1
|
Loizides S, Constantinidou A. Triple negative breast cancer: Immunogenicity, tumor microenvironment, and immunotherapy. Front Genet 2023; 13:1095839. [PMID: 36712858 PMCID: PMC9879323 DOI: 10.3389/fgene.2022.1095839] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a biologically diverse subtype of breast cancer characterized by genomic and transcriptional heterogeneity and exhibiting aggressive clinical behaviour and poor prognosis. In recent years, emphasis has been placed on the identification of mechanisms underlying the complex genomic and biological profile of TNBC, aiming to tailor treatment strategies. High immunogenicity, specific immune activation signatures, higher expression of immunosuppressive genes and higher levels of stromal Tumor Infiltrating Lymphocytes, constitute some of the key elements of the immune driven landscape associated with TNBC. The unprecedented response of TNBC to immunotherapy has undoubtedly changed the standard of care in this disease both in the early and the metastatic setting. However, the extent of interplay between immune infiltration and mutational signatures in TNBC is yet to be fully unravelled. In the present review, we present clinical evidence on the immunogenicity and tumour microenvironment influence on TNBC progression and the current treatment paradigms in TNBC based on immunotherapy.
Collapse
Affiliation(s)
- Sotiris Loizides
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
| | - Anastasia Constantinidou
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
- Medical School, University of Cyprus, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
| |
Collapse
|
2
|
Cook D, Biancalana M, Liadis N, Lopez Ramos D, Zhang Y, Patel S, Peterson JR, Pfeiffer JR, Cole JA, Antony AK. Next generation immuno-oncology tumor profiling using a rapid, non-invasive, computational biophysics biomarker in early-stage breast cancer. Front Artif Intell 2023; 6:1153083. [PMID: 37138891 PMCID: PMC10149754 DOI: 10.3389/frai.2023.1153083] [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: 01/28/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Background Immuno-oncology (IO) therapies targeting the PD-1/PD-L1 axis, such as immune checkpoint inhibitor (ICI) antibodies, have emerged as promising treatments for early-stage breast cancer (ESBC). Despite immunotherapy's clinical significance, the number of benefiting patients remains small, and the therapy can prompt severe immune-related events. Current pathologic and transcriptomic predictions of IO response are limited in terms of accuracy and rely on single-site biopsies, which cannot fully account for tumor heterogeneity. In addition, transcriptomic analyses are costly and time-consuming. We therefore constructed a computational biomarker coupling biophysical simulations and artificial intelligence-based tissue segmentation of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRIs), enabling IO response prediction across the entire tumor. Methods By analyzing both single-cell and whole-tissue RNA-seq data from non-IO-treated ESBC patients, we associated gene expression levels of the PD-1/PD-L1 axis with local tumor biology. PD-L1 expression was then linked to biophysical features derived from DCE-MRIs to generate spatially- and temporally-resolved atlases (virtual tumors) of tumor biology, as well as the TumorIO biomarker of IO response. We quantified TumorIO within patient virtual tumors (n = 63) using integrative modeling to train and develop a corresponding TumorIO Score. Results We validated the TumorIO biomarker and TumorIO Score in a small, independent cohort of IO-treated patients (n = 17) and correctly predicted pathologic complete response (pCR) in 15/17 individuals (88.2% accuracy), comprising 10/12 in triple negative breast cancer (TNBC) and 5/5 in HR+/HER2- tumors. We applied the TumorIO Score in a virtual clinical trial (n = 292) simulating ICI administration in an IO-naïve cohort that underwent standard chemotherapy. Using this approach, we predicted pCR rates of 67.1% for TNBC and 17.9% for HR+/HER2- tumors with addition of IO therapy; comparing favorably to empiric pCR rates derived from published trials utilizing ICI in both cancer subtypes. Conclusion The TumorIO biomarker and TumorIO Score represent a next generation approach using integrative biophysical analysis to assess cancer responsiveness to immunotherapy. This computational biomarker performs as well as PD-L1 transcript levels in identifying a patient's likelihood of pCR following anti-PD-1 IO therapy. The TumorIO biomarker allows for rapid IO profiling of tumors and may confer high clinical decision impact to further enable personalized oncologic care.
Collapse
|
3
|
Makrantonakis AE, Zografos E, Gazouli M, Dimitrakakis K, Toutouzas KG, Zografos CG, Kalapanida D, Tsiakou A, Samelis G, Zagouri F. PD-L1 Gene Polymorphisms rs822336 G>C and rs822337 T>A: Promising Prognostic Markers in Triple Negative Breast Cancer Patients. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101399. [PMID: 36295559 PMCID: PMC9612177 DOI: 10.3390/medicina58101399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 12/02/2022]
Abstract
Background and Objectives: Triple-negative breast cancer (TNBC) is a highly heterogeneous subtype that is associated with unresponsiveness to therapy and hence with high mortality rates. In this study we aimed to investigate the prognostic role of the rs822336 G>C and rs822337 T>A polymorphisms of the PD-L1 (Programmed Death-Ligand 1) in TNBC patients. Materials and methods: Formalin-fixed paraffin-embedded tissues from 114 TNBC patients and blood samples from 124 healthy donors were genotyped, and subsequently extensive statistical analysis was performed in order to investigate the clinical value of these polymorphism in TNBC. Results: Regarding rs822336 G>C, we found that the CG genotype was the most common among women that harbored Stage IV breast tumors (81.8%; p = 0.022), recurred (38.9%; p = 0.02) and died (66.7%; p = 0.04). Similarly, the rs822337 T>A genotype AA is associated with worse prognosis, since it was the most common genotype among stage IV tumors (72.7%; p = 0.04) and in TNBC patients that relapsed (75%; p = 0.021) and died (81.5%; p = 0.004). Our statistical analysis revealed that the rs822336 G>C genotype CG and the rs822337 T>A allele AA are strongly associated with inferior DFS and OS intervals. Moreover, it was revealed that women harboring mutated genotypes of both SNPs had shorter disease-free (Kaplan−Meier; p = 0.037, Cox analysis; p = 0.04) and overall (Kaplan−Meier; p = 0.025, Cox analysis; p = 0.03) survival compared to patients having normal genotype of at least one SNP. Multivariate analysis also showed that the presence of mutated genotypes of both SNPs is a strong and independent marker for predicting shorter DFS (p = 0.02) and OS (p = 0.008). Conclusion: Our study revealed that PD-L1 rs822336 G>C and rs822337 T>A polymorphisms were differentially expressed in our cohort of TNBC patients, and that this distribution was associated with markers of unfavorable prognosis and worse survival.
Collapse
Affiliation(s)
| | - Eleni Zografos
- Department of Basic Medical Sciences, Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
- Correspondence: (E.Z.); (F.Z.)
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Konstantinos Dimitrakakis
- Department of Obstetrics and Gynaecology, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Konstantinos G. Toutouzas
- 1st Propaedeutic Surgical Department, Hippokrateio Hospital, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Constantinos G. Zografos
- 1st Propaedeutic Surgical Department, Hippokrateio Hospital, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Despoina Kalapanida
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Andriani Tsiakou
- First Department of Dermatology, Syggros Hospital, School of Medicine, National and Kapodistrian University of Athens, 161 21 Athens, Greece
| | - George Samelis
- Department of Oncology, Hippocrateion Hospital, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 115 28 Athens, Greece
- Correspondence: (E.Z.); (F.Z.)
| |
Collapse
|
4
|
Pereira IC, Mascarenhas IF, Capetini VC, Ferreira PMP, Rogero MM, Torres-Leal FL. Cellular reprogramming, chemoresistance, and dietary interventions in breast cancer. Crit Rev Oncol Hematol 2022; 179:103796. [PMID: 36049616 DOI: 10.1016/j.critrevonc.2022.103796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/16/2022] [Accepted: 08/21/2022] [Indexed: 10/31/2022] Open
Abstract
Breast cancer (BC) diagnosis has been associated with significant risk factors, including family history, late menopause, obesity, poor eating habits, and alcoholism. Despite the advances in the last decades regarding cancer treatment, some obstacles still hinder the effectiveness of therapy. For example, chemotherapy resistance is common in locally advanced or metastatic cancer, reducing treatment options and contributing to mortality. In this review, we provide an overview of BC metabolic changes, including the impact of restrictive diets associated with chemoresistance, the therapeutic potential of the diet on tumor progression, pathways related to metabolic health in oncology, and perspectives on the future in the area of oncological nutrition.
Collapse
Affiliation(s)
- Irislene Costa Pereira
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil; Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Isabele Frazão Mascarenhas
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | - Paulo Michel Pinheiro Ferreira
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, Sao Paulo, Brazil
| | - Francisco Leonardo Torres-Leal
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil; Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil.
| |
Collapse
|
5
|
Zhou Y, Che Y, Fu Z, Zhang H, Wu H. Triple-Negative Breast Cancer Analysis Based on Metabolic Gene Classification and Immunotherapy. Front Public Health 2022; 10:902378. [PMID: 35875026 PMCID: PMC9296841 DOI: 10.3389/fpubh.2022.902378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) has negative expression of ER, PR and HER-2. TNBC shows high histological grade and positive rate of lymph node metastasis, easy recurrence and distant metastasis. Molecular typing based on metabolic genes can reflect deeper characteristics of breast cancer and provide support for prognostic evaluation and individualized treatment. Metabolic subtypes of TNBC samples based on metabolic genes were determined by consensus clustering. CIBERSORT method was applied to evaluate the score distribution and differential expression of 22 immune cells in the TNBC samples. Linear discriminant analysis (LDA) established a subtype classification feature index. Kaplan-Meier (KM) and receiver operating characteristic (ROC) curves were generated to validate the performance of prognostic metabolic subtypes in different datasets. Finally, we used weighted correlation network analysis (WGCNA) to cluster the TCGA expression profile dataset and screen the co-expression modules of metabolic genes. Consensus clustering of the TCGA cohort/dataset obtained three metabolic subtypes (MC1, MC2, and MC3). The ROC analysis showed a high prognostic performance of the three clusters in different datasets. Specifically, MC1 had the optimal prognosis, MC3 had a poor prognosis, and the three metabolic subtypes had different prognosis. Consistently, the immune characteristic index established based on metabolic subtypes demonstrated that compared with the other two subtypes, MC1 had a higher IFNγ score, T cell lytic activity and lower angiogenesis score, T cell dysfunction and rejection score. TIDE analysis showed that MC1 patients were more likely to benefit from immunotherapy. MC1 patients were more sensitive to immune checkpoint inhibitors and traditional chemotherapy drugs Cisplatin, Paclitaxel, Embelin, and Sorafenib. Multiclass AUC based on RNASeq and GSE datasets were 0.85 and 0.85, respectively. Finally, based on co-expression network analysis, we screened 7 potential gene markers related to metabolic characteristic index, of which CLCA2, REEP6, SPDEF, and CRAT can be used to indicate breast cancer prognosis. Molecular classification related to TNBC metabolism was of great significance for comprehensive understanding of the molecular pathological characteristics of TNBC, contributing to the exploration of reliable markers for early diagnosis of TNBC and predicting metastasis and recurrence, improvement of the TNBC staging system, guiding individualized treatment.
Collapse
Affiliation(s)
- Yu Zhou
- Oncology Department, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yingqi Che
- Hematology-Oncology Department, Long Nan Hospital, Daqing, China
| | - Zhongze Fu
- Gastroenterology Department, The First Hospital of Qiqihar, Qiqihar, China
| | - Henan Zhang
- Oncology Department, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Huiyu Wu
- Third Department of Oncology, People's Hospital of Daqing, Daqing, China
- *Correspondence: Huiyu Wu
| |
Collapse
|
6
|
Li F, Zhao Y, Wei Y, Xi Y, Bu H. Tumor-Infiltrating Lymphocytes Improve Magee Equation-Based Prediction of Pathologic Complete Response in HR-Positive/HER2-Negative Breast Cancer. Am J Clin Pathol 2022; 158:291-299. [PMID: 35486808 DOI: 10.1093/ajcp/aqac041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 04/07/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Magee equation 3 (ME3) is predictive of the pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) in patients with hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer but with insufficient predictive performance. This study was designed to improve predictive ability by combining ME3 with additional clinicopathologic markers. METHODS We retrospectively enrolled 460 patients with HR-positive/HER2-negative breast cancer from 2 centers. We obtained baseline characteristics, the ME3 score, and the number of stromal tumor-infiltrating lymphocytes (sTILs). After performing a logistic regression analysis, a predictive nomogram was built and validated externally. RESULTS ME3 score (adjusted odds ratio [OR], 1.14 [95% confidence interval (CI), 1.10-1.17]; P < .001) and TILs (adjusted OR, 5.21 [95% CI, 3.33-8.14]; P < .001) were independently correlated with pCR. The nomogram (named ME3+) was established using ME3 and sTILs, and it demonstrated an area under the curve of 0.816 and 0.862 in internal and external validation, respectively, outperforming the ME3 score alone. sTILs and ME3 scores were also found to be positively correlated across the entire cohort (P < .001). CONCLUSIONS The combination of sTILs and ME3 score potentially shows better performance for predicting pCR than ME3 alone. Larger validations are required for widespread application of ME3+ nomogram in NAC settings for HR-positive/HER2-negative breast cancer.
Collapse
Affiliation(s)
- Fengling Li
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Zhao
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, China
| | - Yani Wei
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Uddin MN, Wang X. Identification of breast cancer subtypes based on gene expression profiles in breast cancer stroma. Clin Breast Cancer 2022; 22:521-537. [DOI: 10.1016/j.clbc.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022]
|
8
|
An Immune-Related Gene Prognostic Index for Triple-Negative Breast Cancer Integrates Multiple Aspects of Tumor-Immune Microenvironment. Cancers (Basel) 2021; 13:cancers13215342. [PMID: 34771505 PMCID: PMC8582543 DOI: 10.3390/cancers13215342] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Triple-negative breast cancer (TNBC) is the most refractory subtype of breast cancer. Immune checkpoint inhibitor (ICI) therapy has made progress in TNBC treatment. PD-L1 expression is a useful biomarker of ICI therapy efficacy. However, tumor-immune microenvironment (TIME) factors, such as immune cell compositions and tumor-infiltrating lymphocyte (TIL) status, also influence tumor immunity. Therefore, it is necessary to seek biomarkers that are associated with multiple aspects of TIME in TNBC. In this study, we developed an immune-related gene prognostic index (IRGPI) with a substantial prognostic value for TNBC. Moreover, the results from multiple cohorts reproducibly demonstrate that IRGPI is significantly associated with immune cell compositions, the exclusion and dysfunction of TILs, as well as PD-1 and PD-L1 expression in TIME. Therefore, IRGPI is a promising biomarker closely related to patient survival and TIME of TNBC and may have a potential effect on the immunotherapy strategy of TNBC. Abstract Tumor-immune cell compositions and immune checkpoints comprehensively affect TNBC outcomes. With the significantly improved survival rate of TNBC patients treated with ICI therapies, a biomarker integrating multiple aspects of TIME may have prognostic value for improving the efficacy of ICI therapy. Immune-related hub genes were identified with weighted gene co-expression network analysis and differential gene expression assay using The Cancer Genome Atlas TNBC data set (n = 115). IRGPI was constructed with Cox regression analysis. Immune cell compositions and TIL status were analyzed with CIBERSORT and TIDE. The discovery was validated with the Molecular Taxonomy of Breast Cancer International Consortium data set (n = 196) and a patient cohort from our hospital. Tumor expression or serum concentrations of CCL5, CCL25, or PD-L1 were determined with immunohistochemistry or ELISA. The constructed IRGPI was composed of CCL5 and CCL25 genes and was negatively associated with the patient’s survival. IRGPI also predicts the compositions of M0 and M2 macrophages, memory B cells, CD8+ T cells, activated memory CD4 T cells, and the exclusion and dysfunction of TILs, as well as PD-1 and PD-L1 expression of TNBC. IRGPI is a promising biomarker for predicting the prognosis and multiple immune characteristics of TNBC.
Collapse
|
9
|
Desterke C, Dang J, Lorenzo HK, Candelier JJ. Roles of tetraspanins during trophoblast development: bioinformatics and new perspectives. Cell Tissue Res 2021; 386:157-171. [PMID: 34278518 DOI: 10.1007/s00441-021-03502-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/28/2021] [Indexed: 11/24/2022]
Abstract
Tetraspanins are a superfamily of membrane proteins found in all eukaryotic organisms. They act as scaffold molecules that regulate the traffic and function of other membrane/signaling proteins, resulting in important downstream cellular consequences. The aim of this work was to use transcriptomes and bioinformatics analysis to identify the tetraspanins (and their partners) involved in trophoblast differentiation. We built a protein-protein interaction network around tetraspanins which revealed that tetraspanins CD9, CD81, and CD82 show a specific expression during trophoblast differentiation. These proteins appeared to be interconnected and to recruit several membrane partners which include integrins, immune-related molecules, and a variety of receptors. During weeks 8 to 24, a CD9 expression trajectory was identified in extravillous trophoblasts, and a website was developed: ( https://extravillous.shinyapps.io/CD9humanEVT/ ). In conclusion, CD81 may, together with CD9 and CD82, be interconnected in controlling trophoblast invasion in the endometrium. CD9 expression trajectory in extravillous trophoblast between weeks 8 and 24 shows the involvement of CD9 in cell adhesion and migration.
Collapse
Affiliation(s)
- Christophe Desterke
- Université Paris-Saclay, UFR Medicine, Gif-sur-Yvette, France.,INSERM UA9 Hôpital P. Brousse, 14 Avenue P.V. Couturier, 94800, Villejuif, France
| | - Julien Dang
- INSERM U970, 56 rue Leblanc, 75015, Paris, France.,Hôpital Bicêtre, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicetre, France
| | - Hans-Kristian Lorenzo
- Université Paris-Saclay, UFR Medicine, Gif-sur-Yvette, France.,Hôpital Bicêtre, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicetre, France.,INSERM U1197, Hôpital P. Brousse, 14 Avenue P.V. Couturier, 94800, Bâtiment Lavoisier, France
| | - Jean-Jacques Candelier
- Université Paris-Saclay, UFR Medicine, Gif-sur-Yvette, France. .,INSERM U1197, Hôpital P. Brousse, 14 Avenue P.V. Couturier, 94800, Bâtiment Lavoisier, France.
| |
Collapse
|
10
|
Lv Y, Lv D, Lv X, Xing P, Zhang J, Zhang Y. Immune Cell Infiltration-Based Characterization of Triple-Negative Breast Cancer Predicts Prognosis and Chemotherapy Response Markers. Front Genet 2021; 12:616469. [PMID: 33815462 PMCID: PMC8017297 DOI: 10.3389/fgene.2021.616469] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/23/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer represents the number one cause of cancer-associated mortality globally. The most aggressive molecular subtype is triple negative breast cancer (TNBC), of which limited therapeutic options are available. It is well known that breast cancer prognosis and tumor sensitivity toward immunotherapy are dictated by the tumor microenvironment. Breast cancer gene expression profiles were extracted from the METABRIC dataset and two TNBC clusters displaying unique immune features were identified. Activated immune cells formed a large proportion of cells in the high infiltration cluster, which correlated to a good prognosis. Differentially expressed genes (DEGs) extracted between two heterogeneous subtypes were used to further explore the underlying immune mechanism and to identify prognostic biomarkers. Functional enrichment analysis revealed that the DEGs were predominately related to some processes involved in activation and regulation of innate immune signaling. Using network analysis, we identified two modules in which genes were selected for further prognostic investigation. Validation by independent datasets revealed that CXCL9 and CXCL13 were good prognostic biomarkers for TNBC. We also performed comparisons between the above two genes and immune markers (CYT, APM, TILs, and TIS), as well as cell checkpoint marker expressions, and found a statistically significant correlation between them in both METABRIC and TCGA datasets. The potential of CXCL9 and CXCL13 to predict chemotherapy sensitivity was also evaluated. We found that the CXCL9 and CXCL13 were good predictors for chemotherapy and their expressions were higher in chemotherapy-responsive patients in contrast to those who were not responsive. In brief, immune infiltrate characterization on TNBC revealed heterogeneous subtypes with unique immune features allowed for the identification of informative and reliable characteristics representative of the local immune tumor microenvironment and were potential candidates to guide the management of TNBC patients.
Collapse
Affiliation(s)
- Yufei Lv
- Department of Anatomy, Harbin Medical University, Harbin, China
| | - Dongxu Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xiaohong Lv
- Department of Anatomy, Harbin Medical University, Harbin, China
| | - Ping Xing
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianguo Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yafang Zhang
- Department of Anatomy, Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Li C, Li X, Li G, Sun L, Zhang W, Jiang J, Ge Q. Identification of a prognosis‑associated signature associated with energy metabolism in triple‑negative breast cancer. Oncol Rep 2020; 44:819-837. [PMID: 32582991 PMCID: PMC7388543 DOI: 10.3892/or.2020.7657] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
At present, a large number of exciting results have been found regarding energy metabolism within the triple-negative breast cancer (TNBC) field. Apart from aerobic glycolysis, a number of other catabolic pathways have also been demonstrated to participate in energy generation. However, the prognostic value of energy metabolism for TNBC currently remains unclear. In the present study, the association between gene expression profiles of energy metabolism and outcomes in patients with TNBC was examined using datasets obtained from the Gene Expression Omnibus and The Cancer Genome Atlas. In total, four robust TNBC subtypes were identified on the basis of negative matrix factorization clustering and gene expression patterns, which exhibited distinct immunological, molecular and prognostic (disease-free survival) features. The differentially expressed genes were subsequently identified from the subgroup that demonstrated the poorest prognosis compared with the remaining 3 subgroups, where their biological functions were assessed further by means of gene ontology enrichment analysis. Any signatures found to be associated with energy metabolism were then established using the Cox proportional hazards model to assess patient prognosis. According to results of Kaplan-Meier analysis, the constructed signature consisting of eight genes that were associated with energy metabolism distinguished patient outcomes into low- and high-risk groups. In addition, this signature, which was found to be markedly associated with the clinical characteristics of the patients, served as an independent factor in predicting TNBC patient prognosis. According to gene set enrichment analysis, the gene sets related to the high-risk group participated in the MAPK signal transduction pathway, focal adhesion and extracellular matrix receptor interaction, whilst those related to the low-risk group were revealed to be mainly associated with mismatch repair and propanoate metabolism. Findings from the present study shed new light on the role of energy metabolism within TNBC, where the eight-gene signature associated with energy metabolism constructed can be utilized as a new prognostic marker for predicting survival in patients with TNBC.
Collapse
Affiliation(s)
- Chao Li
- Department of Breast Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Xujun Li
- Department of Breast Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Guangming Li
- Department of Breast Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Long Sun
- Department of Breast Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Wei Zhang
- Department of Breast Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Jing Jiang
- Department of Breast Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Qidong Ge
- Department of Breast Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
12
|
Latha NR, Rajan A, Nadhan R, Achyutuni S, Sengodan SK, Hemalatha SK, Varghese GR, Thankappan R, Krishnan N, Patra D, Warrier A, Srinivas P. Gene expression signatures: A tool for analysis of breast cancer prognosis and therapy. Crit Rev Oncol Hematol 2020; 151:102964. [PMID: 32464482 DOI: 10.1016/j.critrevonc.2020.102964] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/25/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Breast Cancer is the most predominant female cancer in developed as well as developing countries. The treatment strategies of breast cancers depends on an array of factors like age at diagnosis, menstrual status, dietary pattern, immunological response, genetic variations of the cancer cells etc. Recent technological advancements in cancer diagnosis lead to the emergence of gene expression pattern for better understanding of the tumor behavior. It has not only bolstered the prognosis, but also the early diagnosis and therapy. The accuracy in disease prognosis can be boosted when gene expression signatures are combined with traditional clinicopathological features. This review explains how the evolution of gene expression signatures for breast cancers, its advantages and future prospects. In addition, an overview of currently available gene expression signature analysis tools and consolidated information on their current status and specific benefits, that can be availed for breast cancer diagnosis are also discussed.
Collapse
Affiliation(s)
- Neetha Rajan Latha
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Arathi Rajan
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Revathy Nadhan
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Sarada Achyutuni
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Satheesh Kumar Sengodan
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India; Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Sreelatha Krishnakumar Hemalatha
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India; Department of Microbiology, Government Medical College, Thiruvananthapuram, Kerala, India
| | - Geetu Rose Varghese
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Ratheeshkumar Thankappan
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India; Research and Development Wing, Life Cell International Pvt Ltd, Chennai, Tamil Nadu, India
| | - Neethu Krishnan
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Dipyaman Patra
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Arathy Warrier
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Priya Srinivas
- Cancer Research Program 6, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
13
|
Coussy F, Lavigne M, de Koning L, Botty RE, Nemati F, Naguez A, Bataillon G, Ouine B, Dahmani A, Montaudon E, Painsec P, Chateau-Joubert S, Laetitia F, Larcher T, Vacher S, Chemlali W, Briaux A, Melaabi S, Salomon AV, Guinebretiere JM, Bieche I, Marangoni E. Response to mTOR and PI3K inhibitors in enzalutamide-resistant luminal androgen receptor triple-negative breast cancer patient-derived xenografts. Theranostics 2020; 10:1531-1543. [PMID: 32042320 PMCID: PMC6993232 DOI: 10.7150/thno.36182] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023] Open
Abstract
Luminal androgen receptor (LAR) breast cancer accounts for 10% of all triple-negative breast cancers (TNBC). Anti-androgen therapy for this subtype is in development, but yields only partial clinical benefits. In this study, we aimed to characterize the genomic alterations of LAR TNBC, to analyze activation of the PI3K signaling pathway and to compare the response to PI3K pathway inhibitors with that to anti-androgen therapy in patient-derived xenografts (PDX) of LAR TNBC. Methods: Four LAR PDX models were identified, on the basis of their transcriptomic profiles, in a cohort of 57 PDX models of TNBC. The expression of AR-related genes, basal and luminal cytokeratins and EMT genes was analyzed by RT-PCR and IHC. AKT1 and PIK3CA mutations were identified by targeted NGS, and activation of the PI3K pathway was analyzed with a reverse-phase protein array. Three LAR PDXs with a PIK3CA or AKT1 mutation were treated with the AR inhibitor enzalutamide, a PI3K inhibitor, a dual PI3K-mTOR inhibitor and a mTORC1-mTORC2 inhibitor. Finally, we screened a clinical cohort of 329 TNBC for PIK3CA and AKT1 hotspot mutations. Results: LAR TNBC PDXs were significantly enriched in PIK3CA and AKT1 mutations, and had higher levels of luminal-androgen-like gene expression and a higher PI3K pathway protein activation score than other TNBC subtypes. Immunohistochemistry analysis revealed strong expression of the luminal cytokeratin CK18 and AR in three LAR PDX models. We found that mTOR and PI3K inhibitors had marked antitumor activity in vivo in PDX harboring genomic alterations of PIK3CA and AKT1 genes that did not respond to the AR antagonist enzalutamide. PIK3CA mutations were detected in more than one third of AR+ TNBC from patients (38%), and only 10% of AR-negative TNBC. Conclusion: Our results for PDX models of LAR TNBC resistant to enzalutamide indicate that PIK3CA and AKT1 are potential therapeutic targets.
Collapse
|
14
|
Thakur V, Kutty RV. Recent advances in nanotheranostics for triple negative breast cancer treatment. J Exp Clin Cancer Res 2019; 38:430. [PMID: 31661003 PMCID: PMC6819447 DOI: 10.1186/s13046-019-1443-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most complex and aggressive type of breast cancer encountered world widely in women. Absence of hormonal receptors on breast cancer cells necessitates the chemotherapy as the only treatment regime. High propensity to metastasize and relapse in addition to poor prognosis and survival motivated the oncologist, nano-medical scientist to develop novel and efficient nanotherapies to solve such a big TNBC challenge. Recently, the focus for enhanced availability, targeted cellular uptake with minimal toxicity is achieved by nano-carriers. These smart nano-carriers carrying all the necessary arsenals (drugs, tracking probe, and ligand) designed in such a way that specifically targets the TNBC cells at site. Articulating the targeted delivery system with multifunctional molecules for high specificity, tracking, diagnosis, and treatment emerged as theranostic approach. In this review, in addition to classical treatment modalities, recent advances in nanotheranostics for early and effective diagnostic and treatment is discussed. This review highlighted the recently FDA approved immunotherapy and all the ongoing clinical trials for TNBC, in addition to nanoparticle assisted immunotherapy. Futuristic but realistic advancements in artificial intelligence (AI) and machine learning not only improve early diagnosis but also assist clinicians for their workup in TNBC. The novel concept of Nanoparticles induced endothelial leakiness (NanoEL) as a way of tumor invasion is also discussed in addition to classical EPR effect. This review intends to provide basic insight and understanding of the novel nano-therapeutic modalities in TNBC diagnosis and treatment and to sensitize the readers for continue designing the novel nanomedicine. This is the first time that designing nanoparticles with stoichiometric definable number of antibodies per nanoparticle now represents the next level of precision by design in nanomedicine.
Collapse
Affiliation(s)
- Vikram Thakur
- Department of Virology, Postgraduate Institute of Medical Education and Research, PGIMER, Chandigarh, 160012 India
| | - Rajaletchumy Veloo Kutty
- Faculty of Chemical and Process Engineering Technology, College of Engineering Technology,University Malaysia Pahang, Tun Razak Highway, 26300 Kuantan, Pahang Malaysia
- Center of Excellence for Advanced Research in Fluid Flow, University Malaysia Pahang, 26300, Kuantan, Pahang Malaysia
| |
Collapse
|
15
|
Decoding Immune Heterogeneity of Triple Negative Breast Cancer and Its Association with Systemic Inflammation. Cancers (Basel) 2019; 11:cancers11070911. [PMID: 31261762 PMCID: PMC6678607 DOI: 10.3390/cancers11070911] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive subtype with limited therapeutic options. New opportunities are emerging from current comprehensive characterization of tumor immune infiltration and fitness. Therefore, effectiveness of current chemotherapies and novel immunotherapies are partially dictated by host inflammatory and immune profiles. However, further progress in breast cancer immuno-oncology is required to reach a detailed awareness of the immune infiltrate landscape and to determine additional reliable and easily detectable biomarkers. In this study, by analyzing gene expression profiles of 54 TNBC cases we identified three TNBC clusters displaying unique immune features. Deep molecular characterization of immune cells cytolytic-activity and tumor-inflammation status reveled variability in the local composition of the immune infiltrate in the TNBC clusters, reconciled by tumor-infiltrating lymphocytes counts. Platelet-to-lymphocyte ratio (PLR), a blood systemic parameter of inflammation evaluated using pre-surgical blood test data, resulted negatively correlated with local tumoral cytolytic activity and T cell–inflamed microenvironment, whereas tumor aggressiveness score signature positively correlated with PLR values. These data highlighted that systemic inflammation parameters may represent reliable and informative markers of the local immune tumor microenvironment in TNBC patients and could be exploited to decipher tumor infiltrate properties and consequently to select the most appropriate therapies.
Collapse
|
16
|
Jézéquel P, Kerdraon O, Hondermarck H, Guérin-Charbonnel C, Lasla H, Gouraud W, Canon JL, Gombos A, Dalenc F, Delaloge S, Lemonnier J, Loussouarn D, Verrièle V, Campone M. Identification of three subtypes of triple-negative breast cancer with potential therapeutic implications. Breast Cancer Res 2019; 21:65. [PMID: 31101122 PMCID: PMC6525459 DOI: 10.1186/s13058-019-1148-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 05/03/2019] [Indexed: 02/06/2023] Open
Abstract
Background Heterogeneity and lack of targeted therapies represent the two main impediments to precision treatment of triple-negative breast cancer (TNBC), and therefore, molecular subtyping and identification of therapeutic pathways are required to optimize medical care. The aim of the present study was to define robust TNBC subtypes with clinical relevance. Methods Gene expression profiling by means of DNA chips was conducted in an internal TNBC cohort composed of 238 patients. In addition, external data (n = 257), obtained by using the same DNA chip, were used for validation. Fuzzy clustering was followed by functional annotation of the clusters. Immunohistochemistry was used to confirm transcriptomics results: CD138 and CD20 were used to test for plasma cell and B lymphocyte infiltrations, respectively; MECA79 and CD31 for tertiary lymphoid structures; and UCHL1/PGP9.5 and S100 for neurogenesis. Results We identified three molecular clusters within TNBC: one molecular apocrine (C1) and two basal-like-enriched (C2 and C3). C2 presented pro-tumorigenic immune response (immune suppressive), high neurogenesis (nerve infiltration), and high biological aggressiveness. In contrast, C3 exhibited adaptive immune response associated with complete B cell differentiation that occurs in tertiary lymphoid structures, and immune checkpoint upregulation. External cohort subtyping by means of the same approach proved the robustness of these results. Furthermore, plasma cell and B lymphocyte infiltrates, tertiary lymphoid structures, and neurogenesis were validated at the protein levels by means of histological evaluation and immunohistochemistry. Conclusion Our work showed that TNBC can be subcategorized in three different subtypes characterized by marked biological features, some of which could be targeted by specific therapies. Electronic supplementary material The online version of this article (10.1186/s13058-019-1148-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pascal Jézéquel
- Département de Biopathologie, Unité Mixte de Génomique du Cancer, Institut de Cancérologie de l'Ouest - site René Gauducheau, Bd Jacques Monod, 44805, Saint Herblain Cedex, France. .,Unité de Bioinfomique, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain Cedex, France. .,CRCINA, UMR 1232 INSERM, Université de Nantes, Université d'Angers, Institut de Recherche en Santé-Université de Nantes, 8 Quai Moncousu, BP 70721, 44007, Nantes Cedex 1, France.
| | - Olivier Kerdraon
- Laboratoire d'Anatomie et Cytologie Pathologiques, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain Cedex, France
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Catherine Guérin-Charbonnel
- Département de Biopathologie, Unité Mixte de Génomique du Cancer, Institut de Cancérologie de l'Ouest - site René Gauducheau, Bd Jacques Monod, 44805, Saint Herblain Cedex, France.,Unité de Bioinfomique, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain Cedex, France.,CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Institut de Recherche en Santé-Université de Nantes, 8 Quai Moncousu, BP 70721, 44007, Nantes Cedex 1, France
| | - Hamza Lasla
- Unité de Bioinfomique, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain Cedex, France
| | - Wilfried Gouraud
- Département de Biopathologie, Unité Mixte de Génomique du Cancer, Institut de Cancérologie de l'Ouest - site René Gauducheau, Bd Jacques Monod, 44805, Saint Herblain Cedex, France.,Unité de Bioinfomique, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain Cedex, France.,CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Institut de Recherche en Santé-Université de Nantes, 8 Quai Moncousu, BP 70721, 44007, Nantes Cedex 1, France
| | - Jean-Luc Canon
- Oncologie-Hématologie, Grand Hôpital de Charleroi, 3 Grand'Rue, 6000, Charleroi, Belgium
| | - Andrea Gombos
- Oncologie Médicale, Institut Jules Bordet, 121 Bd de Waterloo, 1000, Bruxelles, Belgium
| | - Florence Dalenc
- Oncologie Médicale, IUCT-Oncopole, 1 Av Irène Joliot-Curie, 31100, Toulouse, France
| | - Suzette Delaloge
- Oncologie Médicale, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | - Jérôme Lemonnier
- UCBG, R&D UNICANCER, Fédération Nationale des Centres de Lutte Contre le Cancer, 101 rue de Tolbiac, 75013, Paris Cedex 13, France
| | - Delphine Loussouarn
- Départment d'Anatomie et Cytologie Pathologiques, Centre Hospitalo-Universitaire, 1 place Alexis Ricordeau, 44093, Nantes, France
| | - Véronique Verrièle
- Laboratoire d'Anatomie et Cytologie Pathologiques, Institut de Cancérologie de l'Ouest, Bd Jacques Monod, 44805, Saint Herblain Cedex, France
| | - Mario Campone
- CRCINA, UMR 1232 INSERM, Université de Nantes, Université d'Angers, Institut de Recherche en Santé-Université de Nantes, 8 Quai Moncousu, BP 70721, 44007, Nantes Cedex 1, France.,Oncologie Médicale, Institut de Cancérologie de l'Ouest, René Gauducheau, Bd Jacques Monod, 44805, Saint Herblain Cedex, France
| |
Collapse
|
17
|
Jézéquel P, Guette C, Lasla H, Gouraud W, Boissard A, Guérin‐Charbonnel C, Campone M. iTRAQ‐Based Quantitative Proteomic Analysis Strengthens Transcriptomic Subtyping of Triple‐Negative Breast Cancer Tumors. Proteomics 2019; 19:e1800484. [DOI: 10.1002/pmic.201800484] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/28/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Pascal Jézéquel
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- Unité Mixte de Génomique du Cancer Institut de Cancérologie de l'Ouest ‐ René Gauducheau Bd Jacques Monod 44805 Saint Herblain Cedex France
- INSERM U1232 44007 Nantes Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Catherine Guette
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
- Institut de Cancérologie de l'Ouest—Paul Papin 49055 Angers Cedex France
| | - Hamza Lasla
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Wilfried Gouraud
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- Unité Mixte de Génomique du Cancer Institut de Cancérologie de l'Ouest ‐ René Gauducheau Bd Jacques Monod 44805 Saint Herblain Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Alice Boissard
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
- Institut de Cancérologie de l'Ouest—Paul Papin 49055 Angers Cedex France
| | - Catherine Guérin‐Charbonnel
- Unité de Bioinfomique Institut de Cancérologie de l'Ouest Bd Jacques Monod 44805 Saint Herblain Cedex France
- Unité Mixte de Génomique du Cancer Institut de Cancérologie de l'Ouest ‐ René Gauducheau Bd Jacques Monod 44805 Saint Herblain Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
| | - Mario Campone
- INSERM U1232 44007 Nantes Cedex France
- SIRIC ILIAD Institut de Cancérologie de l'Ouest‐René Gauducheau 44805 Saint Herblain Cedex France
- Oncologie Médicale Institut de Cancérologie de l'Ouest—René Gauducheau 44805 Saint Herblain Cedex France
| |
Collapse
|
18
|
Coussy F, de Koning L, Lavigne M, Bernard V, Ouine B, Boulai A, El Botty R, Dahmani A, Montaudon E, Assayag F, Morisset L, Huguet L, Sourd L, Painsec P, Callens C, Chateau-Joubert S, Servely JL, Larcher T, Reyes C, Girard E, Pierron G, Laurent C, Vacher S, Baulande S, Melaabi S, Vincent-Salomon A, Gentien D, Dieras V, Bieche I, Marangoni E. A large collection of integrated genomically characterized patient-derived xenografts highlighting the heterogeneity of triple-negative breast cancer. Int J Cancer 2019; 145:1902-1912. [PMID: 30859564 DOI: 10.1002/ijc.32266] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/26/2018] [Accepted: 02/19/2019] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC) represents 10% of all breast cancers and is a very heterogeneous disease. Globally, women with TNBC have a poor prognosis, and the development of effective targeted therapies remains a real challenge. Patient-derived xenografts (PDX) are clinically relevant models that have emerged as important tools for the analysis of drug activity and predictive biomarker discovery. The purpose of this work was to analyze the molecular heterogeneity of a large panel of TNBC PDX (n = 61) in order to test targeted therapies and identify biomarkers of response. At the gene expression level, TNBC PDX represent all of the various TNBC subtypes identified by the Lehmann classification except for immunomodulatory subtype, which is underrepresented in PDX. NGS and copy number data showed a similar diversity of significantly mutated gene and somatic copy number alteration in PDX and the Cancer Genome Atlas TNBC patients. The genes most commonly altered were TP53 and oncogenes and tumor suppressors of the PI3K/AKT/mTOR and MAPK pathways. PDX showed similar morphology and immunohistochemistry markers to those of the original tumors. Efficacy experiments with PI3K and MAPK inhibitor monotherapy or combination therapy showed an antitumor activity in PDX carrying genomic mutations of PIK3CA and NRAS genes. TNBC PDX reproduce the molecular heterogeneity of TNBC patients. This large collection of PDX is a clinically relevant platform for drug testing, biomarker discovery and translational research.
Collapse
Affiliation(s)
- Florence Coussy
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France.,Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France.,Department of Medical Oncology, Institut Curie, Paris, France
| | - Leanne de Koning
- Translational Research Department, RPPA Platform, Institut Curie Research Center, Paris, France
| | - Marion Lavigne
- Department of Biopathology, Institut Curie, Paris, France
| | - Virginie Bernard
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Berengere Ouine
- Translational Research Department, RPPA Platform, Institut Curie Research Center, Paris, France
| | - Anais Boulai
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Rania El Botty
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Ahmed Dahmani
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Franck Assayag
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Ludivine Morisset
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Lea Huguet
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Laura Sourd
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Pierre Painsec
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Celine Callens
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | | | - Jean-Luc Servely
- BioPôle Alfort, National Veterinary School of Alfort, Maison Alfort, France
| | | | - Cecile Reyes
- Translational Research Department, Genomics Platform, Institut Curie Research Center, Paris, France
| | | | - Gaelle Pierron
- Unit of Somatic Genomics, Department of Genetics, Institut Curie, Paris, France
| | | | - Sophie Vacher
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Sylvain Baulande
- Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, Paris, France
| | - Samia Melaabi
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | | | - David Gentien
- Translational Research Department, Genomics Platform, Institut Curie Research Center, Paris, France
| | | | - Ivan Bieche
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France.,Inserm U1016, Paris Descartes University, Paris, France
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| |
Collapse
|
19
|
He Y, Jiang Z, Chen C, Wang X. Classification of triple-negative breast cancers based on Immunogenomic profiling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:327. [PMID: 30594216 PMCID: PMC6310928 DOI: 10.1186/s13046-018-1002-1] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022]
Abstract
Background Abundant evidence shows that triple-negative breast cancer (TNBC) is heterogeneous, and many efforts have been devoted to identifying TNBC subtypes on the basis of genomic profiling. However, few studies have explored the classification of TNBC specifically based on immune signatures that may facilitate the optimal stratification of TNBC patients responsive to immunotherapy. Methods Using four publicly available TNBC genomics datasets, we classified TNBC on the basis of the immunogenomic profiling of 29 immune signatures. Unsupervised and supervised machine learning methods were used to perform the classification. Results We identified three TNBC subtypes that we named Immunity High (Immunity_H), Immunity Medium (Immunity_M), and Immunity Low (Immunity_L) and demonstrated that this classification was reliable and predictable by analyzing multiple different datasets. Immunity_H was characterized by greater immune cell infiltration and anti-tumor immune activities, as well as better survival prognosis compared to the other subtypes. Besides the immune signatures, some cancer-associated pathways were hyperactivated in Immunity_H, including apoptosis, calcium signaling, MAPK signaling, PI3K–Akt signaling, and RAS signaling. In contrast, Immunity_L presented depressed immune signatures and increased activation of cell cycle, Hippo signaling, DNA replication, mismatch repair, cell adhesion molecule binding, spliceosome, adherens junction function, pyrimidine metabolism, glycosylphosphatidylinositol (GPI)-anchor biosynthesis, and RNA polymerase pathways. Furthermore, we identified a gene co-expression subnetwork centered around five transcription factor (TF) genes (CORO1A, STAT4, BCL11B, ZNF831, and EOMES) specifically significant in the Immunity_H subtype and a subnetwork centered around two TF genes (IRF8 and SPI1) characteristic of the Immunity_L subtype. Conclusions The identification of TNBC subtypes based on immune signatures has potential clinical implications for TNBC treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-1002-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Zehang Jiang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Cai Chen
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China. .,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, Nanjing, 211198, China. .,Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
20
|
Alcalá-Corona SA, Espinal-Enríquez J, de Anda-Jáuregui G, Hernández-Lemus E. The Hierarchical Modular Structure of HER2+ Breast Cancer Network. Front Physiol 2018; 9:1423. [PMID: 30364267 PMCID: PMC6193406 DOI: 10.3389/fphys.2018.01423] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 09/19/2018] [Indexed: 11/13/2022] Open
Abstract
HER2-enriched breast cancer is a complex disease characterized by the overexpression of the ERBB2 amplicon. While the effects of this genomic aberration on the pathology have been studied, genome-wide deregulation patterns in this subtype of cancer are also observed. A novel approach to the study of this malignant neoplasy is the use of transcriptional networks. These networks generally exhibit modular structures, which in turn may be associated to biological processes. This modular regulation of biological functions may also exhibit a hierarchical structure, with deeper levels of modular organization accounting for more specific functional regulation. In this work, we identified the most probable (maximum likelihood) model of the hierarchical modular structure of the HER2-enriched transcriptional network as reconstructed from gene expression data, and analyzed the statistical associations of modules and submodules to biological functions. We found modular structures, independent from direct ERBB2 amplicon regulation, involved in different biological functions such as signaling, immunity, and cellular morphology. Higher resolution submodules were identified in more specific functions, such as micro-RNA regulation and the activation of viral-like immune response. We propose the approach presented here as one that may help to unveil mechanisms involved in the development of the pathology.
Collapse
Affiliation(s)
- Sergio Antonio Alcalá-Corona
- Computational Genomics, National Institute of Genomic Medicine, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | - Jesús Espinal-Enríquez
- Computational Genomics, National Institute of Genomic Medicine, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | | | - Enrique Hernández-Lemus
- Computational Genomics, National Institute of Genomic Medicine, Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| |
Collapse
|
21
|
Liu Z, Li M, Jiang Z, Wang X. A Comprehensive Immunologic Portrait of Triple-Negative Breast Cancer. Transl Oncol 2018; 11:311-329. [PMID: 29413765 PMCID: PMC5884188 DOI: 10.1016/j.tranon.2018.01.011] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/16/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a high-risk malignancy due to its high capacity for invasion and lack of targeted therapy. Immunotherapy continues to demonstrate efficacy in a variety of cancers, and thus may be a promising strategy for TNBC given the limited therapeutic options currently available for TNBC. In this study, we performed an exhaustive analysis of immunogenic signatures in TNBC based on 2 large-scale breast cancer (BC) genomic data. We compared enrichment levels of 26 immune cell activities and pathways among TNBC, non-TNBC, and normal tissue, and within TNBCs of different genotypic or phenotypic features. We found that almost all analyzed immune activities and pathways had significantly higher enrichment levels in TNBC than non-TNBC. Elevated enrichment of these immune activities and pathways was likely to be associated with better survival prognosis in TNBC. This study demonstrated that TNBC likely exhibits the strongest immunogenicity among BC subtypes, and thus warrants the immunotherapeutic option for TNBC.
Collapse
Affiliation(s)
- Zhixian Liu
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Mengyuan Li
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Zehang Jiang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaosheng Wang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
22
|
Foukakis T, Lövrot J, Matikas A, Zerdes I, Lorent J, Tobin N, Suzuki C, Brage SE, Carlsson L, Einbeigi Z, Linderholm B, Loman N, Malmberg M, Fernö M, Skoog L, Bergh J, Hatschek T. Immune gene expression and response to chemotherapy in advanced breast cancer. Br J Cancer 2018; 118:480-488. [PMID: 29370583 PMCID: PMC5830596 DOI: 10.1038/bjc.2017.446] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 12/30/2022] Open
Abstract
Background: Transcriptomic profiles have shown promise as predictors of response to neoadjuvant chemotherapy in breast cancer (BC). This study aimed to explore their predictive value in the advanced BC (ABC) setting. Methods: In a Phase 3 trial of first-line chemotherapy in ABC, a fine needle aspiration biopsy (FNAB) was obtained at baseline. Intrinsic molecular subtypes and gene modules related to immune response, proliferation, oestrogen receptor (ER) signalling and recurring genetic alterations were analysed for association with objective response to chemotherapy. Gene-set enrichment analysis (GSEA) of responders vs non-responders was performed independently. Lymphocytes were enumerated in FNAB smears and the absolute abundance of immune cell types was calculated using the Microenvironment Cell Populations counter method. Results: Gene expression data were available for 109 patients. Objective response to chemotherapy was statistically significantly associated with an immune module score (odds ratio (OR)=1.62; 95% confidence interval (CI), 1.03–2.64; P=0.04). Subgroup analysis showed that this association was restricted to patients with ER-positive or luminal tumours (OR=3.54; 95%, 1.43–10.86; P=0.012 and P for interaction=0.04). Gene-set enrichment analysis confirmed that in these subgroups, immune-related gene sets were enriched in responders. Conclusions: Immune-related transcriptional signatures may predict response to chemotherapy in ER-positive and luminal ABC.
Collapse
Affiliation(s)
- Theodoros Foukakis
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - John Lövrot
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Alexios Matikas
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Ioannis Zerdes
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Julie Lorent
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Nick Tobin
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Chikako Suzuki
- Department of Radiology and Nuclear Medicine, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Suzanne Egyházi Brage
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Lena Carlsson
- Department of Oncology, Sundsvall General Hospital, Sundsvall 85643, Sweden
| | - Zakaria Einbeigi
- Department of Oncology, Sahlgrenska University Hospital, Gothenborg 41345, Sweden
| | - Barbro Linderholm
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden.,Department of Oncology, Sahlgrenska University Hospital, Gothenborg 41345, Sweden
| | - Niklas Loman
- Department of Oncology, Skåne University Hospital, Lund 22241, Sweden
| | - Martin Malmberg
- Department of Oncology, Helsingborg General Hospital, Helsingborg 25187, Sweden
| | - Mårten Fernö
- Department of Oncology, Skåne University Hospital, Lund 22241, Sweden
| | - Lambert Skoog
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| | - Thomas Hatschek
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet and University Hospital, Stockholm 17176, Sweden
| |
Collapse
|
23
|
Sadacca B, Hamy AS, Laurent C, Gestraud P, Bonsang-Kitzis H, Pinheiro A, Abecassis J, Neuvial P, Reyal F. New insight for pharmacogenomics studies from the transcriptional analysis of two large-scale cancer cell line panels. Sci Rep 2017; 7:15126. [PMID: 29123141 PMCID: PMC5680301 DOI: 10.1038/s41598-017-14770-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/12/2017] [Indexed: 12/31/2022] Open
Abstract
One of the most challenging problems in the development of new anticancer drugs is the very high attrition rate. The so-called “drug repositioning process” propose to find new therapeutic indications to already approved drugs. For this, new analytic methods are required to optimize the information present in large-scale pharmacogenomics datasets. We analyzed data from the Genomics of Drug Sensitivity in Cancer and Cancer Cell Line Encyclopedia studies. We focused on common cell lines (n = 471), considering the molecular information, and the drug sensitivity for common drugs screened (n = 15). We propose a novel classification based on transcriptomic profiles of cell lines, according to a biological network-driven gene selection process. Our robust molecular classification displays greater homogeneity of drug sensitivity than cancer cell line grouped based on tissue of origin. We then identified significant associations between cell line cluster and drug response robustly found between both datasets. We further demonstrate the relevance of our method using two additional external datasets and distinct sensitivity metrics. Some associations were still found robust, despite cell lines and drug responses’ variations. This study defines a robust molecular classification of cancer cell lines that could be used to find new therapeutic indications to known compounds.
Collapse
Affiliation(s)
- Benjamin Sadacca
- Residual Tumor & Response to Treatment Laboratory (RT2Lab), PSL Research University, Translational Research Department, F-75248, Paris, France.,U932 Immunity and Cancer; INSERM; Institut Curie, Paris, France.,Laboratoire de Mathématiques et Modélisation d'Evry, Université d'Évry Val d'Essonne, UMR CNRS 8071, ENSIIE, USC INRA, Evry Val d'Essonne, France
| | - Anne-Sophie Hamy
- Residual Tumor & Response to Treatment Laboratory (RT2Lab), PSL Research University, Translational Research Department, F-75248, Paris, France.,U932 Immunity and Cancer; INSERM; Institut Curie, Paris, France
| | - Cécile Laurent
- Residual Tumor & Response to Treatment Laboratory (RT2Lab), PSL Research University, Translational Research Department, F-75248, Paris, France.,U932 Immunity and Cancer; INSERM; Institut Curie, Paris, France
| | - Pierre Gestraud
- Institut Curie, PSL Research University, Mines Paris Tech, Bioinformatics and Computational Systems Biology of Cancer, INSERM U900, F-75005, Paris, France
| | - Hélène Bonsang-Kitzis
- Residual Tumor & Response to Treatment Laboratory (RT2Lab), PSL Research University, Translational Research Department, F-75248, Paris, France.,U932 Immunity and Cancer; INSERM; Institut Curie, Paris, France.,Department of Surgery, Institut Curie, Paris, F-75248, France
| | - Alice Pinheiro
- Residual Tumor & Response to Treatment Laboratory (RT2Lab), PSL Research University, Translational Research Department, F-75248, Paris, France.,U932 Immunity and Cancer; INSERM; Institut Curie, Paris, France
| | - Judith Abecassis
- Residual Tumor & Response to Treatment Laboratory (RT2Lab), PSL Research University, Translational Research Department, F-75248, Paris, France.,U932 Immunity and Cancer; INSERM; Institut Curie, Paris, France.,Mines Paristech, PSL-Research University, CBIO-Centre for Computational Biology, Mines ParisTech, Fontainebleau, F-77300, France.,Institut Curie, PSL Research University, Mines Paris Tech, Bioinformatics and Computational Systems Biology of Cancer, INSERM U900, F-75005, Paris, France
| | - Pierre Neuvial
- Laboratoire de Mathématiques et Modélisation d'Evry, Université d'Évry Val d'Essonne, UMR CNRS 8071, ENSIIE, USC INRA, Evry Val d'Essonne, France.,Institut de Mathématiques de Toulouse; UMR5219 Université de Toulouse; CNRS UPS IMT, F-31062, Toulouse Cedex 9, France
| | - Fabien Reyal
- Residual Tumor & Response to Treatment Laboratory (RT2Lab), PSL Research University, Translational Research Department, F-75248, Paris, France. .,U932 Immunity and Cancer; INSERM; Institut Curie, Paris, France. .,Department of Surgery, Institut Curie, Paris, F-75248, France.
| |
Collapse
|
24
|
Abstract
Immunotherapy is currently the most rapidly advancing area of clinical oncology, and provides the unprecedented opportunity to effectively treat, and even cure, several previously untreatable malignancies. A growing awareness exists of the fact that the success of chemotherapy and radiotherapy, in which the patient's disease can be stabilized well beyond discontinuation of treatment (and occasionally is cured), also relies on the induction of a durable anticancer immune response. Indeed, the local immune infiltrate undergoes dynamic changes that accompany a shift from a pre-existing immune response to a therapy-induced immune response. As a result, the immune contexture, which is determined by the density, composition, functional state and organization of the leukocyte infiltrate of the tumour, can yield information that is relevant to prognosis, prediction of a treatment response and various other pharmacodynamic parameters. Several complementary technologies can be used to explore the immune contexture of tumours, and to derive biomarkers that could enable the adaptation of individual treatment approaches for each patient, as well as monitoring a response to anticancer therapies.
Collapse
|
25
|
Wang ZQ, Milne K, Derocher H, Webb JR, Nelson BH, Watson PH. PD-L1 and intratumoral immune response in breast cancer. Oncotarget 2017; 8:51641-51651. [PMID: 28881675 PMCID: PMC5584276 DOI: 10.18632/oncotarget.18305] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 04/28/2017] [Indexed: 12/20/2022] Open
Abstract
Purpose PD-L1 is thought to play an important role in the antitumor immune response. In this study, we investigated the expression of PD-L1 within breast tumor subsets to better define its prognostic significance. Methods Immunohistochemistry was performed to determine PD-L1 tumor cell expression and to enumerate CD8, CD4 and CD68 tumor-infiltrating leucocytes (TIL) in a cohort of 443 breast cancers categorized by molecular subtype. Results Across the entire cohort, PD-L1 tumor cell expression was observed in 73/443 (16.5%) cases and associated with known indicators of poor prognosis, including low patient age, high tumor grade, ER/PR negative status, but not with outcome. However, in the Triple Negative breast cancer subset PD-L1 was associated with better recurrence free survival (RFS) especially within the Basal-like subset (Hazard ratio = 0.39, 95% CI = 0.22 - 0.86, p = 0.018). Combined PD-L1/epithelial CD8 positive status was also strongly associated with better RFS and OS (Hazard ratio = 0.12, 95% CI = 0.10 - 0.71, p = 0.010 and Hazard ratio = 0.11, 95% CI = 0.11 - 0.68, p = 0.006 respectively) in the Basal-like subgroup. Conclusions PD-L1 expression is associated with better patient survival in Basal-like breast cancer.
Collapse
Affiliation(s)
- Zhi-Qiang Wang
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katy Milne
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - Heather Derocher
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - John R Webb
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Brad H Nelson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada.,Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter H Watson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Hamy AS, Bonsang-Kitzis H, Lae M, Moarii M, Sadacca B, Pinheiro A, Galliot M, Abecassis J, Laurent C, Reyal F. A Stromal Immune Module Correlated with the Response to Neoadjuvant Chemotherapy, Prognosis and Lymphocyte Infiltration in HER2-Positive Breast Carcinoma Is Inversely Correlated with Hormonal Pathways. PLoS One 2016; 11:e0167397. [PMID: 28005906 PMCID: PMC5178998 DOI: 10.1371/journal.pone.0167397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION HER2-positive breast cancer (BC) is a heterogeneous group of aggressive breast cancers, the prognosis of which has greatly improved since the introduction of treatments targeting HER2. However, these tumors may display intrinsic or acquired resistance to treatment, and classifiers of HER2-positive tumors are required to improve the prediction of prognosis and to develop novel therapeutic interventions. METHODS We analyzed 2893 primary human breast cancer samples from 21 publicly available datasets and developed a six-metagene signature on a training set of 448 HER2-positive BC. We then used external public datasets to assess the ability of these metagenes to predict the response to chemotherapy (Ignatiadis dataset), and prognosis (METABRIC dataset). RESULTS We identified a six-metagene signature (138 genes) containing metagenes enriched in different gene ontologies. The gene clusters were named as follows: Immunity, Tumor suppressors/proliferation, Interferon, Signal transduction, Hormone/survival and Matrix clusters. In all datasets, the Immunity metagene was less strongly expressed in ER-positive than in ER-negative tumors, and was inversely correlated with the Hormonal/survival metagene. Within the signature, multivariate analyses showed that strong expression of the "Immunity" metagene was associated with higher pCR rates after NAC (OR = 3.71[1.28-11.91], p = 0.019) than weak expression, and with a better prognosis in HER2-positive/ER-negative breast cancers (HR = 0.58 [0.36-0.94], p = 0.026). Immunity metagene expression was associated with the presence of tumor-infiltrating lymphocytes (TILs). CONCLUSION The identification of a predictive and prognostic immune module in HER2-positive BC confirms the need for clinical testing for immune checkpoint modulators and vaccines for this specific subtype. The inverse correlation between Immunity and hormone pathways opens research perspectives and deserves further investigation.
Collapse
Affiliation(s)
- Anne-Sophie Hamy
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
| | - Hélène Bonsang-Kitzis
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- Department of Surgery, Institut Curie, Paris, France
| | - Marick Lae
- Department of Tumor Biology, Institut Curie, Paris, France
| | - Matahi Moarii
- Mines Paristech, PSL-Research University, CBIO-Centre for Computational Biology, Mines ParisTech, Fontainebleau, France
- U900, INSERM, Institut Curie, Paris, France
| | - Benjamin Sadacca
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- Laboratoire de Mathématiques et Modélisation d’Evry, Université d’Évry Val d’Essonne, Evry, France
| | - Alice Pinheiro
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
| | - Marion Galliot
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
| | - Judith Abecassis
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- Mines Paristech, PSL-Research University, CBIO-Centre for Computational Biology, Mines ParisTech, Fontainebleau, France
- U900, INSERM, Institut Curie, Paris, France
| | - Cecile Laurent
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
| | - Fabien Reyal
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- Department of Surgery, Institut Curie, Paris, France
| |
Collapse
|