1
|
Bouzid RS, Bouzid R, Labed H, Serhani I, Hellal D, Oumeddour L, Boudhiaf I, Ibrir M, Khadraoui H, Belaaloui G. Molecular subtyping and target identification in triple negative breast cancer through immunohistochemistry biomarkers. BMC Cancer 2025; 25:454. [PMID: 40082760 PMCID: PMC11905517 DOI: 10.1186/s12885-025-13832-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The Triple-Negative Breast Cancer (TNBC) molecular subtyping and target identification based on Immunohistochemistry (IHC) is of considerable worth for routine use. Yet, literature on this topic is limited worldwide and needs to be enriched with data from different populations. METHODS We assessed the IHC expression of subtyping biomarkers (Cytokeratins 5, 14 and 17, Epidermal Growth Factor Receptor, Claudins 3 and 7, E-cadherin, Vimentin and Androgen receptor) and predictive biomarkers (Tumor-infiltrating lymphocytes (TILs) density, Breast Cancer Antigen 1 (BRCA1) and P53) in a cohort of TNBC patients. Clinicopathologic parameters and overall survival (OS) were investigated as well. RESULTS The patients were aged 50.11 ± 12.13y (more than 40y in 76.56% of patients), and 23.44% had a BC family history. They were in a non-advanced stage: 51.6% T2 stage, 56.2% negative lymph node involvement, 76.6% without metastasis and 64.1% grade II Scarff-Bloom-Richardson classification (SBR). The IHC subtypes were: 53.1% Basal-like1 (BL1), 6.3% Basal-like2 (BL2), 17.2% Mesenchymal (MES), 9.4% Luminal Androgen Receptor (LAR), 4.7% Mixed subtype and 9.4% "Unclassified" type. The LAR subtype involved the youngest patients (40.17 ± 8.68y, p = 0.02). The "Unclassified" subtype expressed the p53 mutated-type pattern more frequently (100%, p = 0.07). The BRCA1 mutated pattern and TILs infiltration were present in (23.44% and 37.5% of patients, respectively). The OS of the subtypes differed significantly (p = 0.007, log-rank test). The subtypes median OS were, respectively, 15.47 mo. (Unclassified), 18.94 mo. (BL2), 27.23 mo. (MES), 27.28 mo. (Mixed), 30.88 mo. (BL1), and 45.07 mo. (LAR). There was no difference in the OS following age, BRCA1 expression, p53 pattern and TILs density. Though, the OS following the TNM stage was different (p = 0.001). A multivariable Cox proportional hazards regression analysis showed that TNM staging and TNBC subtypes, independently influence the OS (p < 0.001 and p = 0.017, respectively). Hence, IHC is useful in TNBC subtyping for prognostic purposes and in the identification of therapeutic biomarkers. Further investigation is required to confirm our results and to implement IHC as a routine tool to improve patient's care.
Collapse
Affiliation(s)
- Rima Saad Bouzid
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Radhia Bouzid
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Housna Labed
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Iman Serhani
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Dounia Hellal
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Leilia Oumeddour
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Ines Boudhiaf
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Massouda Ibrir
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, University Hospital, Batna, Algeria
| | - Hachani Khadraoui
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Ghania Belaaloui
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria.
- Faculty of Medicine, University Batna 2, Batna, Algeria.
| |
Collapse
|
2
|
Kaewkedsri P, Intarawichian P, Jessadapattarakul S, Kunprom W, Koonmee S, Thanee M, Somintara O, Wongbuddha A, Chadbunchachai P, Nawapun S, Aphivatanasiri C. Programmed Cell Death Ligand 1 (PD-L1) and Major Histocompatibility Complex Class I (MHC Class I) Expression Patterns and Their Pathologic Associations in Triple-Negative Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:123-143. [PMID: 39936074 PMCID: PMC11812676 DOI: 10.2147/bctt.s506833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Purpose This study aims to investigate the clinicopathological characteristics of triple-negative breast cancer (TNBC) in relation to programmed cell death ligand 1 (PD-L1) and major histocompatibility complex class I (MHC class I) expression, with a focus on their prognostic significance. Patients and Methods A retrospective analysis was conducted on formalin-fixed paraffin-embedded (FFPE) tissue samples from 148 TNBC patients diagnosed between 2008 and 2021. Immunohistochemical analysis evaluated PD-L1 and MHC class I expression. PD-L1 was assessed using Combine Positive Scores (CPS), with the threshold set at CPS ≥ 1 and CPS ≥ 10. MHC class I expression was categorized into low and high levels. Associations between these markers, clinicopathological features, overall survival (OS), and disease-free survival (DFS) were analyzed. PD-L1 expression was also compared between older FFPE blocks (2008-2018) versus newer blocks (2019-2021). Results PD-L1 expression was observed in 29.1% of cases with a Combined Positive Score (CPS) ≥1 and 8.8% of CPS ≥10 cases. MHC class I expression was evenly split between low and high levels. Older FFPE blocks (2008-2018) showed lower PD-L1 expression than newer blocks (2019-2021). There was no significant association between PD-L1 expression and overall survival (OS) or disease-free survival (DFS). However, high MHC class I expression was strongly associated with improved OS (HR = 0.469, 95% CI: 0.282-0.780, p=0.004). Patients with negative PD-L1 and high MHC class I expression had the most favorable prognosis, with significant OS for CPS ≥1 (HR = 0.447, 95% CI: 0.236-0.846, p=0.013) and CPS ≥10 (HR = 0.516, 95% CI: 0.307-0.869, p=0.013). Conclusion These findings support the potential of PD-L1 and MHC class I expression as prognostic markers for TNBC, offering insights to guide treatment decisions and improve patient outcomes.
Collapse
Affiliation(s)
- Ponkrit Kaewkedsri
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | | | - Waritta Kunprom
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Supinda Koonmee
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Malinee Thanee
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Ongart Somintara
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Anongporn Wongbuddha
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Payia Chadbunchachai
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Supajit Nawapun
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | |
Collapse
|
3
|
Zhang C, Li N, Zhang P, Jiang Z, Cheng Y, Li H, Pang Z. Advancing precision and personalized breast cancer treatment through multi-omics technologies. Am J Cancer Res 2024; 14:5614-5627. [PMID: 39803662 PMCID: PMC11711544 DOI: 10.62347/mwnz5609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
Breast cancer is the most common malignant tumour in women, with more than 685,000 women dying of breast cancer each year. The heterogeneity of breast cancer complicates both treatment and diagnosis. Traditional methods based on histopathology and hormone receptor status are now no longer sufficient. Recently, advances in multi-omics techniques, including genomic, proteomic, and transcriptomic analyses, have deepened our understanding of breast cancer. Combining these approaches allows for precise molecular subtyping, which is essential for the detection of key mutations, protein interactions and gene expression patterns that are highly relevant to different therapeutic strategies. Genomic analyses have been effectively identifying key mutations in cancer. Meanwhile, proteomics and transcriptomics complement by identifying new therapeutic targets and elucidating gene expression dynamics. Integrating multi-omics and conventional diagnostics improves tumour characterisation and enables prognostic accuracy comparable to established standards and treatment response. Existing and emerging technologies enable real-time enhanced tumour follow-up and data analysis through liquid biopsy and artificial intelligence, respectively. Despite these clinical implementation challenges, multi-omics including clinical phenotyping offers significant potential for precision breast cancer treatment. This article describes recent advances in molecular subtyping and multi-omics technologies that are driving key innovations to optimise patient outcomes and further develop personalised medicine in the context of breast cancer care.
Collapse
Affiliation(s)
- Chenlu Zhang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Nan Li
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
- Heilongjiang Provincial Center for Prevention and Treatment of Cerebral Palsy in Children Postdoctoral Research WorkstationNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Pengxia Zhang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Zhimei Jiang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Yichao Cheng
- School of Rehabilitation Medicine, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Huiqing Li
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| | - Zhenfei Pang
- School of Basic Medical Sciences, Jiamusi UniversityNo. 258, Xuefu Street, Xiangyang District, Jiamusi 154007, Heilongjiang, China
| |
Collapse
|
4
|
Khan S, Bhake A, Sagar S. Deciphering the Role of BRAFV600E Immunohistochemistry in Breast Lesions: A Comprehensive Review. Cureus 2024; 16:e64872. [PMID: 39156294 PMCID: PMC11330685 DOI: 10.7759/cureus.64872] [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: 06/21/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
The BRAFV600E mutation has been extensively studied in various cancers, but its role in breast lesions remains less understood. Immunohistochemistry (IHC) has emerged as a valuable tool for detecting BRAFV600E expression in breast tissue, aiding in diagnosis and prognosis. This comprehensive review examines the significance of BRAFV600E IHC in breast lesions, encompassing its frequency, association with clinicopathological features, and potential clinical implications. We summarize key findings, emphasizing their utility in diagnosis, prognosis prediction, and treatment response assessment. Additionally, we discuss implications for clinical practice, highlighting the need for integrating BRAFV600E IHC into diagnostic algorithms. Recommendations for future research include larger-scale studies to validate findings, optimize detection techniques, and explore therapeutic interventions targeting BRAFV600E in breast cancer. This review contributes to understanding the molecular landscape of breast lesions and informs clinical decision-making in their management.
Collapse
Affiliation(s)
- Simran Khan
- Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Arvind Bhake
- Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Shakti Sagar
- Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
5
|
de Paula B, Crocamo S, de Sousa CAM, Valverde P, Rezende F, Abdelhay E. Triple-Negative Breast Cancer Subclassified by Immunohistochemistry: Correlation with Clinical and Pathological Outcomes in Patients Receiving Neoadjuvant Chemotherapy. Int J Mol Sci 2024; 25:5825. [PMID: 38892013 PMCID: PMC11172922 DOI: 10.3390/ijms25115825] [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: 04/22/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
The intrinsic subtype of triple-negative breast cancer (TNBC) is based on genomic evaluation. In this study, we report the survival and pathological complete response (pCR) rates of TNBC patients subtyped by IHC and treated with neoadjuvant chemotherapy (NACT). A retrospective cohort of 187 TNBC patients who received NACT between 2008 and 2017 was used, and IHC subtyping was performed on biopsy specimens before chemotherapy. The subtyping revealed predominantly basal-like tumors (IHC-BL, 61%), followed by basal-like immune-suppressed tumors (IHC-BLIS, 31%), mesenchymal tumors (12.5%), luminal androgen receptor tumors (IHC-LAR, 12%), and basal-like immune-activated tumors (IHC-BLIA, 10.9%). The pCR rate varied among subtypes, with IHC-BLIA showing the highest (30.0%) and IHC-LAR showing the lowest (4.5%). IHC-BLIS led in recurrence sites. Overall and disease-free survival analyses did not show significant differences among subtypes, although IHC-BLIA demonstrated a trend toward better survival, and IHC-mesenchymal, worse. Patients who achieved pCR exhibited significantly better disease-free survival and overall survival than non-responders. This study underscores the potential of IHC-based subtyping in TNBC management, highlighting distinct response patterns to neoadjuvant chemotherapy and potential implications for treatment strategies. Further research is warranted to validate these findings and explore tailored therapeutic approaches for specific TNBC subtypes.
Collapse
Affiliation(s)
- Bruno de Paula
- Núcleo de Pesquisa Clínica, Hospital do Cancer III, Instituto Nacional de Câncer –, Rio de Janeiro 20560-121, Brazil;
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford GU2 7XH, UK
| | - Susanne Crocamo
- Núcleo de Pesquisa Clínica, Hospital do Cancer III, Instituto Nacional de Câncer –, Rio de Janeiro 20560-121, Brazil;
| | | | - Priscila Valverde
- Divisão de Patologia, COAS, Instituto Nacional de Câncer–INCA, Rio de Janeiro 20220-400, Brazil
| | - Fabiana Rezende
- Divisão de Patologia, COAS, Instituto Nacional de Câncer–INCA, Rio de Janeiro 20220-400, Brazil
| | - Eliana Abdelhay
- Divisão de Laboratórios Especializados, COAS, Instituto Nacional de Câncer–INCA, Rio de Janeiro 202300-130, Brazil
| |
Collapse
|
6
|
Weng L, Zhou J, Guo S, Xu N, Ma R. The molecular subtyping and precision medicine in triple-negative breast cancer---based on Fudan TNBC classification. Cancer Cell Int 2024; 24:120. [PMID: 38555429 PMCID: PMC10981301 DOI: 10.1186/s12935-024-03261-0] [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: 09/29/2023] [Accepted: 02/02/2024] [Indexed: 04/02/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is widely recognized as the most aggressive form of breast cancer, occurring more frequently in younger patients and characterized by high heterogeneity, early distant metastases and poor prognosis. Multiple treatment options have failed to achieve the expected therapeutic effects due to the lack of clear molecular targets. Based on genomics, transcriptomics and metabolomics, the multi-omics analysis further clarifies TNBC subtyping, which provides a greater understanding of tumour heterogeneity and targeted therapy sensitivity. For instance, the luminal androgen receptor subtype (LAR) exhibits responsiveness to anti-AR therapy, and the basal-like immune-suppressed subtype (BLIS) tends to benefit from poly (ADP-ribose) polymerase inhibitors (PARPis) and anti-angiogenic therapy. The efficacy of multi-dimensional combination therapy holds immense importance in guiding personalized and precision medicine for TNBC. This review offers a systematic overview of recent FuDan TNBC molecular subtyping and its role in the instruction of clinical precision therapy.
Collapse
Affiliation(s)
- Lijuan Weng
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jianliang Zhou
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shenchao Guo
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Ruishuang Ma
- Department of Radiotherapy and Chemotherapy, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
7
|
Jing R, Bai S, Zhang P, Ren H, Jia L, Li W, Zheng G. IDO-1 impairs antitumor immunity of natural killer cells in triple-negative breast cancer via up-regulation of HLA-G. Breast Cancer 2024; 31:135-147. [PMID: 37981615 PMCID: PMC10764509 DOI: 10.1007/s12282-023-01522-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/28/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Triple-negative breast cancers (TNBC) are highly aggressive malignancies with poor prognosis. As an essential enzyme in the tryptophan-kynurenine metabolic pathway, indoleamine 2,3 dioxygenase-1 (IDO-1) has been reported to facilitate immune escape of various tumors. However, the mechanism underlying the immunosuppressive role of IDO-1 in TNBC remains largely uncharacterized. METHODS We examined the IDO-1 expression in 93 clinical TNBC tissues and paired adjacent normal tissues, and analyzed the regulation role of environmental cytokines like IFN-γ in IDO-1 expression. The effect of IDO-1 expression in TNBC cells on the function of NK cells were then evaluated and the underlying mechanisms were exploited. RESULTS IDO-1 expressed in 50 of 93 (54.1%) TNBC patients. TNBC patients with high IDO-1 expression tended to have more infiltrated immune cells including NK cells, which are less active than patients with low IDO-1 expression. NK cells could produce IFN-γ, which induced IDO-1 expression in TNBC cells, whereas IDO-1 impaired the cytotoxicity of co-cultured NK cells by upregulation of HLA-G. Blockade of HLA-G improved the antitumor activity of NK cells to TNBC in vivo. CONCLUSION TNBC cells induce dysfunction of NK cells through an IFN-γ/IDO-1/HLA-G pathway, which provide novel insights into the mechanisms of TNBC progression and demonstrate the applicability of IDO-1 and HLA-G targeting in the treatment of TNBC.
Collapse
Affiliation(s)
- Rui Jing
- College of Life Sciences, Northwest University, Xi'an, 710069, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Shukun Bai
- College of Life Sciences, Northwest University, Xi'an, 710069, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Peipei Zhang
- College of Life Sciences, Northwest University, Xi'an, 710069, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Ren
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Lintao Jia
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Weimiao Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xi'an, 710004, China.
| | - Guoxu Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Immunology, Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
8
|
Carvalho FM. Triple-negative breast cancer: from none to multiple therapeutic targets in two decades. Front Oncol 2023; 13:1244781. [PMID: 38023167 PMCID: PMC10666917 DOI: 10.3389/fonc.2023.1244781] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) are more likely to occur in younger patients and have a poor prognosis. They are highly heterogeneous tumors consisting of different molecular subtypes. The only common characteristic among them is the absence of targets for endocrine therapy and human epidermal growth factor receptor 2 (HER2) blockade. In the past two decades, there has been an increased understanding of these tumors from a molecular perspective, leading to their stratification according to new therapeutic strategies. TNBC has ushered breast carcinomas into the era of immunotherapy. The higher frequency of germline BRCA mutations in these tumors enables targeting this repair defect by drugs like PARP inhibitors, resulting in synthetic lethality in neoplastic cells. Additionally, we have the identification of new molecules to which this generation of smart drugs, such as antibody-drug conjugates (ADCs), are directed. In this review, we will discuss the trajectory of this knowledge in a systematic manner, presenting the molecular bases, therapeutic possibilities, and biomarkers.
Collapse
Affiliation(s)
- Filomena Marino Carvalho
- Department of Pathology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|