1
|
Ran R, Zhao S, Zhou Y, Hang X, Wang H, Fan Y, Zhang Y, Qiao Y, Yang J, Dong D. Clinicopathological characteristics, treatment patterns and outcomes in patients with HER2-positive breast cancer based on hormone receptor status: a retrospective study. BMC Cancer 2024; 24:1216. [PMID: 39350043 PMCID: PMC11443648 DOI: 10.1186/s12885-024-12974-4] [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: 12/31/2023] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Different hormone receptor (HR) expression patterns have significant biological and therapeutic implications in patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, the distinction between HR-positive /HER2-positive (HR+/HER2+) and HR-negative/HER2-positive (HR-/HER2+) subtypes remains unclear. METHODS This retrospective study analyzed 828 patients with HER2-positive breast cancer at the First Affiliated Hospital of Xi'an Jiaotong University from 2012 to 2022. Baseline characteristics were compared by chi-square test. Survival outcomes were estimated by Kaplan-Meier method. RESULTS In total, 56.3% (n = 466) had HR-positive and 43.7% (n = 362) had HR-negative disease. Comparatively, HR+/HER2 + breast cancers presented favorable clinicopathological features. At a median follow-up of 49 months, 199 disease-free survival (DFS) events and 99 deaths were observed. HR+/HER2 + patients had significantly better survival outcomes than HR-/HER2 + patients. HR-positive status was an independent protective factor for overall survival (OS) [P = 0.032; hazard ratio, 0.61; 95% confidence interval (CI), 0.39-0.96] and DFS (P = 0.001; hazard ratio, 0.61; 95% CI, 0.46-0.81). HR+/HER2 + patients were significantly less sensitive to neoadjuvant therapy than HR-/HER2 + patients. In the first-line treatment for HR+/HER2 + advanced breast cancer, receiving endocrine therapy significantly improved advanced-OS (P < 0.001; hazard ratio, 0.33; 95% CI, 0.18-0.59) and progression-free survival (PFS) (P < 0.001; hazard ratio, 0.38; 95% CI, 0.25-0.58) compared with not receiving endocrine therapy. Moreover, maintenance endocrine therapy after HER2-targeted therapy and chemotherapy is associated with significant advanced-OS and PFS benefits compared with no maintenance endocrine therapy (advanced-OS: P < 0.001; hazard ratio, 0.05; 95% CI, 0.03-0.12; PFS: P < 0.001; hazard ratio, 0.35; 95% CI, 0.21-0.57). CONCLUSIONS This study reveals the high heterogeneity of HER2-positive breast cancer related to HR status in clinicopathological features, metastasis patterns, and outcomes. Large randomized controlled trials are warranted to optimize treatment strategies for the HER2-positive breast cancer population.
Collapse
Affiliation(s)
- Ran Ran
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shidi Zhao
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhou
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyue Hang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui Wang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuan Fan
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yusi Zhang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yifan Qiao
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Yang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Danfeng Dong
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Ballé JK, Vetter M, Kenea TW, Eber-Schulz P, Reibold C, Ziegenhorn HV, Stückrath K, Wickenhauser C, Addissie A, Santos P, Kantelhardt EJ, Getachew S, Bauer M. PAM50 breast cancer subtypes and survival of patients in rural Ethiopia without adjuvant treatment: a prospective observational study. BMC Cancer 2024; 24:1127. [PMID: 39256703 PMCID: PMC11385137 DOI: 10.1186/s12885-024-12867-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
PURPOSE Survival rates of breast cancer (BC) patients are particularly low in rural regions in sub-Saharan Africa (SSA) which is due to limited access to therapy. In recent years, gene expression profiling (GEP) of BC showed a strong prognostic value in patients with local tumour surgery and (neo)adjuvant treatment. The aim of this study was to evaluate the impact of intrinsic subtypes on survival of patients in rural Ethiopia without any (neo)adjuvant therapy. METHODS In total, 113 female patients from Aira Hospital with histologically proven BC and treated only with surgery were included in this study. All samples were analysed by immunohistochemistry (IHC) for estrogen receptor, progesterone receptor, HER2 and Ki67, as well as RNA-expression analysis for PAM50 subtyping. RESULTS A positive hormone receptor status was found in 69.0% of the tumours and intrinsic subtyping demonstrated Luminal B to be the most common subtype (34.5%). Follow-up data was available for 79 of 113 patients. Two-year overall survival (OS) was 57.3% and a considerably worse OS was observed in patients with Basal-like BC compared to Luminal A BC. Moreover, advanced tumours showed an increased risk of mortality. CONCLUSION The OS was very low in the patient cohort that received no (neo)adjuvant treatment. Immunohistochemistry and GEP confirmed endocrine-sensitive tumours in more than half of the patients, with a large proportion of Luminal B, HER2-enriched and Basal-like tumours so that adjuvant chemotherapy should be recommended.
Collapse
Affiliation(s)
- Judith Katharina Ballé
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Martina Vetter
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | | | - Pia Eber-Schulz
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christian Reibold
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Hannes-Viktor Ziegenhorn
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Anesthesiology and Critical Care Medicine, Carl Gustav Carus University Hospital, Dresden, Germany
| | - Kathrin Stückrath
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Adamu Addissie
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Epidemiology and Biostatistics, School of Public Health, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Pablo Santos
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Eva Johanna Kantelhardt
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sefonias Getachew
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Epidemiology and Biostatistics, School of Public Health, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Marcus Bauer
- Global Health Working Group, Institute of Medical Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| |
Collapse
|
3
|
Regner MJ, Garcia-Recio S, Thennavan A, Wisniewska K, Mendez-Giraldez R, Felsheim B, Spanheimer PM, Parker JS, Perou CM, Franco HL. Defining the Regulatory Logic of Breast Cancer Using Single-Cell Epigenetic and Transcriptome Profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598858. [PMID: 38948758 PMCID: PMC11212881 DOI: 10.1101/2024.06.13.598858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Annotation of the cis-regulatory elements that drive transcriptional dysregulation in cancer cells is critical to improving our understanding of tumor biology. Herein, we present a compendium of matched chromatin accessibility (scATAC-seq) and transcriptome (scRNA-seq) profiles at single-cell resolution from human breast tumors and healthy mammary tissues processed immediately following surgical resection. We identify the most likely cell-of-origin for luminal breast tumors and basal breast tumors and then introduce a novel methodology that implements linear mixed-effects models to systematically quantify associations between regions of chromatin accessibility (i.e. regulatory elements) and gene expression in malignant cells versus normal mammary epithelial cells. These data unveil regulatory elements with that switch from silencers of gene expression in normal cells to enhancers of gene expression in cancer cells, leading to the upregulation of clinically relevant oncogenes. To translate the utility of this dataset into tractable models, we generated matched scATAC-seq and scRNA-seq profiles for breast cancer cell lines, revealing, for each subtype, a conserved oncogenic gene expression program between in vitro and in vivo cells. Together, this work highlights the importance of non-coding regulatory mechanisms that underlie oncogenic processes and the ability of single-cell multi-omics to define the regulatory logic of BC cells at single-cell resolution.
Collapse
Affiliation(s)
- Matthew J. Regner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Aatish Thennavan
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Raul Mendez-Giraldez
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brooke Felsheim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Philip M. Spanheimer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hector L. Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Division of Clinical and Translational Cancer Research, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR 00935
| |
Collapse
|
4
|
Joo EH, Kim S, Park D, Lee T, Park WY, Han KY, Lee JE. Migratory Tumor Cells Cooperate with Cancer Associated Fibroblasts in Hormone Receptor-Positive and HER2-Negative Breast Cancer. Int J Mol Sci 2024; 25:5876. [PMID: 38892065 PMCID: PMC11172245 DOI: 10.3390/ijms25115876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Hormone receptor-positive and HER2-negative breast cancer (HR+/HER2-BC) is the most common type with a favorable prognosis under endocrine therapy. However, it still demonstrates unpredictable progression and recurrences influenced by high tumoral diversity and microenvironmental status. To address these heterogeneous molecular characteristics of HR+/HER2-BC, we aimed to simultaneously characterize its transcriptomic landscape and genetic architecture at the same resolution. Using advanced single-cell RNA and DNA sequencing techniques together, we defined four distinct tumor subtypes. Notably, the migratory tumor subtype was closely linked to genomic alterations of EGFR, related to the tumor-promoting behavior of IL6-positive inflammatory tumor-associated fibroblast, and contributing to poor prognosis. Our study comprehensively utilizes integrated analysis to uncover the complex dynamics of this breast cancer subtype, highlighting the pivotal role of the migratory tumor subtype in influencing surrounding cells. This sheds light on potential therapeutic targets by offering enhanced insights for HR+/HER2-BC treatment.
Collapse
Affiliation(s)
- Eun Hye Joo
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; (E.H.J.); (W.-Y.P.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06355, Republic of Korea
| | - Sangmin Kim
- Department of Breast Cancer Center, Samsung Medical Center, Seoul 06351, Republic of Korea;
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Donghyun Park
- Planit Healthcare Inc., Seoul 06235, Republic of Korea;
| | - Taeseob Lee
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06355, Republic of Korea;
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; (E.H.J.); (W.-Y.P.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06355, Republic of Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; (E.H.J.); (W.-Y.P.)
| | - Jeong Eon Lee
- Department of Breast Cancer Center, Samsung Medical Center, Seoul 06351, Republic of Korea;
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
5
|
Li X, Luo S, Fu W, Huang M, Huang X, Kang S, Zhang J, Wang Q, Song C. Discovery of a proliferation essential gene signature and actin-like 6A as potential biomarkers for predicting prognosis and neoadjuvant chemotherapy response in triple-positive breast cancer. Cancer 2024; 130:1435-1448. [PMID: 38358781 DOI: 10.1002/cncr.35228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 12/26/2023] [Indexed: 02/16/2024]
Abstract
BACKGROUND Patients with triple-positive breast cancer (TPBC) have a higher risk of recurrence and lower survival rates than patients with other luminal breast cancers. However, there are few studies on the predictive biomarkers of prognosis and treatment responses in TPBC. METHODS Proliferation essential genes (PEGs) were acquired from clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9) technology, and cohorts of patients with TPBC were obtained from public databases and our cohort. To develop a TPBC-PEG signature, Cox regression and least absolute shrinkage and selection operator regression analyses were applied. Functional analyses were performed with gene set enrichment analysis. The relationship between candidate genes and neoadjuvant chemotherapy (NACT) sensitivity was explored via real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC) on the basis of clinical samples. RESULTS Among 900 TPBC-PEGs, 437 showed significant differential expression between TPBC and normal tissues. Three prognostic PEGs (actin-like 6A [ACTL6A], chaperonin containing TCP1 subunit 2 [CCT2], and threonyl-TRNA synthetase [TARS]) were identified and used to construct the PEG signature. Patients with high PEG signature scores exhibited a worse overall survival and lower sensitivity to NACT than patients with low PEG signature scores. RT-qPCR results indicated that ACTL6A and CCT2 expression were significantly upregulated in patients who lacked sensitivity to NACT. IHC results showed that the ACTL6A protein was highly expressed in patients with NACT resistance and nonpathological complete responses. CONCLUSIONS This efficient PEG signature prognostic model can predict the outcomes of TPBC. Furthermore, ACTL6A expression level was associated with the response to NACT, and could serve as an important factor in predicting prognosis and drug sensitivity of patients with TPBC.
Collapse
Affiliation(s)
- Xiaofen Li
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Shiping Luo
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Wenfen Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Mingyao Huang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Xiewei Huang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Shaohong Kang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Jie Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Qingshui Wang
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Chuangui Song
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| |
Collapse
|
6
|
Berg T, Jensen MB, Celik A, Talman ML, Misiakou MA, Knoop AS, Nielsen FC, Ejlertsen B, Rossing M. Molecular subtyping improves breast cancer diagnosis in the Copenhagen Breast Cancer Genomics Study. JCI Insight 2024; 9:e178114. [PMID: 38587073 PMCID: PMC11128195 DOI: 10.1172/jci.insight.178114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/16/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUNDIntrinsic molecular subtypes define distinct biological breast cancers and can be used to further improve diagnosis and risk allocation.METHODSThe Copenhagen Breast Cancer Genomics Study (CBCGS) prospectively included women diagnosed with breast cancer at Rigshospitalet from 2014 to 2021. Eligible patients were females with a primary invasive breast cancer (T1c, if N0M0; otherwise, any T, any N, or any M stage) and no prior malignancy. All patients underwent molecular profiling with the CIT256 and PAM50 molecular profile.RESULTSIn the study period, 2,816 patients were included in the CBCGS. Molecular subtyping showed an increase in nonluminal (molecular-apocrine, luminal C, and Basal-like) as compared with luminal (luminal A, luminal B, and Normal-like) subtypes with increasing stage from I to IV. Across all stages, we found a significant difference in survival among subtypes; 91% of patients with LumA were alive at 5 years compared with 91% for LumB, 84% for LumC, 82% for mApo, and 80% for Basal-like. We identified 442 tumors (16%) that were discordant in subtype between CIT256 and IHC. Discordant subtype proved to be a risk factor of death among patients with IHC luminal breast cancer (hazard ratio [HR], 2.08; 95% CI, 1.51-2.86) in a multivariable Cox regression analysis. Discordance occurred more often among patients with N3, stage IV, or grade III disease.CONCLUSIONOur findings indicate that molecular subtypes are a predominant classification for survival. Assessment is particularly crucial for patients with IHC luminal breast cancer with known high-risk factors, since they are at an increased risk of harboring an aggressive molecular subtype.
Collapse
Affiliation(s)
- Tobias Berg
- Danish Breast Cancer Group
- Department of Clinical Oncology
- Center for Genomic Medicine, and
| | | | | | - Maj-Lis Talman
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Finn Cilius Nielsen
- Center for Genomic Medicine, and
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Bent Ejlertsen
- Danish Breast Cancer Group
- Department of Clinical Oncology
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria Rossing
- Center for Genomic Medicine, and
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Di Palma S, Koliou P, Simonovic A, Costa D, Faulkes C, Kobutungi B, Paterson F, Horsnell JD, Pakzad F, Irvine T, Partlett P, Clayton E, Collins N. Breast Cancer Molecular Subtyping in Practice: A Real-World Study of the APIS Breast Cancer Subtyping Assay in a Consecutive Series of Breast Core Biopsies. Int J Mol Sci 2024; 25:2616. [PMID: 38473863 DOI: 10.3390/ijms25052616] [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: 01/08/2024] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 03/14/2024] Open
Abstract
The APIS Breast Cancer Subtyping Kit is an mRNA-based assessment of the seven parameters including three biomarkers routinely assessed in all the newly diagnosed breast cancers (BC), oestrogen receptor (ER), progesterone receptor (PR) and HER-2 and an additional four genes that create a novel proliferation signature, MKI67, PCNA, CCNA2 and KIF23. Taken together, the data are used to produce a molecular subtype for every sample. The kit was evaluated against the current standard protocol of immunohistochemistry (IHC) and/or in situ hybridisation (ISH) in breast cancer patients. The data were presented at the weekly breast multidisciplinary team (MDT) meeting. A total of 98 consecutive cases of pre-operative breast cancer core biopsies and two core biopsies of nodal metastases yielding 100 cases were assessed. IHC and APIS results were available for 100 and 99 cases. ER was concordant in 97% cases, PR was concordant in 89% and HER-2 results were concordant with IHC/ISH in 100% of the cases. Ki-67 IHC was discordant in 3% of cases when compared with MK167 alone but discordant in 24% when compared with the four-gene proliferation signature. In conclusion, our study indicates that the APIS Breast Cancer Subtyping Kit is highly concordant when compared to the results produced for ER/PR/HER-2 by IHC and/or ISH. The assay could play a role in the routine assessment of newly diagnosed breast cancer (BC) specimens.
Collapse
Affiliation(s)
- Silvana Di Palma
- Department of Cellular Pathology, Berkshire & Surrey Pathology Services, The Royal Surrey Hospital NHS Foundation Trust, University of Surrey, Egerton Road, Guildford GU2 7XX, UK
| | - Panagiotis Koliou
- Department of Oncology, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Alex Simonovic
- Department of Cellular Pathology, Berkshire & Surrey Pathology Services, The Royal Surrey Hospital NHS Foundation Trust, University of Surrey, Egerton Road, Guildford GU2 7XX, UK
| | - Daniela Costa
- Molecular Diagnostics, Berkshire & Surrey Pathology Services, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Catherine Faulkes
- Molecular Diagnostics, Berkshire & Surrey Pathology Services, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Brenda Kobutungi
- Molecular Diagnostics, Berkshire & Surrey Pathology Services, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Felicity Paterson
- Department of Oncology, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Jonathan David Horsnell
- Breast Unit, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Farrokh Pakzad
- Breast Unit, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Tracey Irvine
- Breast Unit, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Polly Partlett
- Breast Unit, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Elizabeth Clayton
- Breast Unit, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Nadine Collins
- Molecular Diagnostics, Berkshire & Surrey Pathology Services, The Royal Surrey Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| |
Collapse
|
8
|
Shakyawar SK, Sajja BR, Patel JC, Guda C. iCluF: an unsupervised iterative cluster-fusion method for patient stratification using multiomics data. BIOINFORMATICS ADVANCES 2024; 4:vbae015. [PMID: 38698887 PMCID: PMC11063539 DOI: 10.1093/bioadv/vbae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/10/2023] [Accepted: 01/26/2024] [Indexed: 05/05/2024]
Abstract
Motivation Patient stratification is crucial for the effective treatment or management of heterogeneous diseases, including cancers. Multiomic technologies facilitate molecular characterization of human diseases; however, the complexity of data warrants the need for the development of robust data integration tools for patient stratification using machine-learning approaches. Results iCluF iteratively integrates three types of multiomic data (mRNA, miRNA, and DNA methylation) using pairwise patient similarity matrices built from each omic data. The intermediate omic-specific neighborhood matrices implement iterative matrix fusion and message passing among the similarity matrices to derive a final integrated matrix representing all the omics profiles of a patient, which is used to further cluster patients into subtypes. iCluF outperforms other methods with significant differences in the survival profiles of 8581 patients belonging to 30 different cancers in TCGA. iCluF also predicted the four intrinsic subtypes of Breast Invasive Carcinomas with adjusted rand index and Fowlkes-Mallows scores of 0.72 and 0.83, respectively. The Gini importance score showed that methylation features were the primary decisive players, followed by mRNA and miRNA to identify disease subtypes. iCluF can be applied to stratify patients with any disease containing multiomic datasets. Availability and implementation Source code and datasets are available at https://github.com/GudaLab/iCluF_core.
Collapse
Affiliation(s)
- Sushil K Shakyawar
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Balasrinivasa R Sajja
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Jai Chand Patel
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Genetics, Cell Biology and Anatomy, Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE 68198-5805, United States
| |
Collapse
|
9
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
10
|
Xu Y, Liang Y, Yin G. Detecting the expression of HRs and BCL2 via IHC can help identify luminal A-like subtypes of triple-positive breast cancers. Clin Transl Oncol 2023; 25:1024-1032. [PMID: 36376700 DOI: 10.1007/s12094-022-03007-2] [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: 09/10/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Triple-positive breast cancer (TPBC) is a tumor that simultaneously expresses estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2). Luminal A-like TPBC is a special subtype with a favorable prognosis but benefits less from HER2-targeted therapy. However, little is known about how to identify luminal A-like TPBCs. Therefore, our study aims to explore a clinically feasible method to identify luminal A-like TPBCs using immunohistochemical (IHC) markers. METHODS Our cohort enrolled consecutive 190 patients with early-stage TPBCs diagnosed, treated and followed up in our hospital between 2013 and 2019. Patients whose IHC staining displayed ≥ 50% in both ER and PR scores and B-cell lymphoma 2 (BCL2) positivity were classified as cohort A (n = 64), and the rest were enrolled in cohort B (n = 126). Kaplan-Meier plotter and log-rank test were used to compare the survival difference between cohort A and cohort B and the efficacy of trastuzumab therapy in the two cohorts. RESULTS The disease-free survival (DFS) of patients in cohort A was significantly better than in cohort B (p = 0.031). In cohort A, there was no statistically significant difference in DFS between patients treated with trastuzumab and those without trastuzumab (p = 0.663). While in cohort B, patients treated with trastuzumab had significantly better DFS than those without trastuzumab (p = 0.032). Multivariate survival analysis showed that cohort A was associated with better DFS(95%CI 1.046-11.776, p = 0.042). CONCLUSION TPBCs consist of heterogeneous subtypes. Detecting the expression of ER, PR and BCL2 via IHC can help identify luminal A-like TPBCs. This study will enable individualized treatment of TPBCs.
Collapse
Affiliation(s)
- Yingying Xu
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, 130041, China
| | - Yonghao Liang
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, 130041, China
| | - Guanghao Yin
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
11
|
Dix-Peek T, Phakathi BP, van den Berg EJ, Dickens C, Augustine TN, Cubasch H, Neugut AI, Jacobson JS, Joffe M, Ruff P, Duarte RAB. Discordance between PAM50 intrinsic subtyping and immunohistochemistry in South African women with breast cancer. Breast Cancer Res Treat 2023; 199:1-12. [PMID: 36867282 PMCID: PMC10147771 DOI: 10.1007/s10549-023-06886-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/03/2023] [Indexed: 03/04/2023]
Abstract
PURPOSE Breast cancer is a heterogeneous disease with different gene expression profiles, treatment options and outcomes. In South Africa, tumors are classified using immunohistochemistry. In high-income countries multiparameter genomic assays are being utilized with implications for tumor classification and treatment. METHODS In a cohort of 378 breast cancer patients from the SABCHO study, we investigated the concordance between tumor samples classified by IHC and the PAM50 gene assay. RESULTS IHC classified patients as ER-positive (77.5%), PR-positive (70.6%), and HER2-positive (32.3%). These results, together with Ki67, were used as surrogates for intrinsic subtyping, and showed 6.9% IHC-A-clinical, 72.7% IHC-B-clinical, 5.3% IHC-HER2-clinical and 15.1% triple negative cancer (TNC). Typing using the PAM50 gave 19.3% luminal-A, 32.5% luminal-B, 23.5% HER2-enriched and 24.6% basal-like. The basal-like and TNC had the highest concordance, while the luminal-A and IHC-A group had the lowest concordance. By altering the cutoff for Ki67, and realigning the HER2/ER/PR-positive patients to IHC-HER2, we improved concordance with the intrinsic subtypes. CONCLUSION We suggest that the Ki67 be changed to a cutoff of 20-25% in our population to better reflect the luminal subtype classifications. This change would inform treatment options for breast cancer patients in settings where genomic assays are unaffordable.
Collapse
Affiliation(s)
- Thérèse Dix-Peek
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Johannesburg, 2193, South Africa.
| | - Boitumelo P Phakathi
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of Kwa-Zulu Natal, Durban, 4001, South Africa.,Department of Surgery, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Johannesburg, 2193, South Africa
| | - Eunice J van den Berg
- Department of Histopathology, National Health Laboratory Service, Chris Hani Baragwanath Hospital, 26 Chris Hani Road, Diepkloof, Johannesburg, 1864, South Africa.,Department of Anatomical Pathology, University of the Witwatersrand, 7 York Road, Johannesburg, 2193, South Africa
| | - Caroline Dickens
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Johannesburg, 2193, South Africa
| | - Tanya N Augustine
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193, South Africa
| | - Herbert Cubasch
- Batho Pele Breast Unit, Chris Hani Baragwanath Academic Hospital, 26 Chris Hani Road, Diepkloof, Soweto, 1860, South Africa.,SA MRC Common Epithelial Cancer Research Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Alfred I Neugut
- Herbert Irving Comprehensive Cancer Centre, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Judith S Jacobson
- Herbert Irving Comprehensive Cancer Centre, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Maureen Joffe
- SA MRC Common Epithelial Cancer Research Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa.,Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Paul Ruff
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Johannesburg, 2193, South Africa.,SA MRC Common Epithelial Cancer Research Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Raquel A B Duarte
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Johannesburg, 2193, South Africa
| |
Collapse
|
12
|
Systemically Identifying Triple-Negative Breast Cancer Subtype-Specific Prognosis Signatures, Based on Single-Cell RNA-Seq Data. Cells 2023; 12:cells12030367. [PMID: 36766710 PMCID: PMC9913740 DOI: 10.3390/cells12030367] [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: 11/23/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 01/21/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous disease with different molecular subtypes. Although progress has been made, the identification of TNBC subtype-associated biomarkers is still hindered by traditional RNA-seq or array technologies, since bulk data detected by them usually have some non-disease tissue samples, or they are confined to measure the averaged properties of whole tissues. To overcome these constraints and discover TNBC subtype-specific prognosis signatures (TSPSigs), we proposed a single-cell RNA-seq-based bioinformatics approach for identifying TSPSigs. Notably, the TSPSigs we developed mostly were found to be disease-related and involved in cancer development through investigating their enrichment analysis results. In addition, the prognostic power of TSPSigs was successfully confirmed in four independent validation datasets. The multivariate analysis results showed that TSPSigs in two TNBC subtypes-BL1 and LAR, were two independent prognostic factors. Further, analysis results of the TNBC cell lines revealed that the TSPSigs expressions and drug sensitivities had significant associations. Based on the preceding data, we concluded that TSPSigs could be exploited as novel candidate prognostic markers for TNBC patients and applied to individualized treatment in the future.
Collapse
|
13
|
Hamaneh M, Yu YK. A Simple Method for Robust and Accurate Intrinsic Subtyping of Breast Cancer. Cancer Inform 2023; 22:11769351231159893. [PMID: 37008073 PMCID: PMC10052604 DOI: 10.1177/11769351231159893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/07/2023] [Indexed: 04/04/2023] Open
Abstract
Motivation The PAM50 signature/method is widely used for intrinsic subtyping of breast cancer samples. However, depending on the number and composition of the samples included in a cohort, the method may assign different subtypes to the same sample. This lack of robustness is mainly due to the fact that PAM50 subtracts a reference profile, which is computed using all samples in the cohort, from each sample before classification. In this paper we propose modifications to PAM50 to develop a simple and robust single-sample classifier, called MPAM50, for intrinsic subtyping of breast cancer. Like PAM50, the modified method uses a nearest centroid approach for classification, but the centroids are computed differently, and the distances to the centroids are determined using an alternative method. Additionally, MPAM50 uses unnormalized expression values for classification and does not subtract a reference profile from the samples. In other words, MPAM50 classifies each sample independently, and so avoids the previously mentioned robustness issue. Results A training set was employed to find the new MPAM50 centroids. MPAM50 was then tested on 19 independent datasets (obtained using various expression profiling technologies) containing 9637 samples. Overall good agreement was observed between the PAM50- and MPAM50-assigned subtypes with a median accuracy of 0.792, which (we show) is comparable with the median concordance between various implementations of PAM50. Additionally, MPAM50- and PAM50-assigned intrinsic subtypes were found to agree comparably with the reported clinical subtypes. Also, survival analyses indicated that MPAM50 preserves the prognostic value of the intrinsic subtypes. These observations demonstrate that MPAM50 can replace PAM50 without loss of performance. On the other hand, MPAM50 was compared with 2 previously published single-sample classifiers, and with 3 alternative modified PAM50 approaches. The results indicated a superior performance by MPAM50. Conclusions MPAM50 is a robust, simple, and accurate single-sample classifier of intrinsic subtypes of breast cancer.
Collapse
Affiliation(s)
| | - Yi-Kuo Yu
- Yi-Kuo Yu, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, 8600 Rockville Pike, Bethesda, MD 20894, USA.
| |
Collapse
|
14
|
Ran R, Ma Y, Wang H, Yang J, Yang J. Treatment strategies for hormone receptor-positive, human epidermal growth factor receptor 2-positive (HR+/HER2+) metastatic breast cancer: A review. Front Oncol 2022; 12:975463. [PMID: 36620573 PMCID: PMC9822772 DOI: 10.3389/fonc.2022.975463] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/11/2022] [Indexed: 12/25/2022] Open
Abstract
Hormone receptor-positive HER2-positive (HR+/HER2+) metastatic breast cancer (MBC) is a unique subtype of breast cancer. Most current guidelines recommend that combination regimens based on anti-HER2 therapy should be used as first-line treatment for HER2+ MBC, irrespective of HR status. Endocrine therapy can be applied as maintenance therapy for patients who are intolerant to chemotherapy or post-chemotherapy. Increasing evidence suggests that complex molecular crosstalk between HR and HER2 pathways may affect the sensitivity to both HER2-targeted and endocrine therapy in patients with HR+/HER2+ breast cancer. Recent research and clinical trials have revealed that a combination of endocrine therapy and anti-HER2 approaches without chemotherapy provides along-term disease control for some patients, but the challenge lies in how to accurately identify the subsets of patients who can benefit from such a de-chemotherapy treatment strategy. In this review, we aim to summarize the results of preclinical and clinical studies in HR+/HER2+ MBC and discuss the possibility of sparing chemotherapy in this subgroup of patients.
Collapse
Affiliation(s)
| | | | | | - Jin Yang
- *Correspondence: Jin Yang, ; Jiao Yang,
| | - Jiao Yang
- *Correspondence: Jin Yang, ; Jiao Yang,
| |
Collapse
|
15
|
Carausu M, Carton M, Diéras V, Petit T, Guiu S, Gonçalves A, Augereau P, Ferrero JM, Levy C, Ung M, Desmoulins I, Debled M, Bachelot T, Pistilli B, Frenel JS, Mailliez A, Chevrot M, Cabel L. Association of Endocrine Therapy for HR+/ERBB2+ Metastatic Breast Cancer With Survival Outcomes. JAMA Netw Open 2022; 5:e2247154. [PMID: 36520434 PMCID: PMC9856509 DOI: 10.1001/jamanetworkopen.2022.47154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE Evidence suggests that patients with human epidermal growth factor receptor 2-positive (ERBB2+ [formerly HER2+]) metastatic breast cancer (MBC) have different clinical characteristics and outcomes according to their hormone receptor (HR) status. The place of endocrine therapy (ET) for patients with HR+/ERBB2+ is still not clearly defined in this setting. OBJECTIVE To evaluate the association of HR status and first-line inclusion of ET with outcomes among patients with ERBB2+ MBC. DESIGN, SETTING, AND PARTICIPANTS This cohort study was an analysis of clinical data from the French clinical Epidemiological Strategy and Medical Economics (ESME) cohort, including patients with MBC who started treatment between 2008 and 2017. The last date of follow-up was June 18, 2020. Data were analyzed from May 2021 to May 2022. EXPOSURES Patients were treated with first-line ERBB2-targeted therapy and either chemotherapy (CT) with or without ET or ET alone. For the study of the association of maintenance ET with outcomes, we included patients treated with first-line ERBB2-targeted therapy with CT and with or without maintenance ET. MAIN OUTCOMES AND MEASURES Median overall survival (OS) and median first-line progression-free survival (PFS) were reported using the Kaplan-Meier method. Cox proportional hazards models and a propensity score were constructed to report and adjust for prognostic factors. Multivariable analysis included age at MBC, time to MBC, number of metastatic sites, type of metastases, and Eastern Cooperative Oncology Group performance status. RESULTS Among 4145 women with ERBB2+ MBC, 2696 patients had HR+ (median [IQR] age, 58.0 [47.0-67.0] years) and 1449 patients had HR- (56.0 [47.0-64.0] years) tumors. The median OS for patients with HR+ vs HR- tumors was 55.9 months (95% CI, 53.7-59.4 months) vs 42.0 months (95% CI, 38.8-45.2 months), confirmed in multivariable analysis (hazard ratio, 1.40; 95% CI, 1.26-1.56; P < .001). The median PFS for patients with HR+ vs HR- tumors was 12.2 months (95% CI, 11.5-12.9 months) vs 9.8 months (95% CI, 9.2-11.0 months; P = .01), and the HR was 1.15 (95% CI, 1.06-1.26; P < .001). In multivariable analysis, no significant difference was found in OS or PFS for 1520 patients treated with ERBB2-targeted therapy with CT and with or without ET vs 203 patients receiving ERBB2-targeted therapy with ET, regardless of type of ERBB2-targeted therapy (trastuzumab or trastuzumab with pertuzumab). This result was confirmed by matching patients using a propensity score. Using the time-dependent ET variable among patients with ERBB2-targeted therapy with CT, those with maintenance ET had significantly better PFS (hazard ratio, 0.70; 95% CI, 0.60-0.82; P < .001) and OS (hazard ratio, 0.47; 95% CI, 0.39-0.57; P < .001). CONCLUSIONS AND RELEVANCE These results suggest that ET-containing first-line regimens may be associated with benefits among a subgroup of patients with HR+/ERBB2+ MBC.
Collapse
Affiliation(s)
- Marcela Carausu
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Matthieu Carton
- Department of Biostatistics, Institut Curie, Saint-Cloud, France
| | - Véronique Diéras
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Thierry Petit
- Department of Medical Oncology, Centre Paul Strauss/ICANS, Strasbourg, France
| | - Séverine Guiu
- Department of Medical Oncology, Institut régional du Cancer Montpellier, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Paule Augereau
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Angers, France
| | - Jean Marc Ferrero
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Mony Ung
- Department of Medical Oncology, Institut Claudius Regaud, Toulouse, France
| | - Isabelle Desmoulins
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Marc Debled
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Barbara Pistilli
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | | | - Audrey Mailliez
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Michaël Chevrot
- Health Data and Partnership Department, Unicancer, Paris, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| |
Collapse
|
16
|
Risk Stratification for Breast Cancer Patient by Simultaneous Learning of Molecular Subtype and Survival Outcome Using Genetic Algorithm-Based Gene Set Selection. Cancers (Basel) 2022; 14:cancers14174120. [PMID: 36077657 PMCID: PMC9454699 DOI: 10.3390/cancers14174120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Patient stratification is clinically important because it allows us to understand the characteristics and establish treatment strategies for a group. Transcriptomic data play an important role in determining molecular subtypes and predicting survival. In the case of breast cancer, although the order of prognosis according to molecular subtypes is well known, there is heterogeneity even within a subtype. Therefore, patient stratification considering both molecular subtypes and survival outcomes is required. In this study, a methodology to handle this problem is presented. A genetic algorithm is used to select a set of genes, and a risk score is assigned to each patient using their expression level. According to the risk score, patients are ordered and stratified considering molecular subtypes and survival outcomes. Consequently, informative genes for patient stratification with respect to both aspects could be nominated, and the usefulness of the risk score was shown through comparison with other indicators. Abstract Patient stratification is a clinically important task because it allows us to establish and develop efficient treatment strategies for particular groups of patients. Molecular subtypes have been successfully defined using transcriptomic profiles, and they are used effectively in clinical practice, e.g., PAM50 subtypes of breast cancer. Survival prediction contributed to understanding diseases and also identifying genes related to prognosis. It is desirable to stratify patients considering these two aspects simultaneously. However, there are no methods for patient stratification that consider molecular subtypes and survival outcomes at once. Here, we propose a methodology to deal with the problem. A genetic algorithm is used to select a gene set from transcriptome data, and their expression quantities are utilized to assign a risk score to each patient. The patients are ordered and stratified according to the score. A gene set was selected by our method on a breast cancer cohort (TCGA-BRCA), and we examined its clinical utility using an independent cohort (SCAN-B). In this experiment, our method was successful in stratifying patients with respect to both molecular subtype and survival outcome. We demonstrated that the orders of patients were consistent across repeated experiments, and prognostic genes were successfully nominated. Additionally, it was observed that the risk score can be used to evaluate the molecular aggressiveness of individual patients.
Collapse
|
17
|
Understanding Breast Cancers through Spatial and High-Resolution Visualization Using Imaging Technologies. Cancers (Basel) 2022; 14:cancers14174080. [PMID: 36077616 PMCID: PMC9454728 DOI: 10.3390/cancers14174080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer is the most common cancer affecting women worldwide. Although many analyses and treatments have traditionally targeted the breast cancer cells themselves, recent studies have focused on investigating entire cancer tissues, including breast cancer cells. To understand the structure of breast cancer tissues, including breast cancer cells, it is necessary to investigate the three-dimensional location of the cells and/or proteins comprising the tissues and to clarify the relationship between the three-dimensional structure and malignant transformation or metastasis of breast cancers. In this review, we aim to summarize the methods for analyzing the three-dimensional structure of breast cancer tissue, paying particular attention to the recent technological advances in the combination of the tissue-clearing method and optical three-dimensional imaging. We also aimed to identify the latest methods for exploring the relationship between the three-dimensional cell arrangement in breast cancer tissues and the gene expression of each cell. Finally, we aimed to describe the three-dimensional imaging features of breast cancer tissues using noninvasive photoacoustic imaging methods.
Collapse
|
18
|
Molecular Subtyping of Invasive Breast Cancer Using a PAM50-Based Multigene Expression Test-Comparison with Molecular-Like Subtyping by Tumor Grade/Immunohistochemistry and Influence on Oncologist's Decision on Systemic Therapy in a Real-World Setting. Int J Mol Sci 2022; 23:ijms23158716. [PMID: 35955851 PMCID: PMC9368794 DOI: 10.3390/ijms23158716] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
In intermediate risk hormone receptor (HR) positive, HER2 negative breast cancer (BC), the decision regarding adjuvant chemotherapy might be facilitated by multigene expression tests. In all, 142 intermediate risk BCs were investigated using the PAM50-based multigene expression test Prosigna® in a prospective multicentric study. In 119/142 cases, Prosigna® molecular subtyping was compared with local and two central (C1 and C6) molecular-like subtypes relying on both immunohistochemistry (IHC; HRs, HER2, Ki-67) and IHC + tumor grade (IHC+G) subtyping. According to local IHC, 35.4% were Luminal A-like and 64.6% Luminal B-like subtypes (local IHC+G subtype: 31.9% Luminal A-like; 68.1% Luminal B-like). In contrast to local and C1 subtyping, C6 classified >2/3 of cases as Luminal A-like. Pairwise agreement between Prosigna® subtyping and molecular-like subtypes was fair to moderate depending on molecular-like subtyping method and center. The best agreement was observed between Prosigna® (53.8% Luminal A; 44.5% Luminal B) and C1 surrogate subtyping (Cohen’s kappa = 0.455). Adjuvant chemotherapy was suggested to 44.2% and 88.6% of Prosigna® Luminal A and Luminal B cases, respectively. Out of all Luminal A-like cases (locally IHC/IHC+G subtyping), adjuvant chemotherapy was recommended if Prosigna® testing classified as Prosigna® Luminal A at high / intermediate risk or upgraded to Prosigna® Luminal B.
Collapse
|
19
|
Prognostic significance of different molecular typing methods and immune status based on RNA sequencing in HR-positive and HER2-negative early-stage breast cancer. BMC Cancer 2022; 22:548. [PMID: 35568835 PMCID: PMC9107692 DOI: 10.1186/s12885-022-09656-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 05/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study was conducted to evaluate the prognostic significance of different molecular typing methods and immune status based on RNA sequencing (RNA-seq) in hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative (HR + /HER2-) early-stage breast cancer and develop a modified immunohistochemistry (IHC)-based surrogate for intrinsic subtype analysis. METHODS The gene expression profiles of samples from 87 HR + /HER2- early-stage breast cancer patients were evaluated using the RNA-seq of Oncotype Dx recurrence score (RS), PAM50 risk of recurrence (ROR), and immune score. Intrinsic tumor subtypes were determined using both PAM50- and IHC-based detection of estrogen receptor, progesterone receptor, Ki-67, epidermal growth factor receptor, and cytokeratins 14 and 5/6. Prognostic variables were analyzed through Cox regression analysis of disease-free survival (DFS) and distant metastasis-free survival (DMFS). RESULTS Survival analysis showed that ROR better predicted recurrence and distant metastasis compared to RS (for DFS: ROR, P = 0.000; RS, P = 0.027; for DMFS, ROR, P = 0.047; RS, P = 0.621). Patients with HR + /HER2- early-stage breast cancer was classified into the luminal A, luminal B, HER2-enriched, and basal-like subtypes by PAM50. Basal-like subgroups showed the shortest DFS and DMFS. A modified IHC-based surrogate for intrinsic subtype analysis improved the concordance with PAM50 from 66.7% to 73.6%, particularly for basal-like subtype identification. High level of TILs and high expression of immune genes predicted poor prognosis. Multi-factor Cox analysis showed that IHC-based basal-like markers were the only independent factors affecting DMFS. CONCLUSIONS Prognosis is better evaluated by PAM50 ROR in early-stage HR + /HER2- breast cancer and significantly differs among intrinsic subtypes. The modified IHC-based subtype can improve the basal-like subtype identification of PAM50. High immunity status and IHC-based basal-like markers are negative prognostic factors.
Collapse
|
20
|
Llera AS, Abdelhay ESFW, Artagaveytia N, Daneri-Navarro A, Müller B, Velazquez C, Alcoba EB, Alonso I, Alves da Quinta DB, Binato R, Bravo AI, Camejo N, Carraro DM, Castro M, Castro-Cervantes JM, Cataldi S, Cayota A, Cerda M, Colombo A, Crocamo S, Del Toro-Arreola A, Delgadillo-Cisterna R, Delgado L, Dreyer-Breitenbach M, Fejerman L, Fernández EA, Fernández J, Fernández W, Franco-Topete RA, Gabay C, Gaete F, Garibay-Escobar A, Gómez J, Greif G, Gross TG, Guerrero M, Henderson MK, Lopez-Muñoz ME, Lopez-Vazquez A, Maldonado S, Morán-Mendoza AJ, Nagai MA, Oceguera-Villanueva A, Ortiz-Martínez MA, Quintero J, Quintero-Ramos A, Reis RM, Retamales J, Rivera-Claisse E, Rocha D, Rodríguez R, Rosales C, Salas-González E, Sanchotena V, Segovia L, Sendoya JM, Silva-García AA, Trinchero A, Valenzuela O, Vedham V, Zagame L, Podhajcer OL. The Transcriptomic Portrait of Locally Advanced Breast Cancer and Its Prognostic Value in a Multi-Country Cohort of Latin American Patients. Front Oncol 2022; 12:835626. [PMID: 35433488 PMCID: PMC9007037 DOI: 10.3389/fonc.2022.835626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Purposes Most molecular-based published studies on breast cancer do not adequately represent the unique and diverse genetic admixture of the Latin American population. Searching for similarities and differences in molecular pathways associated with these tumors and evaluating its impact on prognosis may help to select better therapeutic approaches. Patients and Methods We collected clinical, pathological, and transcriptomic data of a multi-country Latin American cohort of 1,071 stage II-III breast cancer patients of the Molecular Profile of Breast Cancer Study (MPBCS) cohort. The 5-year prognostic ability of intrinsic (transcriptomic-based) PAM50 and immunohistochemical classifications, both at the cancer-specific (OSC) and disease-free survival (DFS) stages, was compared. Pathway analyses (GSEA, GSVA and MetaCore) were performed to explore differences among intrinsic subtypes. Results PAM50 classification of the MPBCS cohort defined 42·6% of tumors as LumA, 21·3% as LumB, 13·3% as HER2E and 16·6% as Basal. Both OSC and DFS for LumA tumors were significantly better than for other subtypes, while Basal tumors had the worst prognosis. While the prognostic power of traditional subtypes calculated with hormone receptors (HR), HER2 and Ki67 determinations showed an acceptable performance, PAM50-derived risk of recurrence best discriminated low, intermediate and high-risk groups. Transcriptomic pathway analysis showed high proliferation (i.e. cell cycle control and DNA damage repair) associated with LumB, HER2E and Basal tumors, and a strong dependency on the estrogen pathway for LumA. Terms related to both innate and adaptive immune responses were seen predominantly upregulated in Basal tumors, and, to a lesser extent, in HER2E, with respect to LumA and B tumors. Conclusions This is the first study that assesses molecular features at the transcriptomic level in a multicountry Latin American breast cancer patient cohort. Hormone-related and proliferation pathways that predominate in PAM50 and other breast cancer molecular classifications are also the main tumor-driving mechanisms in this cohort and have prognostic power. The immune-related features seen in the most aggressive subtypes may pave the way for therapeutic approaches not yet disseminated in Latin America. Clinical Trial Registration ClinicalTrials.gov (Identifier: NCT02326857).
Collapse
Affiliation(s)
- Andrea Sabina Llera
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | | | - Nora Artagaveytia
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | | | | | | | - Elsa B Alcoba
- Hospital Municipal de Oncología María Curie, Buenos Aires, Argentina
| | - Isabel Alonso
- Centro Hospitalario Pereira Rossell, Montevideo, Uruguay
| | - Daniela B Alves da Quinta
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina.,Universidad Argentina de la Empresa (UADE), Instituto de Tecnología (INTEC), Buenos Aires, Argentina
| | - Renata Binato
- Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | - Natalia Camejo
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | - Dirce Maria Carraro
- Laboratory of Genomics and Molecular Biology/Centro Internacional de Pesquisa (CIPE), AC Camargo Cancer Center, Sao Paulo, Brazil
| | - Mónica Castro
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | | | | | | | - Mauricio Cerda
- Integrative Biology Program, Instituto de Ciencias Biomédicas (ICBM), Centro de Informática Médica y Telemedicina, Facultad de Medicina, Instituto de Neurociencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo
- Department of Pathology, Facultad de Medicina y Hospital Clínico, Universidad de Chile, Santiago, Chile
| | - Susanne Crocamo
- Oncology Department, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | | | - Lucía Delgado
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | - Marisa Dreyer-Breitenbach
- Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laura Fejerman
- Department of Public Health Sciences and Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| | - Elmer A Fernández
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas [Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE) CONICET/Universidad Católica de Córdoba], Córdoba, Argentina.,Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | | | - Ramón A Franco-Topete
- Organismo Público Descentralizado (OPD), Hospital Civil de Guadalajara, Universidad de Guadalajara, Guadalajara, Mexico
| | - Carolina Gabay
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | | | | | - Jorge Gómez
- Texas A&M University, Houston, TX, United States
| | | | - Thomas G Gross
- Center for Global Health, National Cancer Institute, Rockville, MD, United States
| | | | - Marianne K Henderson
- Center for Global Health, National Cancer Institute, Rockville, MD, United States
| | | | | | | | | | - Maria Aparecida Nagai
- Center for Translational Research in Oncology, Cancer Institute of São Paulo (ICESP), Sao Paulo University Medical School, Sao Paulo, Brazil
| | | | | | | | | | - Rui M Reis
- Molecular Oncology Research Center, Hospital de Câncer de Barretos, Barretos, Brazil
| | - Javier Retamales
- Grupo Oncológico Cooperativo Chileno de Investigación, Santiago, Chile
| | | | - Darío Rocha
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Cristina Rosales
- Hospital Municipal de Oncología María Curie, Buenos Aires, Argentina
| | | | | | | | - Juan Martín Sendoya
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | - Aida A Silva-García
- Organismo Público Descentralizado (OPD), Hospital Civil de Guadalajara, Universidad de Guadalajara, Guadalajara, Mexico
| | | | | | - Vidya Vedham
- Center for Global Health, National Cancer Institute, Rockville, MD, United States
| | - Livia Zagame
- Instituto Jalisciense de Cancerologia, Guadalajara, Mexico
| | | | - Osvaldo L Podhajcer
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| |
Collapse
|
21
|
Li F, Ju Q, Gao C, Li J, Wang X, Yan M, Zhang L, Huang M, Long Q, Jin X, Li N. Association of HER-2/CEP17 Ratio and HER-2 Copy Number With pCR Rate in HER-2-Positive Breast Cancer After Dual-Target Neoadjuvant Therapy With Trastuzumab and Pertuzumab. Front Oncol 2022; 12:819818. [PMID: 35311143 PMCID: PMC8931257 DOI: 10.3389/fonc.2022.819818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/11/2022] [Indexed: 12/02/2022] Open
Abstract
Objective To explore the correlation between HER-2 status and pathological complete response (pCR) in HER-2-positive breast cancer after dual anti-HER-2 neoadjuvant therapy with trastuzumab and pertuzumab. Methods A total of 57 HER-2-positive breast cancer patients admitted to the Second Affiliated Hospital of Anhui Medical University and Xijing Hospital Affiliated to Air Force Military Medical University, between January 1, 2019 and September 30, 2020, were enrolled in this multicenter retrospective study. HER-2 status, including HER-2/CEP17 ratio and HER-2/cell number ratio, was detected by FISH. The correlation between HER-2 status/clinicopathological data and pCR was analyzed. The ROC curve was drawn to determine the cutoff value. Results IHC assessment revealed 40 (70.18%) patients with IHC 3+ and 17 (29.82%) with IHC 2+. 41/57 (71.93%) patients achieved pCR. FISH revealed that the ratio of HER-2/chromosome 17 centromere (HER-2/CEP17) (p<0.001) and the ratio of HER-2/cell number (p<0.001) was significantly correlated with the pCR rate. ROC analysis showed that patients with an HER-2/CEP17 ratio ≥4.495 or HER-2/cell number ≥11.650 have a high pCR rate after dual anti-HER-2 neoadjuvant therapy, suggesting its predictive significance. Conclusion The response to dual-targeted neoadjuvant therapy with trastuzumab and pertuzumab was adequate in hormone receptor-negative, HER-2-positive breast cancer patients. HER-2/CEP17 ratio and HER-2/cell number ratio were crucial for predicting efficacy.
Collapse
Affiliation(s)
- Fanfan Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Fanfan Li, ; Nanlin Li,
| | - Qian Ju
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cong Gao
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jingjing Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaolei Wang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Yan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liying Zhang
- Department of Pathology, Xijing Hospital of Air Force Military Medical University, Xi ‘an, China
| | - Meiling Huang
- Department of Breast Surgery, Xijing Hospital of Air Force Military Medical University, Xi ‘an, China
| | - Qihe Long
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiangting Jin
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Nanlin Li
- Department of Breast Surgery, Xijing Hospital of Air Force Military Medical University, Xi ‘an, China
- *Correspondence: Fanfan Li, ; Nanlin Li,
| |
Collapse
|
22
|
Bueno Muiño C, Martín M, del Monte-Millán M, García-Saénz JÁ, López-Tarruella S. HER2+ Breast Cancer Escalation and De-Escalation Trial Design: Potential Role of Intrinsic Subtyping. Cancers (Basel) 2022; 14:512. [PMID: 35158778 PMCID: PMC8833556 DOI: 10.3390/cancers14030512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/09/2022] [Accepted: 01/16/2022] [Indexed: 12/29/2022] Open
Abstract
Long-term outcomes in breast cancer patients differ based on the molecular subtype, with HER2-E being the most aggressive one. Advances in clinical practice have dramatically shifted HER2+ breast cancer prognosis. Risk adapted strategies to individualize therapies are necessary. De-escalation approaches have been encouraged based on the risks of clinical-pathological factors. Molecular gene subtyping could further accurately define HER2 addicted tumours that are sensitive to anti-HER2 therapies, thus sparing unnecessary treatments. The transition from immunochemistry to molecular profiling in HER2+ breast cancer is discussed.
Collapse
Affiliation(s)
- Coralia Bueno Muiño
- Medical Oncology Department, Hospital Infanta Cristina (Parla), Fundación de Investigación Biomédica del H.U. Puerta de Hierro, Majadahonda, 28009 Madrid, Spain;
| | - Miguel Martín
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigacion Sanitaria Gregorio Marañon (IiSGM), CIBERONC, Geicam, Universidad Complutense, 28007 Madrid, Spain;
| | - María del Monte-Millán
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigacion Sanitaria Gregorio Marañon (IiSGM) CIBERONC, 28007 Madrid, Spain;
| | - José Ángel García-Saénz
- Medical Oncology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), CIBERONC, 28040 Madrid, Spain;
| | - Sara López-Tarruella
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigacion Sanitaria Gregorio Marañon (IiSGM), CIBERONC, Geicam, Universidad Complutense, 28007 Madrid, Spain;
| |
Collapse
|
23
|
Zhao D, Fu X, Rohr J, Wang Y, Li M, Zhang X, Qin J, Xu M, Li C, Sun G, Wang Z, Guo S. Poor histologic tumor response after adjuvant therapy in basal-like HER2-positive breast carcinoma. Pathol Res Pract 2021; 228:153677. [PMID: 34775151 DOI: 10.1016/j.prp.2021.153677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 12/21/2022]
Abstract
AIMS HER2-positive breast carcinomas are all treated with first-line anti-HER2 therapy. However, immunohistochemical and molecular profiling demonstrates significant heterogeneity among HER2-positive carcinomas. Basal-like HER2-positive breast carcinomas are poorly differentiated from pure HER2-positive breast carcinomas. MATERIALS AND METHODS Seventy-five patients with HER2-positive, ER- and PR-negative breast carcinomas who received anti-HER2 based neoadjuvant therapy were retrospectively analyzed. Thirty-seven cases were classified as basal-like HER2-positive breast carcinoma with any positivity for CK5/6, and thirty-eight cases were classified as pure HER2-positive breast carcinoma with completely negativity for CK5/6. The clinicopathological features and tumor responses after neoadjuvant therapy and outcomes were analyzed. RESULTS Compared to non-basal HER2-positive breast carcinoma, basal-like HER2-positive breast carcinoma showed distinctive histologic features including poor differentiation and syncytial tumor cells with pushing, invasive borders and a significantly higher proportion of apocrine metaplasia. They also demonstrated significantly higher histologic grade; 18/37 (48.6%) of basal-like carcinomas were grade 3, whereas only 5/38 (13.2%) of non-basal carcinomas were grade 3 (p = 0.001), Furthermore, basal-like HER2-positive breast carcinomas were more likely to be positive or completely negative for p53 (p = 0.009), and demonstrated a higher percentage of TP53 mutation (p = 0.17). These tumors were less responsive to anti-HER2 based neoadjuvant therapy, with Miller-Payne grades 1-3 higher than pure HER2-positive breast carcinoma (25/37 [67.6%] vs 16/38 [42.1%]), and the percentage of grade 4-5 was lower (12/37 [32.4%] vs 22/38 [57.9%]; p = 0.027). CONCLUSIONS Basal-like HER2-positive breast carcinoma has distinctive clinicopathological features and less histologic tumor response after neoadjuvant therapy. There is urgent need to recognize basal-like HER2-positive breast carcinoma to be treated precisely.
Collapse
Affiliation(s)
- Danhui Zhao
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Xin Fu
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Joseph Rohr
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, 68105, NE, USA
| | - Yingmei Wang
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Mingyang Li
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Xiuming Zhang
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Junhui Qin
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Mengwei Xu
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Chao Li
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Guorui Sun
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China
| | - Zhe Wang
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China.
| | - Shuangping Guo
- Department of Pathology, the First Affinity Hospital of the Air Force Military Medical University, Xi'an, Shaan Xi Province, 710032, China.
| |
Collapse
|
24
|
Hao J, Lyu Y, Zou J, Zhang Y, Xie S, Jing L, Tang F, Lyu J, Zhang W, Zhang J, Wang X, Chen K, Zhang J. Improving Prognosis of Surrogate Assay for Breast Cancer Patients by Absolute Quantitation of Ki67 Protein Levels Using Quantitative Dot Blot (QDB) Method. Front Oncol 2021; 11:737781. [PMID: 34604077 PMCID: PMC8485584 DOI: 10.3389/fonc.2021.737781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022] Open
Abstract
Background Immunohistochemistry (IHC)-based surrogate assay is the prevailing method in daily clinical practice to determine the necessity of chemotherapy for Luminal-like breast cancer patients worldwide. It relies on Ki67 scores to separate Luminal A-like from Luminal B-like breast cancer subtypes. Yet, IHC-based Ki67 assessment is known to be plagued with subjectivity and inconsistency to undermine the performance of the surrogate assay. A novel method needs to be explored to improve the clinical utility of Ki67 in daily clinical practice. Materials and Methods The Ki67 protein levels in a cohort of 253 specimens were assessed with IHC and quantitative dot blot (QDB) methods, respectively, and used to assign these specimens into Luminal A-like and Luminal B-like subtypes accordingly. Their performances were compared with the Kaplan–Meier, univariate, and multivariate survival analyses of the overall survival (OS) of Luminal-like patients. Results The surrogate assay based on absolutely quantitated Ki67 levels (cutoff at 2.31 nmol/g) subtyped the Luminal-like patients more effectively than that based on Ki67 scores (cutoff at 14%) (Log rank test, p = 0.00052 vs. p = 0.031). It is also correlated better with OS in multivariate survival analysis [hazard ratio (HR) at 6.89 (95% CI: 2.66–17.84, p = 0.0001) vs. 2.14 (95% CI: 0.89–5.11, p = 0.087)]. Conclusions Our study showed that the performance of the surrogate assay may be improved significantly by measuring Ki67 levels absolutely, quantitatively, and objectively using the QDB method.
Collapse
Affiliation(s)
- Junmei Hao
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Yan Lyu
- Yantai Quanticision Diagnostics, Inc., Yantai, China
| | - Jiarui Zou
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Yunyun Zhang
- Yantai Quanticision Diagnostics, Inc., Yantai, China
| | - Shuishan Xie
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Lili Jing
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Fangrong Tang
- Yantai Quanticision Diagnostics, Inc., Yantai, China
| | - Jiahong Lyu
- Yantai Quanticision Diagnostics, Inc., Yantai, China
| | - Wenfeng Zhang
- Yantai Quanticision Diagnostics, Inc., Yantai, China
| | - Jianbo Zhang
- Yantai Quanticision Diagnostics, Inc., Yantai, China
| | - Xunting Wang
- Department of Imaging, Linglong Yingcheng Hospital, Zhaoyuan, China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Tumor Pathology, Zhengzhou, China
| | - Jiandi Zhang
- Yantai Quanticision Diagnostics, Inc., Yantai, China
| |
Collapse
|
25
|
A single-cell and spatially resolved atlas of human breast cancers. Nat Genet 2021; 53:1334-1347. [PMID: 34493872 PMCID: PMC9044823 DOI: 10.1038/s41588-021-00911-1] [Citation(s) in RCA: 595] [Impact Index Per Article: 198.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 07/08/2021] [Indexed: 01/05/2023]
Abstract
Breast cancers are complex cellular ecosystems where heterotypic interactions play central roles in disease progression and response to therapy. However, our knowledge of their cellular composition and organization is limited. Here we present a single-cell and spatially resolved transcriptomics analysis of human breast cancers. We developed a single-cell method of intrinsic subtype classification (SCSubtype) to reveal recurrent neoplastic cell heterogeneity. Immunophenotyping using cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) provides high-resolution immune profiles, including new PD-L1/PD-L2+ macrophage populations associated with clinical outcome. Mesenchymal cells displayed diverse functions and cell-surface protein expression through differentiation within three major lineages. Stromal-immune niches were spatially organized in tumors, offering insights into antitumor immune regulation. Using single-cell signatures, we deconvoluted large breast cancer cohorts to stratify them into nine clusters, termed 'ecotypes', with unique cellular compositions and clinical outcomes. This study provides a comprehensive transcriptional atlas of the cellular architecture of breast cancer.
Collapse
|
26
|
Zhang Y, Zhang D, Meng Q, Liu Z, Xie H, Liu L, Xu F, Chen X. Precision treatment exploration of breast cancer based on heterogeneity analysis of lncRNAs at the single-cell level. BMC Cancer 2021; 21:918. [PMID: 34388989 PMCID: PMC8361656 DOI: 10.1186/s12885-021-08617-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Breast cancer (BC) is a complex disease with high heterogeneity, which often leads to great differences in treatment results. Current common molecular typing method is PAM50, which shows positive results for precision medicine; however, room for improvement still remains because of the different prognoses of subtypes. Therefore, in this article, we used lncRNAs, which are more tissue-specific and developmental stage-specific than other RNAs, as typing markers and combined single-cell expression profiles to retype BC, to provide a new method for BC classification and explore new precise therapeutic strategies based on this method. METHODS Based on lncRNA expression profiles of 317 single cells from 11 BC patients, SC3 was used to retype BC, and differential expression analysis and enrichment analysis were performed to identify biological characteristics of new subtypes. The results were validated for survival analysis using data from TCGA. Then, the downstream regulatory genes of lncRNA markers of each subtype were searched by expression correlation analysis, and these genes were used as targets to screen therapeutic drugs, thus proposing new precision treatment strategies according to the different subtype compositions of patients. RESULTS Seven lncRNA subtypes and their specific biological characteristics are obtained. Then, 57 targets and 210 drugs of 7 subtypes were acquired. New precision medicine strategies were proposed according to the different compositions of patient subtypes. CONCLUSIONS For patients with different subtype compositions, we propose a strategy to select different drugs for different patients, which means using drugs targeting multi subtype or combinations of drugs targeting a single subtype to simultaneously kill different cancer cells by personalized treatment, thus reducing the possibility of drug resistance and even recurrence.
Collapse
Affiliation(s)
- Yan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, P. R. China
| | - Denan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, P. R. China
| | - Qingkang Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, P. R. China
| | - Ziqi Liu
- Department of Pharmacy, The First Affiliated Hospital, Harbin Medical University, Harbin, 150001, Heilongjiang Province, P. R. China
| | - Hongbo Xie
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, P. R. China
| | - Lei Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, P. R. China
| | - Fei Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, P. R. China
| | - Xiujie Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, P. R. China.
| |
Collapse
|
27
|
Schoenherr RM, Voytovich UJ, Whiteaker JR, Gadi VK, Gralow JR, Paulovich AG. Quantification of Estrogen Receptor, Progesterone Receptor, and Human Epidermal Growth Factor Receptor 2 Protein Expression in Bone Biopsies by Targeted Mass Spectrometry without Acid Decalcification. Clin Chem 2021; 66:1459-1461. [PMID: 33106847 DOI: 10.1093/clinchem/hvaa220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2020] [Indexed: 01/24/2023]
Affiliation(s)
- Regine M Schoenherr
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Uliana J Voytovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jeffrey R Whiteaker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Vijayakrishna K Gadi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- University of Washington School of Medicine, Seattle, WA
- Seattle Cancer Care Alliance, Seattle, WA
- Department of Medicine, University of Illinois at Chicago, IL
| | - Julie R Gralow
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- University of Washington School of Medicine, Seattle, WA
- Seattle Cancer Care Alliance, Seattle, WA
| | - Amanda G Paulovich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
28
|
Wang L, Li Q, Aushev VN, Neugut AI, Santella RM, Teitelbaum S, Chen J. PAM50- and immunohistochemistry-based subtypes of breast cancer and their relationship with breast cancer mortality in a population-based study. Breast Cancer 2021; 28:1235-1242. [PMID: 34003448 DOI: 10.1007/s12282-021-01261-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/06/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE We evaluated the prognostic ability of immunohistochemistry (IHC)-based vs. PAM50-based subtypes for breast cancer mortality in a population-based study of breast cancer. METHODS We included a total of 463 breast cancer cases from the population-based Long Island Breast Cancer Study Project (LIBCSP). IHC-based markers were abstracted from the medical records, while the PAM50-based intrinsic subtypes were assessed from tumor tissues using NanoString nCounter® Analysis System. Cox proportional hazards models were used to estimate hazards ratios (HRs) for breast cancer-specific mortality associated with subtypes. RESULTS For IHC-based hormone receptor-positive (HR+) tumors (n = 361), 68.7% were classified as luminal subtypes by PAM50; for HR- tumors (n = 102), 95.1% were classified as non-luminal subtypes. Compared to HR+/HER2- subtype, HR- patients had significantly higher breast cancer mortality (HR-/HER2+: HR = 2.84, 95% CI = 1.58-5.11; triple-negative breast cancer: HR = 2.42, 95% CI = 1.44-4.06). Compared to luminal A, a higher mortality rate was observed for all other PAM50-based subtypes: luminal B (HR = 4.03, 95% CI = 1.97-8.22), HER2-enriched (HR = 6.82, 95% CI = 3.29-14.14) and basal-like (HR = 4.71, 95% CI = 2.24-9.93). Additional subtyping of HR+ patients by PAM50 provided future risk stratification where luminal B patients in this group had significant higher mortality than luminal A patients (HR = 3.93, 95% CI = 1.92-8.03). Similar results were also observed among 291 HR+/HER2- patients, but not among the HR- patients. CONCLUSIONS Our study suggests that for HR+ patients, especially HR+/HER2- patients, additional PAM50-based subtyping would provide better prognostic stratification and improve disease management.
Collapse
Affiliation(s)
- Lin Wang
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY, 10029 , USA.,Department of Oncology, The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Li
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY, 10029 , USA
| | - Vasily N Aushev
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY, 10029 , USA
| | - Alfred I Neugut
- Department of Medicine and Epidemiology, Columbia University, New York, USA
| | - Regina M Santella
- Division of Environmental Health Sciences, Joseph L. Mailman School of Public Health, Columbia University, New York, USA
| | - Susan Teitelbaum
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY, 10029 , USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, Box 1057, New York, NY, 10029 , USA.
| |
Collapse
|
29
|
Valle F, Osella M, Caselle M. A Topic Modeling Analysis of TCGA Breast and Lung Cancer Transcriptomic Data. Cancers (Basel) 2020; 12:E3799. [PMID: 33339347 PMCID: PMC7766023 DOI: 10.3390/cancers12123799] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 01/18/2023] Open
Abstract
Topic modeling is a widely used technique to extract relevant information from large arrays of data. The problem of finding a topic structure in a dataset was recently recognized to be analogous to the community detection problem in network theory. Leveraging on this analogy, a new class of topic modeling strategies has been introduced to overcome some of the limitations of classical methods. This paper applies these recent ideas to TCGA transcriptomic data on breast and lung cancer. The established cancer subtype organization is well reconstructed in the inferred latent topic structure. Moreover, we identify specific topics that are enriched in genes known to play a role in the corresponding disease and are strongly related to the survival probability of patients. Finally, we show that a simple neural network classifier operating in the low dimensional topic space is able to predict with high accuracy the cancer subtype of a test expression sample.
Collapse
Affiliation(s)
- Filippo Valle
- Physics Department, University of Turin and INFN, via P. Giuria 1, 10125 Turin, Italy; (M.O.); (M.C.)
| | | | | |
Collapse
|
30
|
Kim JY, Park K, Im SA, Jung KH, Sohn J, Lee KS, Kim JH, Yang Y, Park YH. Clinical implications of HER2 mRNA expression and intrinsic subtype in refractory HER2-positive metastatic breast cancer treated with pan-HER inhibitor, poziotinib. Breast Cancer Res Treat 2020; 184:743-753. [PMID: 32860168 DOI: 10.1007/s10549-020-05891-0] [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: 07/14/2020] [Accepted: 08/17/2020] [Indexed: 11/28/2022]
Abstract
INTRODUCTION We explored clinical implication of intrinsic molecular subtype in human epidermal growth factor receptor 2 (HER2) + metastatic breast cancer (BC) with pan-HER inhibitor from a phase II clinical trial of poziotinib in refractory HER2+BC patients. METHODS For this translational research correlated with phase II clinical trial, we performed an nCounter expression assay, using gene panel including 50 genes for PAM50 prediction and targeted deep sequencing. RESULTS From 106 participants, we obtained 97 tumor tissues and analyzed gene expression in 91 of these samples. Of 91 HER2+BCs, 40 (44.0%) were HER2-enriched (E) intrinsic molecular subtype, 17 (18.7%) of Luminal A, 16 (17.6%) of Basal-like, 14 (15.4%) of Luminal B and 4 (4.4%) of Normal-like. HER2-E subtype was associated with hormone receptor negativity (odds ratio [OR] 2.93; p = 0.019), 3 + of HER2 immunohistochemistry(IHC) (OR 5.64; p = 0.001), high mRNA expression of HER2 (OR 14.43; p = 0.001) and copy number(CN) amplification of HER2 (OR 12.80; p = 0.005). In genetic alterations, alteration was more frequently observed in HER2-E subtype (OR 3.84; p = 0.022) but there was no association between PIK3CA alteration and HER2-E subtype (p = 0.655). In terms of drug efficacy, high mRNA expression of HER2 was the most powerful predictor of poziotinib response (median progression-free survival [PFS): 4.63 months [high] vs. 2.56 [low]; p < .001). In a combination prediction model, median PFS of intrinsic subtypes except Her2-E with high HER2 mRNA expression without PIK3CA genetic alteration was 6.83 months and that of the remaining group was 1.74 months (p < .001). CONCLUSION HER2-E subtype was associated with hormone receptor status, HER2 IHC, CN and mRNA expression and TP53 mutation. In survival analysis, the information of level of HER2 mRNA expression, intrinsic molecular subtype and PI3K pathway alteration would be independent predictors to poziotinib treatment. ClinicalTrials.gov identifier: NCT02418689.
Collapse
Affiliation(s)
- Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea
| | - Kyunghee Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Seock-Ah Im
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul National University School of Medicine, Seoul, 03080, Korea
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Joohyuk Sohn
- Department of Internal Medicine, Yonsei Cancer Center, Seoul, 03722, Korea
| | - Keun Seok Lee
- Center for Breast Cancer, National Cancer Center Hospital, Goyang, 10408, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620, Korea
| | - Yaewon Yang
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, 28644, Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea.
| |
Collapse
|
31
|
Han B, Zhang H, Zhu Y, Han X, Wang Z, Gao Z, Yuan Y, Tian R, Zhang F, Niu R. Subtype-specific risk models for accurately predicting the prognosis of breast cancer using differentially expressed autophagy-related genes. Aging (Albany NY) 2020; 12:13318-13337. [PMID: 32649310 PMCID: PMC7377895 DOI: 10.18632/aging.103437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/25/2020] [Indexed: 01/08/2023]
Abstract
Emerging evidence suggests that the dysregulation of autophagy-related genes (ARGs) is coupled with the carcinogenesis and progression of breast cancer (BRCA). We constructed three subtype-specific risk models using differentially expressed ARGs. In Luminal, Her-2, and Basal-like BRCA, four- (BIRC5, PARP1, ATG9B, and TP63), three- (ITPR1, CCL2, and GAPDH), and five-gene (PRKN, FOS, BAX, IFNG, and EIF4EBP1) risk models were identified, which all have a receiver operating characteristic > 0.65 in the training and testing dataset. Multivariable Cox analysis showed that those risk models can accurately and independently predict the overall survival of BRCA patients. Comprehensive analysis showed that the 12 identified ARGs were correlated with the overall survival of BRCA patients; six of the ARGs (PARP1, TP63, CCL2, GAPDH, FOS, and EIF4EBP1) were differentially expressed between BRCA and normal breast tissue at the protein level. In addition, the 12 identified ARGs were highly interconnected and displayed high frequency of copy number variation in BRCA samples. Gene set enrichment analysis suggested that the deactivation of the immune system was the important driving force for the progression of Basal-like BRCA. This study demonstrated that the 12 ARG signatures were potential multi-dimensional biomarkers for the diagnosis, prognosis, and treatment of BRCA.
Collapse
Affiliation(s)
- Baoai Han
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - He Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Yuying Zhu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Xingxing Han
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Zhiyong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Zicong Gao
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Yue Yuan
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Ruinan Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Fei Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| | - Ruifang Niu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin 300060, China
| |
Collapse
|
32
|
Yu J, Wu J, Huang O, He J, Li Z, Chen W, Li Y, Chen X, Shen K. Do 21-Gene Recurrence Score Influence Chemotherapy Decisions in T1bN0 Breast Cancer Patients? Front Oncol 2020; 10:708. [PMID: 32477946 PMCID: PMC7236800 DOI: 10.3389/fonc.2020.00708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/15/2020] [Indexed: 11/28/2022] Open
Abstract
Purpose: Hormone receptor (HR)-positive breast cancer patients with tumor size ≤1.0 cm and negative node have favorable outcomes. The 21-gene Recurrence Score (RS) could predict response to chemotherapy for HR+ breast cancer, but its role in T1bN0 disease is challenging. Methods: T1bN0 breast cancer patients diagnosed between January 2014 and June 2019 with RS results were included and categorized as Low- (RS < 18), Intermediate- (RS 18–30), or High-risk (RS > 30) groups. Univariate and multivariate analysis were used to assess factors associated with RS distribution and chemotherapy recommendation. Chemotherapy decisions change and patient adherence after 21-gene RS testing were also evaluated. Results: Among 237 patients with T1bN0 tumors, proportions of Low-, Intermediate-, and High-risk RS were 19.8, 63.3, and 16.9%, respectively. Multivariate analysis found that ER expression (P = 0.011), PR expression (P < 0.001), and Ki-67 index (P = 0.001) were independently associated with RS distribution. Adjuvant chemotherapy was recommended for 31.6% of patients, which was more frequently given to patients with higher tumor grade [Odds ratio (OR) = 2.99 for grade II, OR = 59.19 for grade III, P = 0.006], lymph vascular invasion (OR = 8.22, P = 0.032), Luminal-B subtype (OR = 5.68, P < 0.001), and Intermediate-to High-risk RS (OR = 10.01 for Intermediate-risk, OR = 192.42 for High-risk, P < 0.001). Chemotherapy decision change was found in 18.6% of patients, mainly in those with Intermediate- to High-risk RS tumor with the majority from no-chemotherapy to chemotherapy. The treatment compliance rate after the 21-gene RS testing with MDT was 95.4%. Conclusion: RS category was related to ER, PR, and Ki-67 expression, which was recognized as an independent factor of chemotherapy recommendation in T1bN0 breast cancer. The 21-gene RS testing would lead to a chemotherapy decision change rate of 18.6% as well as a high treatment adherence, which can be applied in T1bN0 patients.
Collapse
Affiliation(s)
- Jing Yu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayi Wu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ou Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianrong He
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhu Li
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiguo Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafen Li
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Lei L, Wang XJ, Mo YY, Cheng SHC, Zhou Y. DGM-CM6: A New Model to Predict Distant Recurrence Risk in Operable Endocrine-Responsive Breast Cancer. Front Oncol 2020; 10:783. [PMID: 32528885 PMCID: PMC7263173 DOI: 10.3389/fonc.2020.00783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/22/2020] [Indexed: 01/10/2023] Open
Abstract
To investigate the prognostic value of DGM-CM6 (Distant Genetic Model-Clinical variable Model 6) for endocrine-responsive breast cancer (ERBC) patients, we analyzed 752 operable breast cancer patients treated in a Taiwan cancer center from 2005 to 2014. Among them, 490 ERBC patients (identified by the PAM50 or immunohistochemistry method) were classified by DGM-CM6 into low- and high-risk groups (cutoff <33 and ≥33, respectively). Significant differences were observed between the DGM-CM6 low- and high-risk groups for 10-year distant recurrence-free survival (DRFS) in both lymph node (LN)- (P < 0.05) and LN+ patients (P < 0.05). Multivariate analysis confirmed the independent strength of DGM-CM6 for the prediction of high- vs. low- risk groups for DRFS (P < 0.0001, HR: 6.76, 95% CI, 1.8–25.42) and overall survival (P = 0.01, HR: 6.06, 95% CI:1.55–23.47), respectively. In summary, DGM-CM6 may be used to classify low- and high-risk groups for 10-year distant recurrence in both LN- and LN+ ERBC patients in the Asian population. A large scale clinical trial is warranted.
Collapse
Affiliation(s)
- Lei Lei
- Department of Breast Medical Oncology, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, China.,Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Xiao-Jia Wang
- Department of Breast Medical Oncology, Chinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital), Hangzhou, China
| | - Yin-Yuan Mo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Skye Hung-Chun Cheng
- Department of Radiation Oncology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Yunyun Zhou
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
34
|
Treatment selection for patients with equivocal HER2 status and in luminal versus HER2-enriched disease. Breast 2020; 48 Suppl 1:S49-S52. [PMID: 31839160 DOI: 10.1016/s0960-9776(19)31123-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Equivocal HER2 status has been variably defined in the past, and its clinical implications have long been debated. In the 2018 focused update, ASCO/CAP guidelines recommended that tumours with double-equivocal (by immunohistochemistry and in situ hybridization assays) HER2 status should be considered HER2-negative due to the lack of evidence for any benefit of HER2-targeted therapy. The biology and the response to systemic therapies of tumours co-expressing HR and HER2 is quite complex. There is an extensive bi-directional cross-talk between these 2 pathways, that may result in both intrinsic and acquired resistance to endocrine agents, as well as in lower sensitivity to HER2-targeted therapies. In fact, neoadjuvant studies indicate that pCR rates are significantly lower in HER2-positive/ER-positive than ER-negative tumours, regardless the type of HER2 targeted treatment. The recent identification of different subtypes of HER2-positive breast cancer, according to the co-expression of HR and/or the molecular (intrinsic) subtyping, has prompted a renewed interest for clinical studies aimed at better tailoring the systemic therapy for these patients. A subgroup of them might not need chemotherapy if treated with dual HER2 blockade, and this option has been tested in a number of neo-adjuvant trials. In addition, triple targeting of HR, HER2, and CDK4/6 pathways simultaneously may be an effective treatment and overcome the drug resistance mechanisms that are typical of the disease. Finally, HER2-positive breast cancer may well benefit from immunotherapeutic interventions with anti-programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) agents.
Collapse
|
35
|
Yoon S, Won HS, Kang K, Qiu K, Park WJ, Ko YH. Hormone Receptor-Status Prediction in Breast Cancer Using Gene Expression Profiles and Their Macroscopic Landscape. Cancers (Basel) 2020; 12:cancers12051165. [PMID: 32380759 PMCID: PMC7281553 DOI: 10.3390/cancers12051165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/26/2020] [Accepted: 05/03/2020] [Indexed: 11/24/2022] Open
Abstract
The cost of next-generation sequencing technologies is rapidly declining, making RNA-seq-based gene expression profiling (GEP) an affordable technique for predicting receptor expression status and intrinsic subtypes in breast cancer patients. Based on the expression levels of co-expressed genes, GEP-based receptor-status prediction can classify clinical subtypes more accurately than can immunohistochemistry (IHC). Using data from The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA BRCA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets, we identified common predictor genes found in both datasets and performed receptor-status prediction based on these genes. By assessing the survival outcomes of patients classified using GEP- or IHC-based receptor status, we compared the prognostic value of the two methods. We found that GEP-based HR prediction provided higher concordance with the intrinsic subtypes and a stronger association with treatment outcomes than did IHC-based hormone receptor (HR) status. GEP-based prediction improved the identification of patients who could benefit from hormone therapy, even in patients with non-luminal breast cancer. We also confirmed that non-matching subgroup classification affected the survival of breast cancer patients and that this could be largely overcome by GEP-based receptor-status prediction. In conclusion, GEP-based prediction provides more reliable classification of HR status, improving therapeutic decision making for breast cancer patients.
Collapse
Affiliation(s)
- Seokhyun Yoon
- Department of Electronics Eng., College of Engineering, Dankook University, Yongin-si 16890, Korea; (S.Y.); (K.Q.)
| | - Hye Sung Won
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Keunsoo Kang
- Department of Microbiology, College of Natural Sciences, Dankook University, Cheonan-si 31116, Korea;
| | - Kexin Qiu
- Department of Electronics Eng., College of Engineering, Dankook University, Yongin-si 16890, Korea; (S.Y.); (K.Q.)
| | - Woong June Park
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan-si 31116, Korea;
| | - Yoon Ho Ko
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
36
|
Dual HER2 Blockade versus a Single Agent in Trastuzumab-Containing Regimens for HER2-Positive Early Breast Cancer: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. JOURNAL OF ONCOLOGY 2020; 2020:5169278. [PMID: 32256583 PMCID: PMC7102417 DOI: 10.1155/2020/5169278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/22/2020] [Accepted: 02/08/2020] [Indexed: 01/19/2023]
Abstract
Purpose Although trastuzumab is the standard of care for patients with human epidermal growth factor receptor 2 (HER2)- positive early breast cancer (EBC), drug resistance and disease relapse occur. Therefore, we performed a meta-analysis to assess the efficacy and safety of trastuzumab-containing dual anti-HER2 therapy compared to trastuzumab alone. Methods A systematic search was performed to identify eligible randomized controlled trials (RCTs). Main outcomes including event-free survival/invasive disease-free survival (EFS/iDFS), overall survival (OS), and safety were considered. Results Ten RCTs were included (15,284 patients). Significant improvements were observed in both EFS/iDFS (HR 0.86, p=0.0003) and OS (HR 0.86, p=0.02) with trastuzumab-based dual anti-HER2 therapy, especially in adjuvant treatment, while in the neoadjuvant setting, dual-targeted therapy also achieved a substantial pathological complete response (pCR) benefit (HR 1.34, p=0.0002). Subgroup analysis revealed that the EFS/iDFS benefit was slightly higher with trastuzumab plus pertuzumab or plus neratinib than trastuzumab plus lapatinib, while OS benefit was significant with trastuzumab plus lapatinib, but there were no subgroup differences (interaction test, p=0.80 and 0.24, resp.). In addition, EFS/iDFS benefit was unrelated to hormone receptor status but pronounced in the lymph node-positive (LN+) subgroup, which should be interpreted cautiously for lacking interaction (p=0.18). Besides, patients receiving dual therapy, especially with the lapatinib-containing regimen, experienced more toxicity, but no increase in cardiotoxicity. Conclusions Despite being associated with more toxicity, trastuzumab-containing dual anti-HER2 therapy is superior to trastuzumab single agent for HER2-positive EBC independent of hormone receptor status. The correlation between survival and LN status needs further verification. Trastuzumab plus pertuzumab or plus neratinib is the preferred regimen with substantial efficacy and lower toxicity.
Collapse
|
37
|
Morganti S, Curigliano G. Moving beyond endocrine therapy for luminal metastatic breast cancer in the precision medicine era: looking for new targets. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1720508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Stefania Morganti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
38
|
Klæstad E, Opdahl S, Engstrøm MJ, Ytterhus B, Wik E, Bofin AM, Valla M. MRPS23 amplification and gene expression in breast cancer; association with proliferation and the non-basal subtypes. Breast Cancer Res Treat 2020; 180:73-86. [PMID: 31950385 PMCID: PMC7031208 DOI: 10.1007/s10549-020-05532-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 01/09/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE MRPS23 is recognized as a driver of proliferation in luminal breast cancer. The aims of the present study were to describe MRPS23 copy number change in breast cancer, and to assess associations between MRPS23 copy number change and molecular subtype, proliferation and prognosis, and between MRPS23 gene expression and molecular subtype and prognosis. METHODS Using fluorescence in situ hybridization (FISH), we examined MRPS23 and centromere 17 copy number in 590 formalin-fixed, paraffin-embedded primary tumours and 144 corresponding lymph node metastases from a cohort of Norwegian breast cancer patients. Furthermore, we analysed MRPS23 gene expression data in 1971 primary breast cancer tumours from the METABRIC dataset. We used Pearson's χ2 test to assess associations between MRPS23 copy number and molecular subtype and proliferation, and between MRPS23 expression and molecular subtype. We studied prognosis by estimating hazard ratios and cumulative incidence of death from breast cancer according to MRPS23 copy number and MRPS23 expression status. RESULTS We found MRPS23 amplification (mean MRPS23 copy number ≥ 6 and/or MRPS23/chromosome 17 ratio ≥ 2) in 8% of primary tumours. Copy number increase associated with non-basal subtypes and higher tumour cell proliferation (Ki67). Higher MRPS23 expression associated with the Luminal B subtype. We found no significant association between MRPS23 amplification or MRSP23 gene expression, and prognosis. CONCLUSION Amplification of MRPS23 is associated with higher proliferation and non-basal subtypes in breast cancer. High MRPS23 expression is associated with the Luminal B subtype.
Collapse
Affiliation(s)
- Elise Klæstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gate, 7030, Trondheim, Norway.
| | - Signe Opdahl
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Monica Jernberg Engstrøm
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gate, 7030, Trondheim, Norway.,Department of Breast and Endocrine Surgery, St. Olav's Hospital, Trondheim University Hospital, 7006, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gate, 7030, Trondheim, Norway
| | - Elisabeth Wik
- Department of Clinical Medicine, Section for Pathology, Centre for Cancer Biomarkers CCBIO, University of Bergen, 5021, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Anna Mary Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gate, 7030, Trondheim, Norway
| | - Marit Valla
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Erling Skjalgssons gate, 7030, Trondheim, Norway.,Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, 7006, Trondheim, Norway
| |
Collapse
|
39
|
Zhao S, Liu XY, Jin X, Ma D, Xiao Y, Shao ZM, Jiang YZ. Molecular portraits and trastuzumab responsiveness of estrogen receptor-positive, progesterone receptor-positive, and HER2-positive breast cancer. Am J Cancer Res 2019; 9:4935-4945. [PMID: 31410192 PMCID: PMC6691389 DOI: 10.7150/thno.35730] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/08/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Estrogen receptor-positive, progesterone receptor-positive, and HER2-positive breast cancers (triple-positive breast cancers, TPBCs) account for 5% to 10% of all breast cancers. The clinical and molecular features of TPBCs remain elusive. In this study, we aim to analyze the multiomics landscape and responsiveness of TPBCs to trastuzumab. Methods: We employed five cohorts. The first cohort was from the Surveillance, Epidemiology, and End Results database (n=32,056) and was used to determine the clinical characteristics of TPBC. The second, third and fourth cohorts were from The Cancer Genome Atlas (n=162), GSE2603 (n=37) and GSE2109 (n=30) datasets, respectively, and were used to examine the genomic features and molecular classification of TPBC. The fifth cohort comprised TPBC patients treated at Fudan University Shanghai Cancer Center (FUSCC, n=171) and was used to investigate an immunohistochemistry-defined luminal A-like subgroup of TPBC. Results: Patients with TPBC had a significantly better prognosis than those with ER-PR-HER2+ breast cancer. Genomic analysis revealed that TPBCs showed a lower TP53 mutation rate (30% vs. 69%, P < 0.001) and lower levels of HER2 mRNA and protein expression than ER-PR-HER2+ breast cancers. More than 40% of TPBCs were classified as the luminal A intrinsic subtype, with an even lower HER2 expression level. Based on the immunohistochemical detection of CDCA8, BCL2 and STC2, we identified a luminal A-like subgroup of TPBCs in the FUSCC cohort (CDCA8-negative, BCL2- and/or STC2-positive). Patients with luminal A-like TPBC had a better prognosis and benefited less from trastuzumab than those with TPBC of other subtypes. Conclusions: TPBCs consist of clinically and genomically heterogeneous subgroups that may require different therapeutic strategies. The luminal A-like subgroup of TPBCs is associated with a better prognosis and reduced benefit from trastuzumab.
Collapse
|