjects. We used CGX1321, an inhibitor to the porcupine (PORCN) enzyme that is necessary for secretion of WNT ligand, in mice with established ID8 tumors, a murine ovarian cancer cell line. In order to investigate the effect of decreased Wnt/β–catenin pathway activity in the dendritic cells (DCs), we injected ID8 cells in mice that lacked β–catenin specifically in DCs. Furthermore, to understand how much the effects of blocking the Wnt/β–catenin pathway are dependent on CD8+ T cells, we injected ID8 cells into mice with CD8+ T cell depletion. We confirmed a negative correlation between Wnt activity and T cell signature in our cohort. Decreasing WNT ligand production resulted in increases in T cell, macrophage and dendritic cell functions, decreased tumor burden and improved survival. Reduced tumor growth was found in mice that lacked β–catenin specifically in DCs. When CD8+ T cells were depleted, CGX1321 treatment did not have the same magnitude of effect on tumor growth. Our investigation confirmed an increase in Wnt activity correlated with a decreased T-cell-inflamed environment; a relationship that was further supported in our pre-clinical model that suggests inhibiting the Wnt/β–catenin pathway was associated with decreased tumor growth and improved survival via a partial dependence on CD8+ T cells.
Collapse
Affiliation(s)
- Whitney N. Goldsberry
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
- Correspondence:
| | - Selene Meza-Perez
- Division of Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.M.-P.); (T.D.R.)
| | - Angelina I. Londoño
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
| | - Ashwini A. Katre
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
| | - Bryan T. Mott
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
| | - Brandon M. Roane
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
| | - Nidhi Goel
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
| | - Jaclyn A. Wall
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
| | - Sara J. Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA;
| | - Lyse A. Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Troy D. Randall
- Division of Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.M.-P.); (T.D.R.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael J. Birrer
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rebecca C. Arend
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (A.I.L.); (A.A.K.); (B.T.M.); (B.M.R.); (N.G.); (J.A.W.); (M.J.B.); (R.C.A.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
28
|
Prognostic value of tumor-infiltrating lymphocytes in patients with triple-negative breast cancer: a systematic review and meta-analysis. BMC Cancer 2020; 20:179. [PMID: 32131780 PMCID: PMC7057662 DOI: 10.1186/s12885-020-6668-z] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background The objective of this systematic review and meta-analysis was to determine the prognostic value of total tumor-infiltrating lymphocytes (TILs) and subtypes of TILs (CD4+, CD8+, and FOXP3+) in triple-negative breast cancer (TNBC). Methods A systematic search of the MEDLINE, EMBASE, and Web of Science databases was conducted to identified eligible articles published before August 2019. Study screening, data extraction, and risk of bias assessment were performed by two independent reviewers. Risk of bias on the study level was assessed using the ROBINS I tool and Quality in Prognosis Studies (QUIPS) tool. We performed a meta-analysis to obtain a pooled estimate of the prognostic role of TILs using Review Manager 5.3. Results In total, 37 studies were included in the final analysis. Compared to TNBC patients with low TIL levels, TNBC patients with high TIL levels showed a higher rate of pathological complete response (pCR) to treatment (odds ratio [OR] 2.14, 95% confidence interval [CI] 1.43–3.19). With each 10% increase in percentage of TILs, patients with TNBC had an increased pCR (OR 1.09, 95% CI 1.02–1.16). Compared to TNBC patients with low TIL levels, patients with high TIL levels had better overall survival (OS; hazard ratio [HR] 0.58, 95% CI 0.48–0.71) and disease-free survival (DFS; HR 0.66, 95% CI 0.57–0.76). Additionally, with a continuous increase in TIL levels, patients with TNBC had improved OS (HR 0.90, 95% CI 0.87–0.93) and DFS (HR 0.92, 95% CI 0.90–0.95). A high CD4+ TIL level was associated with better OS (HR 0.49, 95% CI 0.32–0.76) and DFS (HR 0.54, 95% CI 0.36–0.80). A high CD8+ TIL level was associated better DFS only (HR 0.55, 95% CI 0.38–0.81), as no statistical association was found with OS (HR 0.70, 95% CI 0.46–1.06). A high FOXP3+ TIL level also was associated with only DFS (HR 0.50, 95% CI 0.33–0.75) and not OS (HR 1.28, 95% CI 0.24–6.88). Conclusions TNBC with a high level of TILs showed better short-term and long-term prognoses. High levels of specific phenotypes of TILs (CD4+, CD8+, and FOXP3+) were predictive of a positive long-term prognosis for TNBC.
Collapse
|
29
|
Badr N, Berditchevski F, Shaaban A. The Immune Microenvironment in Breast Carcinoma: Predictive and Prognostic Role in the Neoadjuvant Setting. Pathobiology 2019; 87:61-74. [DOI: 10.1159/000504055] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 10/09/2019] [Indexed: 11/19/2022] Open
|
30
|
Baker GM, King TA, Schnitt SJ. Evaluation of Breast and Axillary Lymph Node Specimens in Breast Cancer Patients Treated With Neoadjuvant Systemic Therapy. Adv Anat Pathol 2019; 26:221-234. [PMID: 31149907 DOI: 10.1097/pap.0000000000000237] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Breast and axillary lymph node specimens from breast cancer patients treated with neoadjuvant systemic therapy are being encountered by pathologists with increasing frequency. Evaluation of these specimens presents challenges that differ from those encountered during the examination of other types of breast specimens. This article reviews the key issues regarding the gross and microscopic evaluation of post-neoadjuvant systemic therapy breast and lymph node specimens, and emphasizes the importance of accurate specimen evaluation in assessing treatment response.
Collapse
|
31
|
Goldsberry WN, Londoño A, Randall TD, Norian LA, Arend RC. A Review of the Role of Wnt in Cancer Immunomodulation. Cancers (Basel) 2019; 11:cancers11060771. [PMID: 31167446 PMCID: PMC6628296 DOI: 10.3390/cancers11060771] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/17/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Alterations in the Wnt signaling pathway are associated with the advancement of cancers; however, the exact mechanisms responsible remain largely unknown. It has recently been established that heightened intratumoral Wnt signaling correlates with tumor immunomodulation and immune suppression, which likely contribute to the decreased efficacy of multiple cancer therapeutics. Here, we review available literature pertaining to connections between Wnt pathway activation in the tumor microenvironment and local immunomodulation. We focus specifically on preclinical and clinical data supporting the hypothesis that strategies targeting Wnt signaling could act as adjuncts for cancer therapy, either in combination with chemotherapy or immunotherapy, in a variety of tumor types.
Collapse
Affiliation(s)
- Whitney N Goldsberry
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Angelina Londoño
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Troy D Randall
- Division of Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Lyse A Norian
- Department of Nutritional Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Rebecca C Arend
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
32
|
Caziuc A, Schlanger D, Amarinei G, Dindelegan GC. Can Tumor-Infiltrating Lymphocytes (TILs) Be a Predictive Factor for Lymph Nodes Status in Both Early Stage and Locally Advanced Breast Cancer? J Clin Med 2019; 8:jcm8040545. [PMID: 31013579 PMCID: PMC6517873 DOI: 10.3390/jcm8040545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/18/2019] [Accepted: 04/19/2019] [Indexed: 12/25/2022] Open
Abstract
The status of axillary lymph nodes is an important prognostic factor in the outcome of breast cancer tumors. New trials changed the attitude towards axillary clearance. In the era of development of new immune therapies for breast cancer, it is important to identify a biomarker that can predict lymph node status. Tumor-infiltrating lymphocytes (TILs) are a valuable indicator of the immune microenvironment that plays the central role in new anticancer drugs. Although the correlation between TILs and response to chemotherapy was established by previous studies, our retrospective study investigated the correlation between TILs and lymph node status. We analyzed data on 172 patients. According to stage, patients were divided in two groups: patients who underwent primary surgical treatment (breast-conserving or mastectomy and sentinel lymph node (SLN) biopsy +/− axillary clearance in conformity with lymph node status) and patients who received chemotherapy prior to surgical treatment (breast-conserving or mastectomy + axillary clearance). We showed a good inverse correlation between TILs and lymph nodes status for both early stage and locally advanced breast cancers. Moreover, TILs are a predictor for positive lymph nodes in the axilla in patients undergoing axillary clearance after SLN biopsy, with no statistical difference between the intrinsic or histological subtype of breast cancers. We also obtained a significant correlation between TILs and response to chemotherapy with no significative difference according to histological subtype. Although further data have still to be gathered before meeting the criteria for clinical utility, this study demonstrates that TILs are one of the most accredited forthcoming biomarkers for breast cancer (BC) patients.
Collapse
Affiliation(s)
- Alexandra Caziuc
- First Surgical Clinic, University of Medicine and Pharmacy "Iuliu Hatieganu", Victor Babes Street, no. 8, 400000 Cluj-Napoca, Romania.
| | - Diana Schlanger
- First Surgical Clinic, University of Medicine and Pharmacy "Iuliu Hatieganu", Victor Babes Street, no. 8, 400000 Cluj-Napoca, Romania.
| | - Giorgiana Amarinei
- First Surgical Clinic, University of Medicine and Pharmacy "Iuliu Hatieganu", Victor Babes Street, no. 8, 400000 Cluj-Napoca, Romania.
| | - George Calin Dindelegan
- First Surgical Clinic, University of Medicine and Pharmacy "Iuliu Hatieganu", Victor Babes Street, no. 8, 400000 Cluj-Napoca, Romania.
| |
Collapse
|