1
|
Fang K, Ohihoin AG, Liu T, Choppavarapu L, Nosirov B, Wang Q, Yu XZ, Kamaraju S, Leone G, Jin VX. Integrated single-cell analysis reveals distinct epigenetic-regulated cancer cell states and a heterogeneity-guided core signature in tamoxifen-resistant breast cancer. Genome Med 2024; 16:134. [PMID: 39558215 PMCID: PMC11572372 DOI: 10.1186/s13073-024-01407-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Inter- and intra-tumor heterogeneity is considered a significant factor contributing to the development of endocrine resistance in breast cancer. Recent advances in single-cell RNA sequencing (scRNA-seq) and single-cell ATAC sequencing (scATAC-seq) allow us to explore inter- and intra-tumor heterogeneity at single-cell resolution. However, such integrated single-cell analysis has not yet been demonstrated to characterize the transcriptome and chromatin accessibility in breast cancer endocrine resistance. METHODS In this study, we conducted an integrated analysis combining scRNA-seq and scATAC-seq on more than 80,000 breast tissue cells from two normal tissues (NTs), three primary tumors (PTs), and three tamoxifen-treated recurrent tumors (RTs). A variety of cell types among breast tumor tissues were identified, PT- and RT-specific cancer cell states (CSs) were defined, and a heterogeneity-guided core signature (HCS) was derived through such integrated analysis. Functional experiments were performed to validate the oncogenic role of BMP7, a key gene within the core signature. RESULTS We observed a striking level of cell-to-cell heterogeneity among six tumor tissues and delineated the primary to recurrent tumor progression, underscoring the significance of these single-cell level tumor cell clusters classified from scRNA-seq data. We defined nine CSs, including five PT-specific, three RT-specific, and one PT-RT-shared CSs, and identified distinct open chromatin regions of CSs, as well as a HCS of 137 genes. In addition, we predicted specific transcription factors (TFs) associated with the core signature and novel biological/metabolism pathways that mediate the communications between CSs and the tumor microenvironment (TME). We finally demonstrated that BMP7 plays an oncogenic role in tamoxifen-resistant breast cancer cells through modulating MAPK signaling pathways. CONCLUSIONS Our integrated single-cell analysis provides a comprehensive understanding of the tumor heterogeneity in tamoxifen resistance. We envision this integrated single-cell epigenomic and transcriptomic measure will become a powerful approach to unravel how epigenetic factors and the tumor microenvironment govern the development of tumor heterogeneity and to uncover potential therapeutic targets that circumvent heterogeneity-related failures.
Collapse
Affiliation(s)
- Kun Fang
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Aigbe G Ohihoin
- Cell and Developmental Biology PhD Program, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Tianxiang Liu
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Lavanya Choppavarapu
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Bakhtiyor Nosirov
- Department of Cancer Research, Luxembourg Institute of Health, NORLUX Neuro-Oncology Laboratory and Multiomics Data Science Research Group, Strassen, L-1445, Luxembourg
| | - Qianben Wang
- Department of Pathology and Duke Cancer Institute, Duke University, Durham, NC, 27710, USA
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sailaja Kamaraju
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Gustavo Leone
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Victor X Jin
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
2
|
Zhang X, Kong H, Liu X, Li Q, Fang X, Wang J, Qin Z, Hu N, Tian J, Cui H, Zhang L. Nomograms for predicting recurrence of HER2-positive breast cancer with different HR status based on ultrasound and clinicopathological characteristics. Cancer Med 2024; 13:e70146. [PMID: 39248049 PMCID: PMC11381954 DOI: 10.1002/cam4.70146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
PURPOSE This study aimed to identify ultrasound and clinicopathological characteristics related to recurrence in HER2-positive (HER2+) breast cancer, and to develop nomograms for predicting recurrence. METHODS In this dual-center study, we retrospectively enrolled 570 patients with HER2+ breast cancer. The ultrasound and clinicopathological characteristics of hormone receptor (HR)-/HER2+ patients and HR+/HER2+ patients were analyzed separately according to HR status. Eighty percent of the original samples from HR-/HER2+ and HR+/HER2+ patients were extracted by bootstrap sampling as the training cohorts, while the remaining 20% were used as the external validation cohorts. Informative characteristics were screened through univariate and multivariable Cox regression in the training cohorts and used to develop nomograms for predicting recurrence. The predictive accuracy was calculated using Harrell's C-index and calibration curves. RESULTS Three informative characteristics (axillary nodal status, calcification, and Adler degree) were identified in HR-/HER2+ patients, and another three (histological grade, axillary nodal status, and echogenic halo) in HR+/HER2+ patients. Based on these, two separate nomograms were constructed to assess recurrence risk. In the training cohorts, the C-index was 0.740 (95% CI: 0.667-0.811) for HR-/HER2+ nomogram, and 0.749 (95% CI: 0.679-0.820) for HR+/HER2+ nomogram. In the validation cohorts, the C-index was 0.708 (95% CI: 0.540-0.877) for HR-/HER2+ group, and 0.705 (95% CI: 0.557-0.853) for HR+/HER2+ group. The calibration curves also indicated the excellent accuracy of the nomograms. CONCLUSIONS Ultrasound performance of HER2+ breast cancers with different HR status was significantly different. Nomograms integrating ultrasound and clinicopathological characteristics exhibited favorable performance and have the potential to serve as a reliable method for predicting recurrence in heterogeneous breast cancer.
Collapse
Affiliation(s)
- Xudong Zhang
- Department of Abdominal Ultrasound, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hanqing Kong
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoxue Liu
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingxiang Li
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinran Fang
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junjia Wang
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zihao Qin
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Nana Hu
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiawei Tian
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Ultrasound molecular imaging Joint laboratory of Heilongjiang province (International Cooperation), Harbin, Heilongjiang, China
| | - Hao Cui
- Department of Ultrasound Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Ultrasound molecular imaging Joint laboratory of Heilongjiang province (International Cooperation), Harbin, Heilongjiang, China
| | - Lei Zhang
- Department of Abdominal Ultrasound, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Ultrasound molecular imaging Joint laboratory of Heilongjiang province (International Cooperation), Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Woodcock CL, Alsaleem M, Toss MS, Lothion-Roy J, Harris AE, Jeyapalan JN, Blatt N, Rizvanov AA, Miftakhova RR, Kariri YA, Madhusudan S, Green AR, Rutland CS, Fray RG, Rakha EA, Mongan NP. The role of the ALKBH5 RNA demethylase in invasive breast cancer. Discov Oncol 2024; 15:343. [PMID: 39127986 PMCID: PMC11317455 DOI: 10.1007/s12672-024-01205-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) is the most common internal RNA modification and is involved in regulation of RNA and protein expression. AlkB family member 5 (ALKBH5) is a m6A demethylase. Given the important role of m6A in biological mechanisms, m6A and its regulators, have been implicated in many disease processes, including cancer. However, the contribution of ALKBH5 to invasive breast cancer (BC) remains poorly understood. The aim of this study was to evaluate the clinicopathological value of ALKBH5 in BC. METHODS Publicly available data were used to investigate ALKBH5 mRNA alterations, prognostic significance, and association with clinical parameters at the genomic and transcriptomic level. Differentially expressed genes (DEGs) and enriched pathways with low or high ALKBH5 expression were investigated. Immunohistochemistry (IHC) was used to assess ALKBH5 protein expression in a large well-characterised BC series (n = 1327) to determine the clinical significance and association of ALKBH5 expression. RESULTS Reduced ALKBH5 mRNA expression was significantly associated with poor prognosis and unfavourable clinical parameters. ALKBH5 gene harboured few mutations and/or copy number alternations, but low ALKBH5 mRNA expression was seen. Patients with low ALKBH5 mRNA expression had a number of differentially expressed genes and enriched pathways, including the cytokine-cytokine receptor interaction pathway. Low ALKBH5 protein expression was significantly associated with unfavourable clinical parameters associated with tumour progression including larger tumour size and worse Nottingham Prognostic Index group. CONCLUSION This study implicates ALKBH5 in BC and highlights the need for further functional studies to decipher the role of ALKBH5 and RNA m6A methylation in BC progression.
Collapse
Affiliation(s)
- Corinne L Woodcock
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK.
| | - Mansour Alsaleem
- Nottingham Breast Cancer Research Centre, School of Medicine, Academic Unit for Translational Medical Sciences, University of Nottingham, Nottingham, UK
- Unit of Scientific Research, Applied College, Qassim University, Qassim, Saudi Arabia
| | - Michael S Toss
- Nottingham Breast Cancer Research Centre, School of Medicine, Academic Unit for Translational Medical Sciences, University of Nottingham, Nottingham, UK
| | - Jennifer Lothion-Roy
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Anna E Harris
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Jennie N Jeyapalan
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Nataliya Blatt
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
- Institute for Fundamental Medicine and Science, Kazan Federal University, Kazan, Tatarstan, Russia
| | - Albert A Rizvanov
- Institute for Fundamental Medicine and Science, Kazan Federal University, Kazan, Tatarstan, Russia
| | - Regina R Miftakhova
- Institute for Fundamental Medicine and Science, Kazan Federal University, Kazan, Tatarstan, Russia
| | - Yousif A Kariri
- Nottingham Breast Cancer Research Centre, School of Medicine, Academic Unit for Translational Medical Sciences, University of Nottingham, Nottingham, UK
- Department of Clinical Laboratory Science, Faculty of Applied Medical Science, Shaqra University 33, 11961, Shaqra, Saudi Arabia
| | - Srinivasan Madhusudan
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, School of Medicine, Academic Unit for Translational Medical Sciences, University of Nottingham, Nottingham, UK
| | - Catrin S Rutland
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Rupert G Fray
- School of Biosciences, Plant Science Division, University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Nottingham Breast Cancer Research Centre, School of Medicine, Academic Unit for Translational Medical Sciences, University of Nottingham, Nottingham, UK
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
- Pathology Department, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Nigel P Mongan
- University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK.
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK.
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Al-Ruwishan A, Amer B, Salem A, Abdi A, Chimpandu N, Esa A, Melemenis A, Saleem MZ, Mathew R, Gamallat Y. Advancements in Understanding the Hide-and-Seek Strategy of Hibernating Breast Cancer Cells and Their Implications in Oncology from a Broader Perspective: A Comprehensive Overview. Curr Issues Mol Biol 2024; 46:8340-8367. [PMID: 39194709 DOI: 10.3390/cimb46080492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Despite recent advancements in technology, breast cancer still poses a significant threat, often resulting in fatal consequences. While early detection and treatments have shown some promise, many breast cancer patients continue to struggle with the persistent fear of the disease returning. This fear is valid, as breast cancer cells can lay dormant for years before remerging, evading traditional treatments like a game of hide and seek. The biology of these dormant breast cancer cells presents a crucial yet poorly understood challenge in clinical settings. In this review, we aim to explore the mysterious world of dormant breast cancer cells and their significant impact on patient outcomes and prognosis. We shed light on the elusive role of the G9a enzyme and many other epigenetic factors in breast cancer recurrence, highlighting its potential as a target for eliminating dormant cancer cells and preventing disease relapse. Through this comprehensive review, we not only emphasise the urgency of unravelling the dynamics of dormant breast cancer cells to improve patient outcomes and advance personalised oncology but also provide a guide for fellow researchers. By clearly outlining the clinical and research gaps surrounding dormant breast cancer cells from a molecular perspective, we aim to inspire further exploration of this critical area, ultimately leading to improved patient care and treatment strategies.
Collapse
Affiliation(s)
- Aiman Al-Ruwishan
- Space for Research Initiative, Research Horizons, London NW10 2PU, UK
| | - Bushra Amer
- Department of Family Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Ahmed Salem
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 53210 Pardubice, Czech Republic
| | - Ahmed Abdi
- Independent Researcher, Uxbridge UB9 6JH, UK
| | | | | | | | - Muhammad Zubair Saleem
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Roselit Mathew
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yaser Gamallat
- Department of Oncology, Biochemistry and Molecular Biology, and Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
5
|
Fu D, Weng X, Su Y, Hong B, Zhao A, Lin J. Establishing a model composed of immune-related gene-modules to predict tumor immunotherapy response. Sci Rep 2024; 14:16630. [PMID: 39025898 PMCID: PMC11258235 DOI: 10.1038/s41598-024-67742-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
At present, tumor immunotherapy has been widely applied to treat various cancers. However, the accuracy of predicting treatment efficacy has not yet achieved a significant breakthrough. This study aimed to construct a prediction model based on the modified WGCNA algorithm to precisely judge the anti-tumor immune response. First, we used a murine colon cancer model to screen corresponding DEGs according to different groups. GSEA was used to analyze the potential mechanisms of the immune-related DEGs (irDEGs) in each group. Subsequently, the intersection of the irDEGs in every group was acquired, and 7 gene-modules were mapped. Finally, 4 gene-modules including cogenes, antiPD-1 immu-genes, chemo immu-genes and comb immu-genes, were selected for subsequent study. Furthermore, a clinical dataset of gastric cancer patients receiving immunotherapy was enrolled, and the irDEGs were identified. A total of 34 vital irDEGs were obtained from the intersections of the vital irDEGs and the four gene-modules. Next, the vital irDEGs were analyzed by the modified WGCNA algorithm, and the correlation coefficients between the 4 gene-modules and the response status to immunotherapy were calculated. Thus, a prediction model based on correlation coefficients was built, and the corresponding model scores were acquired. The AUC calculated according to the model score was 0.727, which was non-inferior to that of the ESTIMATE score and the TIDE score. Meanwhile, the AUC calculated according to the classification of the model scores was 0.705, which was non-inferior to that of the ESTIMATE classification and the TIDE classification. The prediction accuracy of the model was validated in clinical datasets of other cancers.
Collapse
Affiliation(s)
- Deqiang Fu
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiaoyuan Weng
- Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Quanzhou Medical College, Quanzhou, China
| | - Yunxia Su
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Binhuang Hong
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Aiyue Zhao
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| | - Jianqing Lin
- Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
6
|
Shin E, Yoo TK, Kim J, Chung IY, Ko BS, Kim HJ, Lee JW, Son BH, Lee SB. Association of residual ductal carcinoma in situ with breast cancer treatment outcomes after neoadjuvant chemotherapy according to hormone receptor status. Discov Oncol 2024; 15:288. [PMID: 39017974 PMCID: PMC11254890 DOI: 10.1007/s12672-024-01157-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/15/2024] [Indexed: 07/18/2024] Open
Abstract
PURPOSE This research aimed to clarify the impact of residual ductal carcinoma in situ(DCIS) in surgical specimens obtained after neoadjuvant chemotherapy(NAC) for breast cancer on the associated prognosis outcomes. METHODS This retrospective study was performed on a cohort of 1,009 patients who achieved pCR following NAC for breast cancer and underwent subsequent breast surgery at a single institution between January 2008 and December 2019. Overall survival, local recurrence-free survival, distant metastasis-free survival, and disease-free survival of the residual and non-residual DCIS groups were the outcomes compared, with further subgroup analysis performed according to hormone receptor status. RESULTS 260 individuals (25.8%) presented with residual DCIS. Based on a median follow-up of 54.0 months, no significant differences in outcomes were observed between the two groups. Patients with residual DCIS and hormone receptor-negative (HR-) breast cancer demonstrated a significant decrease in distant metastasis-free survival (p = 0.030) compared to those without residual DCIS. In the HR + cohort, no significant difference was observed between the two groups. Multivariate analysis of the HR- cohort demonstrated a significant association between residual DCIS and an elevated risk for distant recurrence (hazard ratio = 2.3, 95% confidence interval = 1.01-5.20, p = 0.047). CONCLUSIONS Residual DCIS following NAC may impact breast cancer outcomes, particularly with respect to the occurrence of distant metastasis in HR- patients. Therefore, clinicians must vigilantly monitor patients with residual DCIS after NAC, and further research is needed to expand our understanding of the clinical implications of residual DCIS.
Collapse
Affiliation(s)
- Eunju Shin
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Tae-Kyung Yoo
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Jisun Kim
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Il Yong Chung
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Beom Seok Ko
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Jong Won Lee
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Byung Ho Son
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Sae Byul Lee
- Division of Breast Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Korea.
| |
Collapse
|
7
|
Han YJ, Liu S, Hardeman A, Rajagopal PS, Mueller J, Khramtsova G, Sanni A, Ajani M, Clayton W, Hurley IW, Yoshimatsu TF, Zheng Y, Parker J, Perou CM, Olopade OI. The VEGF-Hypoxia Signature Is Upregulated in Basal-like Breast Tumors from Women of African Ancestry and Associated with Poor Outcomes in Breast Cancer. Clin Cancer Res 2024; 30:2609-2618. [PMID: 38564595 DOI: 10.1158/1078-0432.ccr-23-1526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/21/2023] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Black women experience the highest breast cancer mortality rate compared with women of other racial/ethnic groups. To gain a deeper understanding of breast cancer heterogeneity across diverse populations, we examined a VEGF-hypoxia gene expression signature in breast tumors from women of diverse ancestry. EXPERIMENTAL DESIGN We developed a NanoString nCounter gene expression panel and applied it to breast tumors from Nigeria (n = 182) and the University of Chicago (Chicago, IL; n = 161). We also analyzed RNA sequencing data from Nigeria (n = 84) and The Cancer Genome Atlas (TCGA) datasets (n = 863). Patient prognosis was analyzed using multiple datasets. RESULTS The VEGF-hypoxia signature was highest in the basal-like subtype compared with other subtypes, with greater expression in Black women compared with White women. In TCGA dataset, necrotic breast tumors had higher scores for the VEGF-hypoxia signature compared with non-necrosis tumors (P < 0.001), with the highest proportion in the basal-like subtype. Furthermore, necrotic breast tumors have higher scores for the proliferation signature, suggesting an interaction between the VEGF-hypoxia signature, proliferation, and necrosis. T-cell gene expression signatures also correlated with the VEGF-hypoxia signature when testing all tumors in TCGA dataset. Finally, we found a significant association of the VEGF-hypoxia profile with poor outcomes when using all patients in the METABRIC (P < 0.0001) and SCAN-B datasets (P = 0.002). CONCLUSIONS These data provide further evidence for breast cancer heterogeneity across diverse populations and molecular subtypes. Interventions selectively targeting VEGF-hypoxia and the immune microenvironment have the potential to improve overall survival in aggressive breast cancers that disproportionately impact Black women in the African Diaspora.
Collapse
Affiliation(s)
- Yoo Jane Han
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Siyao Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Ashley Hardeman
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Padma Sheila Rajagopal
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Jeffrey Mueller
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Galina Khramtsova
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Ayodele Sanni
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Mustapha Ajani
- Department of Pathology, College of Medicine, University of Ibadan/University College Hospital, Ibadan, Oyo, Nigeria
| | - Wendy Clayton
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Ian W Hurley
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Toshio F Yoshimatsu
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Joel Parker
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Olufunmilayo I Olopade
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
8
|
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
|
9
|
Kang D, Wang C, Han Z, Zheng L, Guo W, Fu F, Qiu L, Han X, He J, Li L, Chen J. Exploration of the relationship between tumor-infiltrating lymphocyte score and histological grade in breast cancer. BMC Cancer 2024; 24:318. [PMID: 38454386 PMCID: PMC10921807 DOI: 10.1186/s12885-024-12069-0] [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: 09/21/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The histological grade is an important factor in the prognosis of invasive breast cancer and is vital to accurately identify the histological grade and reclassify of Grade2 status in breast cancer patients. METHODS In this study, data were collected from 556 invasive breast cancer patients, and then randomly divided into training cohort (n = 335) and validation cohort (n = 221). All patients were divided into actual low risk group (Grade1) and high risk group (Grade2/3) based on traditional histological grade, and tumor-infiltrating lymphocyte score (TILs-score) obtained from multiphoton images, and the TILs assessment method proposed by International Immuno-Oncology Biomarker Working Group (TILs-WG) were also used to differentiate between high risk group and low risk group of histological grade in patients with invasive breast cancer. Furthermore, TILs-score was used to reclassify Grade2 (G2) into G2 /Low risk and G2/High risk. The coefficients for each TILs in the training cohort were retrieved using ridge regression and TILs-score was created based on the coefficients of the three kinds of TILs. RESULTS Statistical analysis shows that TILs-score is significantly correlated with histological grade, and is an independent predictor of histological grade (odds ratio [OR], 2.548; 95%CI, 1.648-3.941; P < 0.0001), but TILs-WG is not an independent predictive factor for grade (P > 0.05 in the univariate analysis). Moreover, the risk of G2/High risk group is higher than that of G2/Low risk group, and the survival rate of patients with G2/Low risk is similar to that of Grade1, while the survival rate of patients with G2/High risk is even worse than that of patients with G3. CONCLUSION Our results suggest that TILs-score can be used to predict the histological grade of breast cancer and potentially to guide the therapeutic management of breast cancer patients.
Collapse
Affiliation(s)
- Deyong Kang
- Department of Pathology, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Chuan Wang
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Zhonghua Han
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China
| | - Wenhui Guo
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Fangmeng Fu
- Breast Surgery Ward, Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, P. R. China
| | - Lida Qiu
- College of Physics and Electronic Information Engineering, Minjiang University, 350108, Fuzhou, P. R. China
| | - Xiahui Han
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China
| | - Jiajia He
- School of Science, Jimei University, 361021, Xiamen, P. R. China.
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China.
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, 350007, Fuzhou, P. R. China.
| |
Collapse
|
10
|
De Blander H, Tonon L, Fauvet F, Pommier RM, Lamblot C, Benhassoun R, Angileri F, Gibert B, Rodriguez R, Ouzounova M, Morel AP, Puisieux A. Cooperative pro-tumorigenic adaptation to oncogenic RAS through epithelial-to-mesenchymal plasticity. SCIENCE ADVANCES 2024; 10:eadi1736. [PMID: 38354248 PMCID: PMC10866563 DOI: 10.1126/sciadv.adi1736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
In breast cancers, aberrant activation of the RAS/MAPK pathway is strongly associated with mesenchymal features and stemness traits, suggesting an interplay between this mitogenic signaling pathway and epithelial-to-mesenchymal plasticity (EMP). By using inducible models of human mammary epithelial cells, we demonstrate herein that the oncogenic activation of RAS promotes ZEB1-dependent EMP, which is necessary for malignant transformation. Notably, EMP is triggered by the secretion of pro-inflammatory cytokines from neighboring RAS-activated senescent cells, with a prominent role for IL-6 and IL-1α. Our data contrast with the common view of cellular senescence as a tumor-suppressive mechanism and EMP as a process promoting late stages of tumor progression in response to signals from the tumor microenvironment. We highlighted here a pro-tumorigenic cooperation of RAS-activated mammary epithelial cells, which leverages on oncogene-induced senescence and EMP to trigger cellular reprogramming and malignant transformation.
Collapse
Affiliation(s)
- Hadrien De Blander
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Laurie Tonon
- Synergie Lyon Cancer, Plateforme de Bioinformatique ‘Gilles Thomas’, Centre Léon Bérard, Lyon, France
| | - Frédérique Fauvet
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Roxane M. Pommier
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
- Synergie Lyon Cancer, Plateforme de Bioinformatique ‘Gilles Thomas’, Centre Léon Bérard, Lyon, France
| | - Christelle Lamblot
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Rahma Benhassoun
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Francesca Angileri
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Benjamin Gibert
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
- Gastroenterology and Technologies for Health Group, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS5286, Université Lyon 1, 69008, Lyon, France
| | - Raphaël Rodriguez
- Equipe Labellisée Ligue Contre le Cancer, CNRS UMR 3666, INSERM U1143, Paris, France
- Institut Curie, PSL Research University, Paris, France
| | - Maria Ouzounova
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Anne-Pierre Morel
- Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Equipe Labellisée Ligue Contre le Cancer, 69008, Lyon, France
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Alain Puisieux
- Equipe Labellisée Ligue Contre le Cancer, CNRS UMR 3666, INSERM U1143, Paris, France
- Institut Curie, PSL Research University, Paris, France
| |
Collapse
|
11
|
Azizi A, Mansouri N, Tarlan M, Sadeghi M. Analysis of Interleukin-6 Gene Variants ( rs1800795, rs1800796, rs1554606, rs1800797, rs2069840, rs12700386, and rs2069861) as Prognostic Markers in Breast Cancer: A Systematic Review, Meta-Analysis, and Network Analysis. J Interferon Cytokine Res 2024; 44:3-15. [PMID: 38029374 DOI: 10.1089/jir.2023.0090] [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] [Indexed: 12/01/2023] Open
Abstract
Interleukin-6 (IL-6) has obviously tumor-promoting and tumor-inhibitory effects and can induce an epithelial-mesenchymal transition phenotype in human breast cancer (BC) cells and implicate its potential to promote BC metastasis. Herein, we aimed to evaluate the association of IL-6 variants (rs1800795, rs1800796, rs1554606, rs1800797, rs2069840, rs12700386, and rs2069861) with the susceptibility to BC. The databases of PubMed/Medline, Web of Science, Scopus, and Cochrane Library were searched until December 19, 2022, without any restrictions. The quality assessment of each study was performed based on the Newcastle-Ottawa Scale tool. The Review Manager 5.3 software presented the effect sizes including odds ratio (OR) along with a 95% confidence interval (CI). Both publication bias and sensitivity analyses were carried out by the Comprehensive Meta-Analysis version 2.0 software. A total of 2,508 records were identified among databases and at last, 27 articles were entered into the meta-analysis. Seven polymorphisms of IL-6 were entered into the analyses. Just rs1800797 polymorphism in the dominant model (OR = 1.51; 95% CI = 1.15-2.00; P = 0.003) and rs2069840 polymorphism in heterozygous (OR = 0.89; 95% CI = 0.81-0.97; P = 0.008) and dominant (OR = 0.91; 95% CI = 0.84-0.99; P = 0.02) models had a significant association with the BC risk. In conclusion, among 7 polymorphisms and despite a few included cases, the present meta-analysis recommended that the AA+GA genotype of rs1800797 polymorphism had a significantly elevated risk and the GC and the CC+GC genotypes of rs2069840 polymorphism had a protective role in the BC patients.
Collapse
Affiliation(s)
- Ali Azizi
- Social Development and Health Promotion Research Center, Department of Family and Community Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasrin Mansouri
- Department of Obstetrics and Gynecology, Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mitra Tarlan
- Department of Physiology, Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Sadeghi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
12
|
Oumnia B, Maladho D, Amanda G, Wafaa K, Fadila G, Mohamed C, Najdi A, Abderrahmane ALB, Chakib N, Nabil I, Mohamed K. Immunohistochemical-Based Molecular Subtypes of Female Breast Cancer: A Retrospective Cross-Sectional Study at Cheikh Khalifa Hospital in Casablanca, Morocco. Cancer Control 2024; 31:10732748241300655. [PMID: 39528903 PMCID: PMC11555740 DOI: 10.1177/10732748241300655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 10/13/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Breast cancer is a major public health concern worldwide and the most prevalent form of cancer in Morocco. This study aimed to describe the histological and immunohistochemical profiles of breast cancer in women admitted to Cheikh Khalifa Hospital in Casablanca, Morocco. METHODS This is a retrospective cross-sectional study. All histologically confirmed female breast cancer cases diagnosed between January 2017 and May 2021 at the Cheikh Khalifa University Hospital were included in the study. Data were collected from patient electronic medical records. Using an electronic sheet, information was collected about the socio-demographic characteristics of the patients, clinical features, histopathology, molecular characteristics, treatment received, and progression. RESULTS Invasive carcinoma of no special type was the most common type of cancer accounting for 90.7% of all cases. The majority of the tumors (56.1%) were grade II tumors. About 42.1% of tumors were lymph node-positive and only 13.4% developed distant metastasis. Immunohistochemical data revealed that 57.9% of the tumors in this study were hormone receptor-positive (ER+ and PR+), 74.4% were estrogen receptor-positive (ER+), 58.5% were progesterone receptor positive (PR+), and 18.9% were HER2 positive (HER2+). The most common molecular subtype was Luminal A-like (43.9%). A statistically significant difference was found in histological grades across the four molecular subtypes (P < 0.001). CONCLUSIONS Our findings should be used to guide breast cancer management policies in Morocco. Larger cohort studies are needed to determine the specificity of the breast cancer profile in Morocco as well as the epidemiological risk factors specific to every subtype.
Collapse
Affiliation(s)
- Bouaddi Oumnia
- Mohammed VI International School of Public Health, Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat, Morocco
| | - Diaby Maladho
- Mohammed VI International School of Public Health, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Gordon Amanda
- University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Kaikani Wafaa
- Department of Oncology, Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Guessous Fadila
- Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Chahboune Mohamed
- Laboratory of Sciences and Health Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat, Morocco
| | - Adil Najdi
- Department of Epidemiology, Faculty of Medicine and Pharmacy, Tangier, Morocco
| | - AL Bouzidi Abderrahmane
- Laboratory of Pathological Anatomy, Cheikh Khalifa International University Hospital, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Nejjari Chakib
- Faculty of Medicine, Euromed Research Center, Euromed University of Fez (UEMF), Fez, Morocco
- Faculty of Medicine, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ismaili Nabil
- Department of Oncology, Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Khalis Mohamed
- Mohammed VI International School of Public Health, Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Higher Institute of Nursing Professions and Health Techniques, Rabat, Ministry of Health and Social Protection, Rabat, Morocco
| |
Collapse
|
13
|
Desterke C, Xiang Y, Elhage R, Duruel C, Chang Y, Hamaï A. Ferroptosis Inducers Upregulate PD-L1 in Recurrent Triple-Negative Breast Cancer. Cancers (Basel) 2023; 16:155. [PMID: 38201582 PMCID: PMC10778345 DOI: 10.3390/cancers16010155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
(1) Background: Triple-negative breast cancer (TNBC) is a distinct subgroup of breast cancer presenting a high level of recurrence, and neo-adjuvant chemotherapy is beneficial in its therapy management. Anti-PD-L1 immunotherapy improves the effect of neo-adjuvant therapy in TNBC. (2) Methods: Immune-modulation and ferroptosis-related R-packages were developed for integrative omics analyses under ferroptosis-inducer treatments: TNBC cells stimulated with ferroptosis inducers (GSE173905, GSE154425), single cell data (GSE191246) and mass spectrometry on breast cancer stem cells. Clinical association analyses were carried out with breast tumors (TCGA and METABRIC cohorts). Protein-level validation was investigated through protein atlas proteome experiments. (3) Results: Erastin/RSL3 ferroptosis inducers upregulate CD274 in TNBC cells (MDA-MB-231 and HCC38). In breast cancer, CD274 expression is associated with overall survival. Breast tumors presenting high expression of CD274 upregulated some ferroptosis drivers associated with prognosis: IDO1, IFNG and TNFAIP3. At the protein level, the induction of Cd274 and Tnfaip3 was confirmed in breast cancer stem cells under salinomycin treatment. In a 4T1 tumor treated with cyclophosphamide, the single cell expression of Cd274 was found to increase both in myeloid- and lymphoid-infiltrated cells, independently of its receptor Pdcd1. The CD274 ferroptosis-driver score computed on a breast tumor transcriptome stratified patients on their prognosis: low score was observed in the basal subgroup, with a higher level of recurrent risk scores (oncotypeDx, ggi and gene70 scores). In the METABRIC cohort, CD274, IDO1, IFNG and TNFAIP3 were found to be overexpressed in the TNBC subgroup. The CD274 ferroptosis-driver score was found to be associated with overall survival, independently of TNM classification and age diagnosis. The tumor expression of CD274, TNFAIP3, IFNG and IDO1, in a biopsy of breast ductal carcinoma, was confirmed at the protein level (4) Conclusions: Ferroptosis inducers upregulate PD-L1 in TNBC cells, known to be an effective target of immunotherapy in high-risk early TNBC patients who received neo-adjuvant therapy. Basal and TNBC tumors highly expressed CD274 and ferroptosis drivers: IFNG, TNFAIP3 and IDO1. The CD274 ferroptosis-driver score is associated with prognosis and to the risk of recurrence in breast cancer. A potential synergy of ferroptosis inducers with anti-PD-L1 immunotherapy is suggested for recurrent TNBC.
Collapse
Affiliation(s)
- Christophe Desterke
- UFR Médecine-INSERM UMRS1310, Université Paris-Saclay, F-94800 Villejuif, France
| | - Yao Xiang
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France; (Y.X.); (R.E.); (C.D.); (Y.C.)
| | - Rima Elhage
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France; (Y.X.); (R.E.); (C.D.); (Y.C.)
- Team 5/Ferostem Group, F-75015 Paris, France
| | - Clémence Duruel
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France; (Y.X.); (R.E.); (C.D.); (Y.C.)
- Team 5/Ferostem Group, F-75015 Paris, France
| | - Yunhua Chang
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France; (Y.X.); (R.E.); (C.D.); (Y.C.)
| | - Ahmed Hamaï
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France; (Y.X.); (R.E.); (C.D.); (Y.C.)
- Team 5/Ferostem Group, F-75015 Paris, France
| |
Collapse
|
14
|
Kim H, Whitman AA, Wisniewska K, Kakati RT, Garcia-Recio S, Calhoun BC, Franco HL, Perou CM, Spanheimer PM. Tamoxifen Response at Single-Cell Resolution in Estrogen Receptor-Positive Primary Human Breast Tumors. Clin Cancer Res 2023; 29:4894-4907. [PMID: 37747807 PMCID: PMC10690085 DOI: 10.1158/1078-0432.ccr-23-1248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/18/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
PURPOSE In estrogen receptor-positive (ER+)/HER2- breast cancer, multiple measures of intratumor heterogeneity are associated with a worse response to endocrine therapy. We sought to develop a novel experimental model to measure heterogeneity in response to tamoxifen treatment in primary breast tumors. EXPERIMENTAL DESIGN To investigate heterogeneity in response to treatment, we developed an operating room-to-laboratory pipeline for the collection of live normal breast specimens and human tumors immediately after surgical resection for processing into single-cell workflows for experimentation and genomic analyses. Live primary cell suspensions were treated ex vivo with tamoxifen (10 μmol/L) or control media for 12 hours, and single-cell RNA libraries were generated using the 10X Genomics droplet-based kit. RESULTS In total, we obtained and processed normal breast tissue from two women undergoing reduction mammoplasty and tumor tissue from 10 women with ER+/HER2- invasive breast carcinoma. We demonstrate differences in tamoxifen response by cell type and identify distinctly responsive and resistant subpopulations within the malignant cell compartment of human tumors. Tamoxifen resistance signatures from resistant subpopulations predict poor outcomes in two large cohorts of ER+ breast cancer patients and are enriched in endocrine therapy-resistant tumors. CONCLUSIONS This novel ex vivo model system now provides the foundation to define responsive and resistant subpopulations within heterogeneous human tumors, which can be used to develop precise single cell-based predictors of response to therapy and to identify genes and pathways driving therapeutic resistance.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Austin A. Whitman
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Rasha T. Kakati
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Benjamin C. Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hector L. Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
- Computational Medicine Program, University of North Carolina, Chapel Hill, North Carolina
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
- Computational Medicine Program, University of North Carolina, Chapel Hill, North Carolina
| | - Philip M. Spanheimer
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
15
|
Bhise K, Gavande NS, Iyer AK. Leveraging hypoxia in triple-negative breast cancer as a promising treatment strategy. Drug Discov Today 2023; 28:103761. [PMID: 37660983 DOI: 10.1016/j.drudis.2023.103761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/08/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Current treatment strategies for triple-negative breast cancer (TNBC) are based upon conventional chemotherapy, immunotherapy, or a combination of both. The treatment regimen for chemotherapy is often a combination of two or more drugs, either dose dense or low dose for synergy. Anthracyclines, alkylating agents, antimicrotubule agents, and antimetabolites for early-stage TNBC; and antimetabolites, non-taxane microtubule inhibitors, and cross-linker platinums for late-stage TNBC are usually administered in the clinical setting. Newer options for patients with advanced TNBC, such as poly (ADP-ribose) polymerase (PARP) inhibitors and immune checkpoint inhibitors, have recently emerged for cases where surgery is not a viable option and the disease has metastasized. This review outlines the current trends in hypoxia-inspired treatment strategies for TNBC with a focus on clinical trials.
Collapse
Affiliation(s)
- Ketki Bhise
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| | - Navnath S Gavande
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA; Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, USA
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA; Molecular Imaging Program, Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
16
|
Li Y, Chen T, Du F, Wang H, Ma L. Concordance of RT-qPCR with immunohistochemistry and its beneficial role in breast cancer subtyping. Medicine (Baltimore) 2023; 102:e35272. [PMID: 37746948 PMCID: PMC10519502 DOI: 10.1097/md.0000000000035272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/12/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
This study was to compare the concordance of transcription-quantitative polymerase chain reaction (RT-qPCR) with immunohistochemistry (IHC) in determining estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and tumor proliferation index (Ki67) status in breast cancer, and to assess the prognosis based on different subtypes. Totally 323 breast cancer patients were selected, including 216 in the training set and 107 in the validation set. Logistic regression models were constructed using 5-fold cross-validation with the mRNA expression of each biomarker as the predictor and the corresponding IHC expression level as the binary response variable. Receiver operating characteristic curve was used to determine the cutoff value. When the thresholds of ER, PR, HER2, and Ki67 were 0.764, 0.709, 0.161, and 0.554, there existed high concordance rates between IHC and RT-qPCR in ER (94.4%), PR (88.0%) and HER2 (89.4%) and a medium concordance rate in Ki67 (67.8%), which were further confirmed in the validation set (ER: 81.3%, PR: 78.3%, HER2: 80.4%, and Ki67: 69.1%). Based on the subtyping stratified by RT-qPCR, the 5-year recurrence-free interval rates of patients with luminal, HER2-enriched, and triple-negative subtypes were 88% (95% CI: 0.84-0.93), 82% (95% CI: 0.73-0.92) and 58% (95% CI: 0.42-0.80), respectively, which were similar to those assessed by IHC (88%, 78% and 47%). RT-qPCR may be a complementary method to IHC, which can not only provide additional useful information in clinic, but also show more advantages over IHC in determining certain subtypes of breast cancer.
Collapse
Affiliation(s)
- Yilun Li
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Furong Du
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics CO., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
| | - Huimin Wang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics CO., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
| | - Li Ma
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
17
|
Khanna P, Mehta R, Mehta GA, Bhatt V, Guo JY, Gatza ML. SOX4-SMARCA4 complex promotes glycolysis-dependent TNBC cell growth through transcriptional regulation of Hexokinase 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.557071. [PMID: 37745600 PMCID: PMC10515838 DOI: 10.1101/2023.09.10.557071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Tumor cells rely on increased glycolytic capacity to promote cell growth and progression. While glycolysis is known to be upregulated in the majority of triple negative (TNBC) or basal-like subtype breast cancers, the mechanism remains unclear. Here, we used integrative genomic analyses to identify a subset of basal-like tumors characterized by increased expression of the oncogenic transcription factor SOX4 and its co-factor the SWI/SNF ATPase SMARCA4. These tumors are defined by unique gene expression programs that correspond with increased tumor proliferation and activation of key metabolic pathways, including glycolysis. Mechanistically, we demonstrate that the SOX4-SMARCA4 complex mediates glycolysis through direct transcriptional regulation of Hexokinase 2 (HK2) and that aberrant HK2 expression and altered glycolytic capacity are required to mediate SOX4-SMARCA4-dependent cell growth. Collectively, we have defined the SOX4-SMARCA4-HK2 signaling axis in basal-like breast tumors and established that this axis promotes metabolic reprogramming which is required to maintain tumor cell growth.
Collapse
Affiliation(s)
- Pooja Khanna
- Department of Radiation Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Rushabh Mehta
- Department of Radiation Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Gaurav A. Mehta
- Department of Radiation Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Vrushank Bhatt
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
- Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Jessie Y. Guo
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
- Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Michael L. Gatza
- Department of Radiation Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| |
Collapse
|
18
|
Kang B, Lee J, Jung JH, Kim WW, Keum H, Park HY. Differences in clinical outcomes between HER2-negative and HER2-positive luminal B breast cancer. Medicine (Baltimore) 2023; 102:e34772. [PMID: 37653831 PMCID: PMC10470803 DOI: 10.1097/md.0000000000034772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 09/02/2023] Open
Abstract
The clinical features and prognosis of breast cancer can vary widely, depending on the molecular subtype. Luminal B breast cancers are usually either estrogen receptor-positive and/or progesterone receptor-positive with high proliferation of Ki67 index, or HER2 positive (HER2+). The authors compared the clinicopathologic factors and survival rates of different subtypes of luminal B breast cancer according to HER2 status. Between 2009 and 2013, 1131 cases of breast cancer were reviewed and characterized as 1 of 4 different molecular subtypes based on their immunohistochemical results: luminal A, luminal B, HER2+, or triple-negative breast cancer. From these, luminal B breast cancers were extracted and the clinical features and prognosis of the HER2- and the HER2 + subtypes were compared. Survival differed significantly based on the molecular subtype regardless of whether or not the patient received treatment with neoadjuvant chemotherapy. While patients with HER2- luminal B breast cancer who received neoadjuvant chemotherapy had better prognoses, patients with HER2 + luminal B breast cancer who did not receive neoadjuvant chemotherapy had better prognoses. Luminal B breast cancers showed different clinical outcomes and survival rates according to HER2 gene overexpression type. Physicians should consider these results when they establish a treatment strategy.
Collapse
Affiliation(s)
- Byeongju Kang
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Jeeyeon Lee
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Jin Hyang Jung
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Wan Wook Kim
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Heejung Keum
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ho Yong Park
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| |
Collapse
|
19
|
Chiodoni C, Sangaletti S, Lecchi M, Ciniselli CM, Cancila V, Tripodi I, Ratti C, Talarico G, Brich S, De Cecco L, Baili P, Truffi M, Sottotetti F, Piccotti F, Tripodo C, Pruneri G, Triulzi T, Corsi F, Cappelletti V, Di Cosimo S, Verderio P, Colombo MP. A three-gene signature marks the time to locoregional recurrence in luminal-like breast cancer. ESMO Open 2023; 8:101590. [PMID: 37393630 PMCID: PMC10485389 DOI: 10.1016/j.esmoop.2023.101590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Gene expression profiling (GEP)-based prognostic signatures are being rapidly integrated into clinical decision making for systemic management of breast cancer patients. However, GEP remains relatively underdeveloped for locoregional risk assessment. Yet, locoregional recurrence (LRR), especially early after surgery, is associated with poor survival. PATIENTS AND METHODS GEP was carried out on two independent luminal-like breast cancer cohorts of patients developing early (≤5 years after surgery) or late (>5 years) LRR and used, by a training and testing approach, to build a gene signature able to intercept women at risk of developing early LRR. The GEP data of two in silico datasets and of a third independent cohort were used to explore its prognostic value. RESULTS Analysis of the first two cohorts led to the identification of three genes, CSTB, CCDC91 and ITGB1, whose expression, derived by principal component analysis, generated a three-gene signature significantly associated with early LRR in both cohorts (P value <0.001 and 0.005, respectively), overcoming the discriminatory capability of age, hormone receptor status and therapy. Remarkably, the integration of the signature with these clinical variables led to an area under the curve of 0.878 [95% confidence interval (CI) 0.810-0.945]. In in silico datasets we found that the three-gene signature retained its association, showing higher values in the early relapsed patients. Moreover, in the third additional cohort, the signature significantly associated with relapse-free survival (hazard ratio 1.56, 95% CI 1.04-2.35). CONCLUSIONS Our three-gene signature represents a new exploitable tool to aid treatment choice in patients with luminal-like breast cancer at risk of developing early recurrence.
Collapse
Affiliation(s)
- C Chiodoni
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - S Sangaletti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - M Lecchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Epidemiology and Data Science, Unit of Bioinformatics and Biostatistics, Milan
| | - C M Ciniselli
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Epidemiology and Data Science, Unit of Bioinformatics and Biostatistics, Milan
| | - V Cancila
- University of Palermo School of Medicine, Department of Health Sciences, Tumor Immunology Unit, Palermo
| | - I Tripodi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - C Ratti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - G Talarico
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - S Brich
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Pathology, Milan
| | - L De Cecco
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Mechanisms Unit, Milan
| | - P Baili
- Fondazione IRCCS Istituto Nazionale dei Tumori, Analytical Epidemiology and Health Impact Unit, Milan
| | - M Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Laboratory of Nanomedicine, Pavia
| | - F Sottotetti
- Istituti Clinici Scientifici Maugeri IRCCS, Medical Oncology Unit, Pavia
| | - F Piccotti
- Istituti Clinici Scientifici Maugeri IRCCS, Laboratory of Nanomedicine, Pavia
| | - C Tripodo
- University of Palermo School of Medicine, Department of Health Sciences, Tumor Immunology Unit, Palermo; FIRC Institute of Molecular Oncology (IFOM), Milan
| | - G Pruneri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Pathology, Milan
| | - T Triulzi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Targeting Unit, Milan
| | - F Corsi
- Istituti Clinici Scientifici Maugeri IRCCS, Surgery Department, Breast Unit, Pavia; Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan
| | - V Cappelletti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Advanced Diagnostics, Biomarkers Unit, Milan, Italy
| | - S Di Cosimo
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Advanced Diagnostics, Biomarkers Unit, Milan, Italy
| | - P Verderio
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Epidemiology and Data Science, Unit of Bioinformatics and Biostatistics, Milan
| | - M P Colombo
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan.
| |
Collapse
|
20
|
Skjervold AH, Valla M, Ytterhus B, Bofin AM. PAK1 copy number in breast cancer-Associations with proliferation and molecular subtypes. PLoS One 2023; 18:e0287608. [PMID: 37368917 DOI: 10.1371/journal.pone.0287608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION P21-activated kinase 1 (PAK1) is known to be overexpressed in several human tumour types, including breast cancer (BC). It is located on chromosome 11 (11q13.5-q14.1) and plays a significant role in proliferation in BC. In this study we aimed to assess PAK1 gene copy number (CN) in primary breast tumours and their corresponding lymph node metastases, and associations between PAK1 CN and proliferation status, molecular subtype, and prognosis. In addition, we aimed to study associations between CNs of PAK1 and CCND1. Both genes are located on the long arm of chromosome 11 (11q13). METHODS Fluorescence in situ hybridization for PAK1 and Chromosome enumeration probe (CEP)11 were used on tissue microarray sections from a series of 512 BC cases. Copy numbers were estimated by counting the number of fluorescent signals for PAK1 and CEP11 in 20 tumour cell nuclei. Pearson's x2 test was performed to assess associations between PAK1 CN and tumour features, and between PAK1 and CCND1 CNs. Cumulative risk of death from BC and hazard ratios were estimated in analysis of prognosis. RESULTS We found mean PAK1 CN ≥4<6 in 26 (5.1%) tumours, and CN ≥ 6 in 22 (4.3%) tumours. The proportion of cases with copy number increase (mean CN ≥4) was highest among HER2 type and Luminal B (HER2-) tumours. We found an association between PAK1 CN increase, and high proliferation, and high histological grade, but not prognosis. Of cases with PAK1 CN ≥ 6, 30% also had CCND1 CN ≥ 6. CONCLUSIONS PAK1 copy number increase is associated with high proliferation and high histological grade, but not with prognosis. PAK1 CN increase was most frequent in the HER2 type and Luminal B (HER2-) subtype. PAK1 CN increase is associated with CN increase of CCND1.
Collapse
Affiliation(s)
- Anette H Skjervold
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Valla
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Pathology, St. Olav's Hospital, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna M Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
21
|
García-Torralba E, Navarro Manzano E, Luengo-Gil G, De la Morena Barrio P, Chaves Benito A, Pérez-Ramos M, Álvarez-Abril B, Ivars Rubio A, García-Garre E, Ayala de la Peña F, García-Martínez E. A new prognostic model including immune biomarkers, genomic proliferation tumor markers ( AURKA and MYBL2) and clinical-pathological features optimizes prognosis in neoadjuvant breast cancer patients. Front Oncol 2023; 13:1182725. [PMID: 37313470 PMCID: PMC10258327 DOI: 10.3389/fonc.2023.1182725] [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: 03/09/2023] [Accepted: 05/17/2023] [Indexed: 06/15/2023] Open
Abstract
Background Up to 30% of breast cancer (BC) patients treated with neoadjuvant chemotherapy (NCT) will relapse. Our objective was to analyze the predictive capacity of several markers associated with immune response and cell proliferation combined with clinical parameters. Methods This was a single-center, retrospective cohort study of BC patients treated with NCT (2001-2010), in whom pretreatment biomarkers were analyzed: neutrophil-to-lymphocyte ratio (NLR) in peripheral blood, CD3+ tumor-infiltrating lymphocytes (TILs), and gene expression of AURKA, MYBL2 and MKI67 using qRT-PCR. Results A total of 121 patients were included. Median followup was 12 years. In a univariate analysis, NLR, TILs, AURKA, and MYBL2 showed prognostic value for overall survival. In multivariate analyses, including hormone receptor, HER2 status, and response to NCT, NLR (HR 1.23, 95% CI 1.01-1.75), TILs (HR 0.84, 95% CI 0.73-0.93), AURKA (HR 1.05, 95% CI 1.00-1.11) and MYBL2 (HR 1.19, 95% CI 1.05-1.35) remained as independent predictor variables. Conclusion Consecutive addition of these biomarkers to a regression model progressively increased its discriminatory capacity for survival. Should independent cohort studies validate these findings, management of early BC patients may well be changed.
Collapse
Affiliation(s)
- Esmeralda García-Torralba
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Esther Navarro Manzano
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Gines Luengo-Gil
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Pilar De la Morena Barrio
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | | | - Miguel Pérez-Ramos
- Department of Pathology, University Hospital Morales Meseguer, Murcia, Spain
| | - Beatriz Álvarez-Abril
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Alejandra Ivars Rubio
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Elisa García-Garre
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Francisco Ayala de la Peña
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Elena García-Martínez
- Department of Haematology and Medical Oncology, University Hospital Morales Meseguer, Murcia, Spain
- Department of Medicine, Medical School, University of Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
- Medical School, Catholic University of Murcia, Murcia, Spain
| |
Collapse
|
22
|
Stenmark Tullberg A, Sjöström M, Tran L, Niméus E, Killander F, Kovács A, Lundstedt D, Holmberg E, Karlsson P. Combining histological grade, TILs, and the PD-1/PD-L1 pathway to identify immunogenic tumors and de-escalate radiotherapy in early breast cancer: a secondary analysis of a randomized clinical trial. J Immunother Cancer 2023; 11:e006618. [PMID: 37208129 PMCID: PMC10201214 DOI: 10.1136/jitc-2022-006618] [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] [Accepted: 03/17/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND The implementation of immunological biomarkers for radiotherapy (RT) individualization in breast cancer requires consideration of tumor-intrinsic factors. This study aimed to investigate whether the integration of histological grade, tumor-infiltrating lymphocytes (TILs), programmed cell death protein-1 (PD-1), and programmed death ligand-1 (PD-L1) can identify tumors with aggressive characteristics that can be downgraded regarding the need for RT. METHODS The SweBCG91RT trial included 1178 patients with stage I-IIA breast cancer, randomized to breast-conserving surgery with or without adjuvant RT, and followed for a median time of 15.2 years. Immunohistochemical analyses of TILs, PD-1, and PD-L1 were performed. An activated immune response was defined as stromal TILs ≥10% and PD-1 and/or PD-L1 expression in ≥1% of lymphocytes. Tumors were categorized as high-risk or low-risk using assessments of histological grade and proliferation as measured by gene expression. The risk of ipsilateral breast tumor recurrence (IBTR) and benefit of RT were then analyzed with 10 years follow-up based on the integration of immune activation and tumor-intrinsic risk group. RESULTS Among high-risk tumors, an activated immune infiltrate was associated with a reduced risk of IBTR (HR 0.34, 95% CI 0.16 to 0.73, p=0.006). The incidence of IBTR in this group was 12.1% (5.6-25.0) without RT and 4.4% (1.1-16.3) with RT. In contrast, the incidence of IBTR in the high-risk group without an activated immune infiltrate was 29.6% (21.4-40.2) without RT and 12.8% (6.6-23.9) with RT. Among low-risk tumors, no evidence of a favorable prognostic effect of an activated immune infiltrate was seen (HR 2.0, 95% CI 0.87 to 4.6, p=0.100). CONCLUSIONS Integrating histological grade and immunological biomarkers can identify tumors with aggressive characteristics but a low risk of IBTR despite a lack of RT boost and systemic therapy. Among high-risk tumors, the risk reduction of IBTR conferred by an activated immune infiltrate is comparable to treatment with RT. These findings may apply to cohorts dominated by estrogen receptor-positive tumors.
Collapse
Affiliation(s)
- Axel Stenmark Tullberg
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Martin Sjöström
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
| | - Lena Tran
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
| | - Emma Niméus
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Fredrika Killander
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Dan Lundstedt
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Erik Holmberg
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Per Karlsson
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| |
Collapse
|
23
|
Guo X, Gao Y, Song Q, Wei J, Wu J, Dong J, Chen L, Xu S, Wu D, Yang X, Chen L, Li X, Ji G, Lv X, Wei B. Early assessment of circulating exosomal lncRNA-GC1 for monitoring neoadjuvant chemotherapy response in gastric cancer. Int J Surg 2023; 109:1094-1104. [PMID: 37222716 PMCID: PMC10389467 DOI: 10.1097/js9.0000000000000249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/22/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND The timing of surgery for patients with gastric cancer (GC) who undergo neoadjuvant chemotherapy (neoCT) was mainly guided by serial radiologic imaging. However, an earlier assessment was indispensable to avoid delayed treatment for nonresponders and excessive toxicity for responders. Our previous study has identified circulating extracellular vesicles-derived lncRNA-GC1 as a biomarker for early detection and monitoring progression of GC. However, the potential role of neoCT remains poorly understood. METHODS In this explorative biomarker analysis, we conducted a multi-cohort study to examine longitudinal levels of circulating extracellular vesicles-derived lncRNA-GC1 in 798 patients enrolled in the RESONANCE study (NCT01583361). Both circulating extracellular vesicles-derived lncRNA-GC1 and traditional gastrointestinal biomarkers were assessed at defined time nodes. Computed tomography (CT) scans were performed before treatment and 8-10 weeks and assessed based on the RECIST criteria. RESULTS Circulating extracellular vesicles-derived lncRNA-GC1 could be detected in 96.3% of patients at baseline, and significant reductions were observed before cycle 2 (P<0.0001). Levels of circulating extracellular vesicles-derived lncRNA-GC1 showed a stronger correlation with tumor burden and exhibited earlier dynamic changes than the traditional gastrointestinal biomarkers during the first cycle of neoCT. Strong agreement was observed between circulating extracellular vesicles-derived lncRNA-GC1 response (reduction >50%) and radiographic response (Cohen's κ, 0.704). Importantly, circulating extracellular vesicles-derived lncRNA-GC1 maintained predictive value in two external cohorts. Patients with circulating extracellular vesicles-derived lncRNA-GC1 response showed superior disease-free survival [hazard ratio (HR), 0.6238; 95% CI, 0.4095-0.9501; P=0.0118] and overall survival (HR, 0.6131; 95% CI, 0.4016-0.9358; P=0.0090). CONCLUSION Circulating extracellular vesicles-derived lncRNA-GC1 is an early marker of neoCT efficacy and predicts superior survival in GC patients treated with neoCT.
Collapse
Affiliation(s)
- Xin Guo
- Department of Digestive Surgery
- Department of Endoscopic Surgery, Air Force 986th Hospital, Fourth Military Medical University, Xian
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yunge Gao
- Department of Gynecology and Obstetrics
| | - Qiying Song
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | | | | | - Jian Dong
- Department of Gynecology and Obstetrics
| | | | - Shenhui Xu
- Department of Pathology, Xijing Hospital
| | - Di Wu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | | | - Lubin Chen
- Department of Digestive Surgery
- Department of Endoscopic Surgery, Air Force 986th Hospital, Fourth Military Medical University, Xian
| | | | - Gang Ji
- Department of Digestive Surgery
| | | | - Bo Wei
- Department of General Surgery, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
24
|
Kim H, Whitman AA, Wisniewska K, Kakati RT, Garcia-Recio S, Calhoun BC, Franco HL, Perou CM, Spanheimer PM. Tamoxifen Response at Single Cell Resolution in Estrogen Receptor-Positive Primary Human Breast Tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.01.535159. [PMID: 37066379 PMCID: PMC10103953 DOI: 10.1101/2023.04.01.535159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
In ER+/HER2- breast cancer, multiple measures of intra-tumor heterogeneity are associated with worse response to endocrine therapy. To investigate heterogeneity in response to treatment, we developed an operating room-to-laboratory pipeline for the collection of live human tumors and normal breast specimens immediately after surgical resection for processing into single-cell workflows for experimentation and genomic analyses. We demonstrate differences in tamoxifen response by cell type and identify distinctly responsive and resistant subpopulations within the malignant cell compartment of human tumors. Tamoxifen resistance signatures from 3 distinct resistant subpopulations are prognostic in large cohorts of ER+ breast cancer patients and enriched in endocrine therapy resistant tumors. This novel ex vivo model system now provides a foundation to define responsive and resistant sub-populations within heterogeneous tumors, to develop precise single cell-based predictors of response to therapy, and to identify genes and pathways driving resistance to therapy.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Austin A. Whitman
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Rasha T. Kakati
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Benjamin C. Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hector L. Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
- Computational Medicine Program, University of North Carolina, Chapel Hill, NC
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
- Computational Medicine Program, University of North Carolina, Chapel Hill, NC
| | - Philip M. Spanheimer
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
- Department of Surgery, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
25
|
Li J, Chen Y, Ye W, Zhang M, Zhu J, Zhi W, Cheng Q. Molecular breast cancer subtype identification using photoacoustic spectral analysis and machine learning at the biomacromolecular level. PHOTOACOUSTICS 2023; 30:100483. [PMID: 37063308 PMCID: PMC10090435 DOI: 10.1016/j.pacs.2023.100483] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
Breast cancer threatens the health of women worldwide, and its molecular subtypes largely determine the therapy and prognosis of patients. However, an uncomplicated and accurate method to identify subtypes is currently lacking. This study utilized photoacoustic spectral analysis (PASA) based on the partial least squares discriminant algorithm (PLS-DA) to identify molecular breast cancer subtypes at the biomacromolecular level in vivo. The area of power spectrum density (APSD) was extracted to semi-quantify the biomacromolecule content. The feature wavelengths were obtained via the variable importance in projection (VIP) score and the selectivity ratio (Sratio), to identify the biomarkers. The PASA achieved an accuracy of 84%. Most of the feature wavelengths fell into the collagen-dominated absorption waveband, which was consistent with the histopathological results. This paper proposes a successful method for identifying molecular breast cancer subtypes and proves that collagen can be treated as a biomarker for molecular breast cancer subtyping.
Collapse
Affiliation(s)
- Jiayan Li
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Yingna Chen
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Wanli Ye
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Mengjiao Zhang
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Jingtao Zhu
- School of Physics Science and Engineering, Tongji University, Shanghai, China
| | - Wenxiang Zhi
- Department of Ultrasonography, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, China
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China
| |
Collapse
|
26
|
Han S, Lee SB, Gong G, Lee J, Chae SY, Oh JS, Moon DH. Prognostic significance of pretreatment 18F-fluorodeoxyglucose positron emission tomography/computed tomography in patients with T2N1 hormone receptor-positive, ERBB2-negative breast cancer who underwent adjuvant chemotherapy. Breast Cancer Res Treat 2023; 198:207-215. [PMID: 36633721 DOI: 10.1007/s10549-022-06852-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023]
Abstract
PURPOSE To determine whether tumor uptake of 18F-fluorodeoxyglucose (18F-FDG) is associated with invasive disease-free survival (IDFS) in patients with hormone receptor (HR)-positive ERBB2-negative early-stage breast cancer treated with adjuvant chemotherapy. METHODS This is a single-center cohort study of women with breast cancer who underwent surgery between 2008 and 2015 at Asan Medical Center, Seoul, Korea. Patients were enrolled if they were diagnosed with HR-positive ERBB2-negative breast cancer with histology of invasive ductal carcinoma, had an American Joint Committee on Cancer pathologic tumor stage of T2N1 with 1-3 positive axillary nodes, underwent preoperative 18F-FDG positron emission tomography/computed tomography (PET/CT), and underwent breast cancer surgery followed by anthracycline- or taxane-based adjuvant chemotherapy. The primary outcome measure was IDFS. The maximum standardized uptake value (SUVmax) was dichotomized using a predefined cut-off of 4.14. RESULTS A total of 129 patients were included. The median follow-up period for IDFS in those without recurrence was 82 months (interquartile range, 65-106). Multivariable Cox analysis showed that SUVmax was independently associated with IDFS [adjusted hazard ratio 2.49; 95% confidence interval (CI), 1.06-5.84]. Ten-year IDFS estimates via the Kaplan-Meier method were 0.60 (95% CI, 0.42-0.74) and 0.82 (95% CI, 0.65-0.91) for high and low SUVmax groups, respectively. The overall association between SUVmax and IDFS appeared to be consistent across subgroups divided according to age, progesterone receptor status, histologic grade, or presence of lymphovascular invasion. CONCLUSION High SUVmax on preoperative 18F-FDG PET/CT was independently associated with reduced long-term IDFS in T2N1 HR-positive ERBB2-negative breast cancer patients who underwent adjuvant chemotherapy.
Collapse
Affiliation(s)
- Sangwon Han
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sae Byul Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jungbok Lee
- Division of Biostatistics, Center for Medical Research and Information, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sun Young Chae
- Department of Nuclear Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea
| | - Jungsu S Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dae Hyuk Moon
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Circulating tumor DNA reveals complex biological features with clinical relevance in metastatic breast cancer. Nat Commun 2023; 14:1157. [PMID: 36859416 PMCID: PMC9977734 DOI: 10.1038/s41467-023-36801-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 02/14/2023] [Indexed: 03/03/2023] Open
Abstract
Liquid biopsy has proven valuable in identifying individual genetic alterations; however, the ability of plasma ctDNA to capture complex tumor phenotypes with clinical value is unknown. To address this question, we have performed 0.5X shallow whole-genome sequencing in plasma from 459 patients with metastatic breast cancer, including 245 patients treated with endocrine therapy and a CDK4/6 inhibitor (ET + CDK4/6i) from 2 independent cohorts. We demonstrate that machine learning multi-gene signatures, obtained from ctDNA, identify complex biological features, including measures of tumor proliferation and estrogen receptor signaling, similar to what is accomplished using direct tumor tissue DNA or RNA profiling. More importantly, 4 DNA-based subtypes, and a ctDNA-based genomic signature tracking retinoblastoma loss-of-heterozygosity, are significantly associated with poor response and survival outcome following ET + CDK4/6i, independently of plasma tumor fraction. Our approach opens opportunities for the discovery of additional multi-feature genomic predictors coming from ctDNA in breast cancer and other cancer-types.
Collapse
|
28
|
Waliany S, Caswell-Jin J, Riaz F, Myall N, Zhu H, Witteles RM, Neal JW. Pharmacovigilance Analysis of Heart Failure Associated With Anti-HER2 Monotherapies and Combination Regimens for Cancer. JACC CardioOncol 2023; 5:85-98. [PMID: 36875913 PMCID: PMC9982216 DOI: 10.1016/j.jaccao.2022.09.007] [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: 05/01/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 01/18/2023] Open
Abstract
Background Trastuzumab improves outcomes in patients with HER2-overexpressing malignancies but is associated with decreases in left ventricular ejection fraction. Heart failure (HF) risks from other anti-HER2 therapies are less clear. Objectives Using World Health Organization pharmacovigilance data, the authors compared HF odds across anti-HER2 regimens. Methods In VigiBase, 41,976 patients had adverse drug reactions (ADRs) with anti-HER2 monoclonal antibodies (trastuzumab, n = 16,900; pertuzumab, n = 1,856), antibody-drug conjugates (trastuzumab emtansine [T-DM1], n = 3,983; trastuzumab deruxtecan, n = 947), and tyrosine kinase inhibitors (afatinib, n = 10,424; lapatinib, n = 5,704; neratinib, n = 1,507; tucatinib, n = 655); additionally, 36,052 patients had ADRs with anti-HER2-based combination regimens. Most patients had breast cancer (monotherapies, n = 17,281; combinations, n = 24,095). Outcomes included comparison of HF odds with each monotherapy relative to trastuzumab, within each therapeutic class, and among combination regimens. Results Of 16,900 patients with trastuzumab-associated ADRs, 2,034 (12.04%) had HF reports (median time to onset 5.67 months; IQR: 2.85-9.32 months) compared with 1% to 2% with antibody-drug conjugates. Trastuzumab had higher odds of HF reporting relative to other anti-HER2 therapies collectively in the overall cohort (reporting OR [ROR]: 17.37; 99% CI: 14.30-21.10) and breast cancer subgroup (ROR: 17.10; 99% CI: 13.12-22.27). Pertuzumab/T-DM1 had 3.4 times higher odds of HF reporting than T-DM1 monotherapy; tucatinib/trastuzumab/capecitabine had similar odds as tucatinib. Among metastatic breast cancer regimens, HF odds were highest with trastuzumab/pertuzumab/docetaxel (ROR: 1.42; 99% CI: 1.17-1.72) and lowest with lapatinib/capecitabine (ROR: 0.09; 99% CI: 0.04-0.23). Conclusions Trastuzumab and pertuzumab/T-DM1 had higher odds of HF reporting than other anti-HER2 therapies. These data provide large-scale, real-world insight into which HER2-targeted regimens would benefit from left ventricular ejection fraction monitoring.
Collapse
Key Words
- AC-THP, doxorubicin/cyclophosphamide followed by paclitaxel/trastuzumab/pertuzumab
- ACTH, doxorubicin/cyclophosphamide followed by trastuzumab/paclitaxel
- ADC, antibody-drug conjugate
- ADR, adverse drug reaction
- AI, aromatase inhibitor
- FDA, U.S. Food and Drug Administration
- HER2
- HF, heart failure
- IC, information component
- LVEF, left ventricular ejection fraction
- ROR, reporting odds ratio
- T-DM1, trastuzumab emtansine
- T-DXd, trastuzumab deruxtecan
- antibody-drug conjugates
- heart failure
- trastuzumab
- tyrosine kinase inhibitors
Collapse
Affiliation(s)
- Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Jennifer Caswell-Jin
- Division of Oncology, Stanford University School of Medicine, Palo Alto, California, USA.,Stanford Cancer Institute, Palo Alto, California, USA
| | - Fauzia Riaz
- Division of Oncology, Stanford University School of Medicine, Palo Alto, California, USA.,Stanford Cancer Institute, Palo Alto, California, USA
| | - Nathaniel Myall
- Division of Oncology, Stanford University School of Medicine, Palo Alto, California, USA.,Stanford Cancer Institute, Palo Alto, California, USA
| | - Han Zhu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, California, USA
| | - Ronald M Witteles
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Joel W Neal
- Division of Oncology, Stanford University School of Medicine, Palo Alto, California, USA.,Stanford Cancer Institute, Palo Alto, California, USA
| |
Collapse
|
29
|
Finkelman BS, Zhang H, Hicks DG, Turner BM. The Evolution of Ki-67 and Breast Carcinoma: Past Observations, Present Directions, and Future Considerations. Cancers (Basel) 2023; 15:808. [PMID: 36765765 PMCID: PMC9913317 DOI: 10.3390/cancers15030808] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
The 1983 discovery of a mouse monoclonal antibody-the Ki-67 antibody-that recognized a nuclear antigen present only in proliferating cells represented a seminal discovery for the pathologic assessment of cellular proliferation in breast cancer and other solid tumors. Cellular proliferation is a central determinant of prognosis and response to cytotoxic chemotherapy in patients with breast cancer, and since the discovery of the Ki-67 antibody, Ki-67 has evolved as an important biomarker with both prognostic and predictive potential in breast cancer. Although there is universal recognition among the international guideline recommendations of the value of Ki-67 in breast cancer, recommendations for the actual use of Ki-67 assays in the prognostic and predictive evaluation of breast cancer remain mixed, primarily due to the lack of assay standardization and inconsistent inter-observer and inter-laboratory reproducibility. The treatment of high-risk ER-positive/human epidermal growth factor receptor-2 (HER2) negative breast cancer with the recently FDA-approved drug abemaciclib relies on a quantitative assessment of Ki-67 expression in the treatment decision algorithm. This further reinforces the urgent need for standardization of Ki-67 antibody selection and staining interpretation, which will hopefully lead to multidisciplinary consensus on the use of Ki-67 as a prognostic and predictive marker in breast cancer. The goals of this review are to highlight the historical evolution of Ki-67 in breast cancer, summarize the present literature on Ki-67 in breast cancer, and discuss the evolving literature on the use of Ki-67 as a companion diagnostic biomarker in breast cancer, with consideration for the necessary changes required across pathology practices to help increase the reliability and widespread adoption of Ki-67 as a prognostic and predictive marker for breast cancer in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Bradley M. Turner
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14620, USA
| |
Collapse
|
30
|
Klæstad E, Opdahl S, Raj SX, Bofin AM, Valla M. Long term trends of breast cancer incidence according to proliferation status. BMC Cancer 2022; 22:1340. [PMID: 36544164 PMCID: PMC9773605 DOI: 10.1186/s12885-022-10438-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Long-term breast cancer incidence trends according to proliferation status are poorly described. We studied time-trends in breast cancer incidence, using mitotic count and Ki-67 as markers of proliferation. METHODS Among 83,298 Norwegian women followed for breast cancer occurrence 1961-2012, 2995 incident breast cancers were diagnosed. Ki-67 was assessed using immunohistochemistry on tissue microarrays and mitoses were counted on whole sections. We compared incidence rates according to proliferation status among women born 1886-1928 and 1929-1977, estimating age-specific incidence rate ratios. We performed multiple imputations to account for unknown proliferation status. Mean values of Ki-67 and mitotic counts were calculated, according to age and birth year. We performed separate incidence analyses for HER2+ and triple negative breast cancers. RESULTS Among women aged 40-69 years, incidence rates of tumours with low-proliferative activity were higher among those born in 1929 or later, compared to before 1929, according to Ki-67 and mitotic count. Incidence rates of tumours with high-proliferative activity were also higher in women born in 1929 or later compared to before 1929 according to Ki-67, but not according to mitotic count. Mean values of Ki-67 and mitotic count varied according to age and birth year. In subtype-specific analyses we found an increase of high-proliferative HER2+ tumours according to Ki-67 in women born in 1929 or later, compared to before 1929. CONCLUSIONS There has been a temporal increase in both low- and high-proliferative breast cancers.
Collapse
Affiliation(s)
- Elise Klæstad
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Signe Opdahl
- grid.5947.f0000 0001 1516 2393Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sunil Xavier Raj
- grid.52522.320000 0004 0627 3560Cancer Clinic, St. Olav’s Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Anna Mary Bofin
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Valla
- grid.5947.f0000 0001 1516 2393Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway ,grid.52522.320000 0004 0627 3560Department of Pathology, St. Olav’s Hospital, Trondheim University Hospital, 7006 Trondheim, Norway ,grid.52522.320000 0004 0627 3560Clinic of Laboratory Medicine, St. Olav’s Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| |
Collapse
|
31
|
Signatures of Breast Cancer Progression in the Blood: What Could Be Learned from Circulating Tumor Cell Transcriptomes. Cancers (Basel) 2022; 14:cancers14225668. [PMID: 36428760 PMCID: PMC9688726 DOI: 10.3390/cancers14225668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Gene expression profiling has revolutionized our understanding of cancer biology, showing an unprecedented ability to impact patient management especially in breast cancer. The vast majority of breast cancer gene expression signatures derive from the analysis of the tumor bulk, an experimental approach that limits the possibility to dissect breast cancer heterogeneity thoroughly and might miss the message hidden in biologically and clinically relevant cell populations. During disease progression or upon selective pressures, cancer cells undergo continuous transcriptional changes, which inevitably affect tumor heterogeneity, response to therapy and tendency to disseminate. Therefore, metastasis-associated signatures and transcriptome-wide gene expression measurement at single-cell resolution hold great promise for the future of breast cancer clinical care. Seen from this perspective, transcriptomics of circulating tumor cells (CTCs) represent an attractive opportunity to bridge the knowledge gap and develop novel biomarkers. This review summarizes the current state-of-the-science on CTC gene expression analysis in breast cancer, addresses technical and clinical issues related to the application of CTC-derived signatures, and discusses potential research directions.
Collapse
|
32
|
Muzzi JCD, Magno JM, Souza JS, Alvarenga LM, de Moura JF, Figueiredo BC, Castro MAA. Comprehensive Characterization of the Regulatory Landscape of Adrenocortical Carcinoma: Novel Transcription Factors and Targets Associated with Prognosis. Cancers (Basel) 2022; 14:5279. [PMID: 36358698 PMCID: PMC9657296 DOI: 10.3390/cancers14215279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 08/31/2023] Open
Abstract
We reconstructed a transcriptional regulatory network for adrenocortical carcinoma (ACC) using transcriptomic and clinical data from The Cancer Genome Atlas (TCGA)-ACC cohort. We investigated the association of transcriptional regulatory units (regulons) with overall survival, molecular phenotypes, and immune signatures. We annotated the ACC regulons with cancer hallmarks and assessed single sample regulon activities in the European Network for the Study of Adrenal Tumors (ENSAT) cohort. We found 369 regulons associated with overall survival and subdivided them into four clusters: RC1 and RC2, associated with good prognosis, and RC3 and RC4, associated with worse outcomes. The RC1 and RC3 regulons were highly correlated with the 'Steroid Phenotype,' while the RC2 and RC4 regulons were highly correlated with a molecular proliferation signature. We selected two regulons, NR5A1 (steroidogenic factor 1, SF-1) and CENPA (Centromeric Protein A), that were consistently associated with overall survival for further downstream analyses. The CENPA regulon was the primary regulator of MKI-67 (a marker of proliferation KI-67), while the NR5A1 regulon is a well-described transcription factor (TF) in ACC tumorigenesis. We also found that the ZBTB4 (Zinc finger and BTB domain-containing protein 4) regulon, which is negatively associated with CENPA in our transcriptional regulatory network, is also a druggable anti-tumorigenic TF. We anticipate that the ACC regulons may be used as a reference for further investigations concerning the complex molecular interactions in ACC tumors.
Collapse
Affiliation(s)
- João C. D. Muzzi
- Laboratório de Imunoquímica (LIMQ), Pós-Graduação em Microbiologia, Parasitologia e Patologia, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba 81530-990, Brazil
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba 81520-260, Brazil
- Oncology Division, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
| | - Jéssica M. Magno
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba 81520-260, Brazil
- Oncology Division, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
| | - Jean S. Souza
- Oncology Division, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
| | - Larissa M. Alvarenga
- Laboratório de Imunoquímica (LIMQ), Pós-Graduação em Microbiologia, Parasitologia e Patologia, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba 81530-990, Brazil
| | - Juliana F. de Moura
- Laboratório de Imunoquímica (LIMQ), Pós-Graduação em Microbiologia, Parasitologia e Patologia, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba 81530-990, Brazil
| | - Bonald C. Figueiredo
- Oncology Division, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
- Molecular Oncology Laboratory, Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Curitiba 80030-110, Brazil
| | - Mauro A. A. Castro
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba 81520-260, Brazil
| |
Collapse
|
33
|
van Leeuwen JE, Ba-Alawi W, Branchard E, Cruickshank J, Schormann W, Longo J, Silvester J, Gross PL, Andrews DW, Cescon DW, Haibe-Kains B, Penn LZ, Gendoo DMA. Computational pharmacogenomic screen identifies drugs that potentiate the anti-breast cancer activity of statins. Nat Commun 2022; 13:6323. [PMID: 36280687 PMCID: PMC9592602 DOI: 10.1038/s41467-022-33144-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 09/02/2022] [Indexed: 12/25/2022] Open
Abstract
Statins, a family of FDA-approved cholesterol-lowering drugs that inhibit the rate-limiting enzyme of the mevalonate metabolic pathway, have demonstrated anticancer activity. Evidence shows that dipyridamole potentiates statin-induced cancer cell death by blocking a restorative feedback loop triggered by statin treatment. Leveraging this knowledge, we develop an integrative pharmacogenomics pipeline to identify compounds similar to dipyridamole at the level of drug structure, cell sensitivity and molecular perturbation. To overcome the complex polypharmacology of dipyridamole, we focus our pharmacogenomics pipeline on mevalonate pathway genes, which we name mevalonate drug-network fusion (MVA-DNF). We validate top-ranked compounds, nelfinavir and honokiol, and identify that low expression of the canonical epithelial cell marker, E-cadherin, is associated with statin-compound synergy. Analysis of remaining prioritized hits led to the validation of additional compounds, clotrimazole and vemurafenib. Thus, our computational pharmacogenomic approach identifies actionable compounds with pathway-specific activities.
Collapse
Affiliation(s)
- Jenna E. van Leeuwen
- grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Wail Ba-Alawi
- grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Emily Branchard
- grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Jennifer Cruickshank
- grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Wiebke Schormann
- grid.17063.330000 0001 2157 2938Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON M4N 3M5 Canada
| | - Joseph Longo
- grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Jennifer Silvester
- grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Peter L. Gross
- grid.25073.330000 0004 1936 8227Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8 Canada
| | - David W. Andrews
- grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.17063.330000 0001 2157 2938Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, ON M4N 3M5 Canada
| | - David W. Cescon
- grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.17063.330000 0001 2157 2938Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, 27 King’s College Circle, Toronto, ON M5S 1A1 Canada
| | - Benjamin Haibe-Kains
- grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.17063.330000 0001 2157 2938Department of Computer Science, University of Toronto, 10 King’s College Road, Toronto, ON M5S 3G4 Canada ,grid.419890.d0000 0004 0626 690XOntario Institute of Cancer Research, 661 University Avenue, Suite 510, Toronto, ON M5G 0A3 Canada
| | - Linda Z. Penn
- grid.17063.330000 0001 2157 2938Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7 Canada ,grid.231844.80000 0004 0474 0428Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Deena M. A. Gendoo
- grid.6572.60000 0004 1936 7486Centre for Computational Biology, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, Birmingham, B15 2TT UK ,grid.6572.60000 0004 1936 7486Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, Birmingham, B15 2TT UK
| |
Collapse
|
34
|
Wu J, Tian Y, Liu W, Zheng H, Xi Y, Yan Y, Hu Y, Liao B, Wang M, Tang P. A novel twelve-gene signature to predict neoadjuvant chemotherapy response and prognosis in breast cancer. Front Immunol 2022; 13:1035667. [PMID: 36341435 PMCID: PMC9629837 DOI: 10.3389/fimmu.2022.1035667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background Accurate evaluation of the response to neoadjuvant chemotherapy (NAC) provides important information about systemic therapies for breast cancer, which implies pharmacological response, prognosis, and guide further therapy. Gene profiles overcome the shortcomings of the relatively limited detection indicators of the classical pathological evaluation criteria and the subjectivity of observation, but are complicated and expensive. Therefore, it is essential to develop a more accurate, repeatable, and economical evaluation approach for neoadjuvant chemotherapy responses. Methods We analyzed the transcriptional profiles of chemo-resistant breast cancer cell lines and tumors of chemo-resistant breast cancer patients in the GSE25066 dataset. We preliminarily screened out common significantly differentially expressed genes and constructed a NAC response risk model using LASSO regression and univariate and multivariate analyses. The differences in bioinformatic features of tumor cells, immune characteristics, and prognosis were compared between high and low-risk group. The potential drugs that could reverse chemotherapy resistance in breast cancer were screened by the CMap database. Results Thirty-six genes were commonly up/down-regulated in both NAC chemo-resistant tumors and cells compared to the sensitive tumors and wild-type cells. Through LASSO regression, we obtained a risk model composed of 12 genes. The risk model divided patients into high and low-risk groups. Univariate and multivariate Cox regression analyses suggested that the risk score is an independent prognostic factor for evaluating NAC response in breast cancer. Tumors in risk groups exhibited significant differences in molecular biological characteristics, tumor-infiltrating lymphocytes, and immunosuppressive molecule expression. Our results suggested that the risk score was also a good prognostic factor for breast cancer. Finally, we screened potential drugs that could reverse chemotherapy resistance in breast cancer. Conclusion A novel 12 gene-signature could be used to predict NAC response and predict prognosis in breast cancer.
Collapse
Affiliation(s)
- Jin Wu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yuan Tian
- Department of General surgery, Linyi People’s Hospital, Linyi, China
| | - Wei Liu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Hong Zheng
- Department of Neurosurgery, Chongqing General Hospital, Chongqing, China
| | - Yuanyin Xi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yuzhao Yan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Ying Hu
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Bin Liao
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
- *Correspondence: Peng Tang, ; Minghao Wang, ; Bin Liao,
| | - Minghao Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- *Correspondence: Peng Tang, ; Minghao Wang, ; Bin Liao,
| | - Peng Tang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- *Correspondence: Peng Tang, ; Minghao Wang, ; Bin Liao,
| |
Collapse
|
35
|
Yin Q, Chen W, Zhang C, Wei Z. A convolutional neural network model for survival prediction based on prognosis-related cascaded Wx feature selection. J Transl Med 2022; 102:1064-1074. [PMID: 35810236 DOI: 10.1038/s41374-022-00801-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/14/2022] Open
Abstract
Great advances in deep learning have provided effective solutions for prediction tasks in the biomedical field. However, accurate prognosis prediction using cancer genomics data remains challenging due to the severe overfitting problem caused by curse of dimensionality inherent to high-throughput sequencing data. Moreover, there are unique challenges to perform survival analysis, arising from the difficulty in utilizing censored samples whose events of interest are not observed. Convolutional neural network (CNN) models provide us the opportunity to extract meaningful hierarchical features to characterize cancer subtype and prognosis outcomes. On the other hand, feature selection can mitigate overfitting and reduce subsequent model training computation burden by screening out significant genes from redundant genes. To accomplish model simplification, we developed a concise and efficient survival analysis model, named CNN-Cox model, which combines a special CNN framework with prognosis-related feature selection cascaded Wx, with the advantage of less computation demand utilizing light training parameters. Experiment results show that CNN-Cox model achieved consistent higher C-index values and better survival prediction performance across seven cancer type datasets in The Cancer Genome Atlas cohort, including bladder carcinoma, head and neck squamous cell carcinoma, kidney renal cell carcinoma, brain low-grade glioma, lung adenocarcinoma (LUAD), lung squamous cell carcinoma, and skin cutaneous melanoma, compared with the existing state-of-the-art survival analysis methods. As an illustration of model interpretation, we examined potential prognostic gene signatures of LUAD dataset using the proposed CNN-Cox model. We conducted protein-protein interaction network analysis to identify potential prognostic genes and further analyzed the biological function of 13 hub genes, including ANLN, RACGAP1, KIF4A, KIF20A, KIF14, ASPM, CDK1, SPC25, NCAPG, MKI67, HJURP, EXO1, HMMR, whose high expression is significantly associated with poor survival of LUAD patients. These findings confirmed that CNN-Cox model is effective in extracting not only prognosis factors but also biologically meaningful gene features. The codes are available at the GitHub website: https://github.com/wangwangCCChen/CNN-Cox .
Collapse
Affiliation(s)
- Qingyan Yin
- School of Science, Xi'an University of Architecture and Technology, Xi'an, Shaanxi, 710055, China.
| | - Wangwang Chen
- School of Science, Xi'an University of Architecture and Technology, Xi'an, Shaanxi, 710055, China
| | - Chunxia Zhang
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
36
|
The role of HER2 alterations in clinicopathological and molecular characteristics of breast cancer and HER2-targeted therapies: a comprehensive review. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:210. [PMID: 36175719 DOI: 10.1007/s12032-022-01817-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/03/2022] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC) is the most common malignancy in women and one of the leading causes of cancer mortality, despite significant treatment advancements over the last decades. Human epidermal growth factor receptor-2 (HER2) is a member of the ERBB family of receptor tyrosine kinases which have long been known to mediate cancer cell growth and invasion through constitutive activation of oncogenic downstream signaling, such as PI3K/Akt/mTOR and MAPK. Overexpression/amplification of HER2 in various tumors, especially BC, offers the possible therapeutic potential for target therapies. HER2-targeted therapies, either with a combination of chemotherapy or through multi-anti-HER2 therapies without chemotherapy, have significantly improved the prognosis of HER2-positive tumors. In recent years, novel anti-HER2 agents and combination therapies have garnered much attention, especially for heavily treated advanced or metastatic BCs. HER2-positive BC is biologically a heterogeneous group depending on HER2 activation mechanisms, hormone receptor status, genome variations, tumor heterogeneity, and treatment resistance, which affect the treatment benefit and patients' outcomes. This review will discuss HER2 alternations (gene amplification or receptor overexpression) in BC, their correlation with clinicopathological characteristics and molecular characteristics, and HER2-based therapies in tumors with HER2 overexpression/amplification.
Collapse
|
37
|
Wang J, Liang H, Zhang Q, Ma S. Replicability in cancer omics data analysis: measures and empirical explorations. Brief Bioinform 2022; 23:bbac304. [PMID: 35876281 PMCID: PMC9487717 DOI: 10.1093/bib/bbac304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 02/05/2023] Open
Abstract
In biomedical research, the replicability of findings across studies is highly desired. In this study, we focus on cancer omics data, for which the examination of replicability has been mostly focused on important omics variables identified in different studies. In published literature, although there have been extensive attention and ad hoc discussions, there is insufficient quantitative research looking into replicability measures and their properties. The goal of this study is to fill this important knowledge gap. In particular, we consider three sensible replicability measures, for which we examine distributional properties and develop a way of making inference. Applying them to three The Cancer Genome Atlas (TCGA) datasets reveals in general low replicability and significant across-data variations. To further comprehend such findings, we resort to simulation, which confirms the validity of the findings with the TCGA data and further informs the dependence of replicability on signal level (or equivalently sample size). Overall, this study can advance our understanding of replicability for cancer omics and other studies that have identification as a key goal.
Collapse
Affiliation(s)
- Jiping Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Hongmin Liang
- Department of Statistics, School of Economics, Xiamen University, Xiamen, Fujian, China
| | - Qingzhao Zhang
- Department of Statistics, School of Economics, Xiamen University, Xiamen, Fujian, China
- The Wang Yanan Institute for Studies in Economics, Xiamen University, Xiamen, Fujian, China
| | - Shuangge Ma
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
38
|
Kreipe H, Harbeck N, Christgen M. Clinical validity and clinical utility of Ki67 in early breast cancer. Ther Adv Med Oncol 2022; 14:17588359221122725. [PMID: 36105888 PMCID: PMC9465566 DOI: 10.1177/17588359221122725] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Ki67 represents an immunohistochemical nuclear localized marker that is widely
used in surgical pathology. Nuclear immunoreactivity for Ki67 indicates that
cells are cycling and are in G1- to S-phase. The percentage of Ki67-positive
tumor cells (Ki67 index) therefore provides an estimate of the growth fraction
in tumor specimens. In breast cancer (BC), tumor cell proliferation rate is one
of the most relevant prognostic markers and Ki67 is consequently helpful in
prognostication similar to histological grading and mRNA profiling-based BC risk
stratification. In BCs treated with short-term preoperative endocrine therapy,
Ki67 dynamics enable distinguishing between endocrine sensitive and resistant
tumors. Despite its nearly universal use in pathology laboratories worldwide, no
internationally accepted consensus has yet been achieved for some methodological
details related to Ki67 immunohistochemistry (IHC). Controversial issues refer
to choice of IHC antibody clones, scoring methods, inter-laboratory
reproducibility, and the potential value of computer-assisted imaging analysis
and/or artificial intelligence for Ki67 assessment. Prospective clinical trials
focusing on BC treatment have proven that Ki67, as determined by standardized
central pathology assessment, is of clinical validity. Clinical utility has been
demonstrated in huge observational studies.
Collapse
Affiliation(s)
- Hans Kreipe
- Institute of Pathology, Hannover Medical School, Carl-Neubergstraße 1, Hannover 30625, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München (LMU) Frauenklinik Maistrasse-Innenstadt und Klinikum Großhadern, Germany
| | | |
Collapse
|
39
|
Lorusso G, Wyss CB, Kuonen F, Vannini N, Billottet C, Duffey N, Pineau R, Lan Q, Wirapati P, Barras D, Tancredi A, Lyck R, Lehr HA, Engelhardt B, Delorenzi M, Bikfalvi A, Rüegg C. Connexins orchestrate progression of breast cancer metastasis to the brain by promoting FAK activation. Sci Transl Med 2022; 14:eaax8933. [PMID: 36070364 DOI: 10.1126/scitranslmed.aax8933] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Brain metastasis is a complication of increasing incidence in patients with breast cancer at advanced disease stage. It is a severe condition characterized by a rapid decline in quality of life and poor prognosis. There is a critical clinical need to develop effective therapies to prevent and treat brain metastases. Here, we describe a unique and robust spontaneous preclinical model of breast cancer metastasis to the brain (4T1-BM2) in mice that has been instrumental in uncovering molecular mechanisms guiding metastatic dissemination and colonization of the brain. Key experimental findings were validated in the additional murine D2A1-BM2 model and in human MDA231-BrM2 model. Gene expression analyses and functional studies, coupled with clinical transcriptomic and histopathological investigations, identified connexins (Cxs) and focal adhesion kinase (FAK) as master molecules orchestrating breast cancer colonization of the brain. Cx31 promoted homotypic tumor cell adhesion, heterotypic tumor-astrocyte interaction, and FAK phosphorylation. FAK signaling prompted NF-κB activation inducing Lamc2 expression and laminin 332 (laminin 5) deposition, α6 integrin-mediated adhesion, and sustained survival and growth within brain parenchyma. In the MDA231-BrM2 model, the human homologous molecules CX43, LAMA4, and α3 integrin were involved. Systemic treatment with FAK inhibitors reduced brain metastasis progression. In conclusion, we report a spontaneous model of breast cancer metastasis to the brain and identified Cx-mediated FAK-NF-κB signaling as a mechanism promoting cell-autonomous and microenvironmentally controlled cell survival for brain colonization. Considering the limited therapeutic options for brain metastatic disease in cancer patients, we propose FAK as a therapeutic candidate to further pursue in the clinic.
Collapse
Affiliation(s)
- Girieca Lorusso
- Experimental and Translational Oncology, Pathology Unit, Department of Oncology Microbiology and Immunology (OMI), Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland.,Division of Experimental Oncology, Multidisciplinary Oncology Center (CePO), Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Faculty of Biology and Medicine, Epalinges 1066, Switzerland.,National Center for Competence in Research (NCCR) Molecular Oncology, Swiss Institute of Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne (ISREC-EPFL), Lausanne 1015, Switzerland
| | - Christof B Wyss
- Experimental and Translational Oncology, Pathology Unit, Department of Oncology Microbiology and Immunology (OMI), Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - François Kuonen
- Division of Experimental Oncology, Multidisciplinary Oncology Center (CePO), Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Faculty of Biology and Medicine, Epalinges 1066, Switzerland.,National Center for Competence in Research (NCCR) Molecular Oncology, Swiss Institute of Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne (ISREC-EPFL), Lausanne 1015, Switzerland
| | - Nicola Vannini
- Ludwig Institute for Cancer Research (LICR), Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Epalinges 1066, Switzerland
| | | | - Nathalie Duffey
- Experimental and Translational Oncology, Pathology Unit, Department of Oncology Microbiology and Immunology (OMI), Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Raphael Pineau
- INSERM U1029 and University of Bordeaux, Pessac Cedex 33615, France
| | - Qiang Lan
- Experimental and Translational Oncology, Pathology Unit, Department of Oncology Microbiology and Immunology (OMI), Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland.,Division of Experimental Oncology, Multidisciplinary Oncology Center (CePO), Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Faculty of Biology and Medicine, Epalinges 1066, Switzerland.,National Center for Competence in Research (NCCR) Molecular Oncology, Swiss Institute of Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne (ISREC-EPFL), Lausanne 1015, Switzerland
| | - Pratyaksha Wirapati
- Bioinformatics Core Facility, Swiss Institute for Bioinformatics (SIB), Lausanne 1015, Switzerland
| | - David Barras
- Bioinformatics Core Facility, Swiss Institute for Bioinformatics (SIB), Lausanne 1015, Switzerland
| | - Alessandro Tancredi
- Experimental and Translational Oncology, Pathology Unit, Department of Oncology Microbiology and Immunology (OMI), Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern (UNIBE), Bern 3012, Switzerland
| | - Hans-Anton Lehr
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne 1011, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern (UNIBE), Bern 3012, Switzerland
| | - Mauro Delorenzi
- Bioinformatics Core Facility, Swiss Institute for Bioinformatics (SIB), Lausanne 1015, Switzerland
| | - Andreas Bikfalvi
- INSERM U1029 and University of Bordeaux, Pessac Cedex 33615, France
| | - Curzio Rüegg
- Experimental and Translational Oncology, Pathology Unit, Department of Oncology Microbiology and Immunology (OMI), Faculty of Science and Medicine, University of Fribourg, Fribourg 1700, Switzerland.,Division of Experimental Oncology, Multidisciplinary Oncology Center (CePO), Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Faculty of Biology and Medicine, Epalinges 1066, Switzerland.,National Center for Competence in Research (NCCR) Molecular Oncology, Swiss Institute of Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne (ISREC-EPFL), Lausanne 1015, Switzerland
| |
Collapse
|
40
|
Assessment of MRI to estimate metastatic dissemination risk and prometastatic effects of chemotherapy. NPJ Breast Cancer 2022; 8:101. [PMID: 36056005 PMCID: PMC9440218 DOI: 10.1038/s41523-022-00463-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
Metastatic dissemination in breast cancer is regulated by specialized intravasation sites called “tumor microenvironment of metastasis” (TMEM) doorways, composed of a tumor cell expressing the actin-regulatory protein Mena, a perivascular macrophage, and an endothelial cell, all in stable physical contact. High TMEM doorway number is associated with an increased risk of distant metastasis in human breast cancer and mouse models of breast carcinoma. Here, we developed a novel magnetic resonance imaging (MRI) methodology, called TMEM Activity-MRI, to detect TMEM-associated vascular openings that serve as the portal of entry for cancer cell intravasation and metastatic dissemination. We demonstrate that TMEM Activity-MRI correlates with primary tumor TMEM doorway counts in both breast cancer patients and mouse models, including MMTV-PyMT and patient-derived xenograft models. In addition, TMEM Activity-MRI is reduced in mouse models upon treatment with rebastinib, a specific and potent TMEM doorway inhibitor. TMEM Activity-MRI is an assay that specifically measures TMEM-associated vascular opening (TAVO) events in the tumor microenvironment, and as such, can be utilized in mechanistic studies investigating molecular pathways of cancer cell dissemination and metastasis. Finally, we demonstrate that TMEM Activity-MRI increases upon treatment with paclitaxel in mouse models, consistent with prior observations that chemotherapy enhances TMEM doorway assembly and activity in human breast cancer. Our findings suggest that TMEM Activity-MRI is a promising precision medicine tool for localized breast cancer that could be used as a non-invasive test to determine metastatic risk and serve as an intermediate pharmacodynamic biomarker to monitor therapeutic response to agents that block TMEM doorway-mediated dissemination.
Collapse
|
41
|
Athira K, Gopakumar G. Breast cancer stage prediction: a computational approach guided by transcriptome analysis. Mol Genet Genomics 2022; 297:1467-1479. [PMID: 35922530 DOI: 10.1007/s00438-022-01932-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/17/2022] [Indexed: 11/25/2022]
Abstract
Breast cancer is the second leading cancer among women in terms of mortality rate. In recent years, its incidence frequency has been continuously rising across the globe. In this context, the new therapeutic strategies to manage the deadly disease attracts tremendous research focus. However, finding new prognostic predictors to refine the selection of therapy for the various stages of breast cancer is an unattempted issue. Aberrant expression of genes at various stages of cancer progression can be studied to identify specific genes that play a critical role in cancer staging. Moreover, while many schemes for subtype prediction in breast cancer have been explored in the literature, stage-wise classification remains a challenge. These observations motivated the proposed two-phased method: stage-specific gene signature selection and stage classification. In the first phase, meta-analysis of gene expression data is conducted to identify stage-wise biomarkers that were then used in the second phase of cancer classification. From the analysis, 118, 12 and 4 genes respectively in stage I, stage II and stage III are determined as potential biomarkers. Pathway enrichment, gene network and literature analysis validate the significance of the identified genes in breast cancer. In this study, machine learning methods were combined with principal component and posterior probability analysis. Such a scheme offers a unique opportunity to build a meaningful model for predicting breast cancer staging. Among the machine learning models compared, Support Vector Machine (SVM) is found to perform the best for the selected datasets with an accuracy of 92.21% during test data evaluation. Perhaps, biomarker identification performed here for stage-specific cancer treatment would be a meaningful step towards predictive medicine. Significantly, the determination of correct cancer stage using the proposed 134 gene signature set can possibly act as potential target for breast cancer therapeutics.
Collapse
Affiliation(s)
- K Athira
- Department of Computer Science and Engineering, NIT Campus PO, National Institute of Technology Calicut, Calicut, Kerala, India.
| | - G Gopakumar
- Department of Computer Science and Engineering, NIT Campus PO, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
42
|
Bhardwaj A, Josse C, Van Daele D, Poulet C, Chavez M, Struman I, Van Steen K. Deeper insights into long-term survival heterogeneity of pancreatic ductal adenocarcinoma (PDAC) patients using integrative individual- and group-level transcriptome network analyses. Sci Rep 2022; 12:11027. [PMID: 35773268 PMCID: PMC9247075 DOI: 10.1038/s41598-022-14592-1] [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: 07/09/2021] [Accepted: 06/09/2022] [Indexed: 11/22/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is categorized as the leading cause of cancer mortality worldwide. However, its predictive markers for long-term survival are not well known. It is interesting to delineate individual-specific perturbed genes when comparing long-term (LT) and short-term (ST) PDAC survivors and integrate individual- and group-based transcriptome profiling. Using a discovery cohort of 19 PDAC patients from CHU-Liège (Belgium), we first performed differential gene expression analysis comparing LT to ST survivor. Second, we adopted systems biology approaches to obtain clinically relevant gene modules. Third, we created individual-specific perturbation profiles. Furthermore, we used Degree-Aware disease gene prioritizing (DADA) method to develop PDAC disease modules; Network-based Integration of Multi-omics Data (NetICS) to integrate group-based and individual-specific perturbed genes in relation to PDAC LT survival. We identified 173 differentially expressed genes (DEGs) in ST and LT survivors and five modules (including 38 DEGs) showing associations to clinical traits. Validation of DEGs in the molecular lab suggested a role of REG4 and TSPAN8 in PDAC survival. Via NetICS and DADA, we identified various known oncogenes such as CUL1 and TGFB1. Our proposed analytic workflow shows the advantages of combining clinical and omics data as well as individual- and group-level transcriptome profiling.
Collapse
Affiliation(s)
- Archana Bhardwaj
- GIGA-R Centre, BIO3 - Medical Genomics, University of Liège, Avenue de L'Hôpital, 11, 4000, Liège, Belgium.
| | - Claire Josse
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU), Liège, Belgium
- Medical Oncology Department, CHU Liège, Liège, Belgium
| | - Daniel Van Daele
- Department of Gastro-Enterology, University Hospital (CHU), Liège, Belgium
| | - Christophe Poulet
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU), Liège, Belgium
- Laboratory of Rheumatology, GIGA-R, University Hospital (CHULiege), Liège, Belgium
| | - Marcela Chavez
- Department of Medicine, Division of Hematology, University Hospital (CHU), Liège, Belgium
| | - Ingrid Struman
- GIGA-R Centre, Laboratory of Molecular Angiogenesis, University of Liège, Liège, Belgium
| | - Kristel Van Steen
- GIGA-R Centre, BIO3 - Medical Genomics, University of Liège, Avenue de L'Hôpital, 11, 4000, Liège, Belgium
| |
Collapse
|
43
|
Sarhangi N, Hajjari S, Heydari SF, Ganjizadeh M, Rouhollah F, Hasanzad M. Breast cancer in the era of precision medicine. Mol Biol Rep 2022; 49:10023-10037. [PMID: 35733061 DOI: 10.1007/s11033-022-07571-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 01/02/2023]
Abstract
Breast cancer is a heterogeneous disorder with different molecular subtypes and biological characteristics for which there are diverse therapeutic approaches and clinical outcomes specific to any molecular subtype. It is a global health concern due to a lack of efficient therapy regimens that might be used for all disease subtypes. Therefore, treatment customization for each patient depending on molecular characteristics should be considered. Precision medicine for breast cancer is an approach to diagnosis, treatment, and prevention of the disease that takes into consideration the patient's genetic makeup. Precision medicine provides the promise of highly individualized treatment, in which each individual breast cancer patient receives the most appropriate diagnostics and targeted therapies based on the genetic profile of cancer. The knowledge about the molecular features and development of breast cancer treatment approaches has increased, which led to the development of new targeted therapeutics. Tumor genomic profiling is the standard of care for breast cancer that could contribute to taking steps to better management of malignancies. It holds great promise for accurate prognostication, prediction of response to common systemic therapies, and individualized monitoring of the disease. The emergence of targeted treatment has significantly enhanced the survival of patients with breast cancer and contributed to reducing the economic costs of the health system. In this review, we summarized the therapeutic approaches associated with the molecular classification of breast cancer to help the best treatment selection specific to the target patient.
Collapse
Affiliation(s)
- Negar Sarhangi
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrzad Hajjari
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyede Fatemeh Heydari
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Ganjizadeh
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rouhollah
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
44
|
Thompson KJ, Leon-Ferre RA, Sinnwell JP, Zahrieh D, Suman V, Metzger F, Asad S, Stover D, Carey L, Sikov W, Ingle J, Liu M, Carter J, Klee E, Weinshilboum R, Boughey J, Wang L, Couch F, Goetz M, Kalari K. Luminal androgen receptor breast cancer subtype and investigation of the microenvironment and neoadjuvant chemotherapy response. NAR Cancer 2022; 4:zcac018. [PMID: 35734391 PMCID: PMC9204893 DOI: 10.1093/narcan/zcac018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/28/2022] [Accepted: 06/13/2022] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with low overall survival rates and high molecular heterogeneity; therefore, few targeted therapies are available. The luminal androgen receptor (LAR) is the most consistently identified TNBC subtype, but the clinical utility has yet to be established. Here, we constructed a novel genomic classifier, LAR-Sig, that distinguishes the LAR subtype from other TNBC subtypes and provide evidence that it is a clinically distinct disease. A meta-analysis of seven TNBC datasets (n = 1086 samples) from neoadjuvant clinical trials demonstrated that LAR patients have significantly reduced response (pCR) rates than non-LAR TNBC patients (odds ratio = 2.11, 95% CI: 1.33, 2.89). Moreover, deconvolution of the tumor microenvironment confirmed an enrichment of luminal epithelium corresponding with a decrease in basal and myoepithelium in LAR TNBC tumors. Increased immunosuppression in LAR patients may lead to a decreased presence of cycling T-cells and plasma cells. While, an increased presence of myofibroblast-like cancer-associated cells may impede drug delivery and treatment. In summary, the lower levels of tumor infiltrating lymphocytes (TILs), reduced immune activity in the micro-environment, and lower pCR rates after NAC, suggest that new therapeutic strategies for the LAR TNBC subtype need to be developed.
Collapse
Affiliation(s)
- Kevin J Thompson
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, USA
| | | | - Jason P Sinnwell
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, USA
| | - David M Zahrieh
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, USA
| | - Vera J Suman
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, USA
| | | | - Sarah Asad
- The Ohio State University Wexner Medical Center, Molecular, Cellular, and Developmental Biology, Columbus, OH, USA
| | - Daniel G Stover
- The Ohio State University Wexner Medical Center, Molecular, Cellular, and Developmental Biology, Columbus, OH, USA
| | - Lisa Carey
- University of North Carolina at Chapel Hill School of Medicine, Medical Science, Chapel Hill, NC, USA
| | - William M Sikov
- Warren Alpert Medical School of Brown University, Department of Medicine Women, Providence, RI, USA
- Infants Hospital of Rhode Island, Department of Obstetrics & Gynecology, Providence, RI, USA
| | - James N Ingle
- Mayo Clinic, Department of Oncology, Rochester, MN, USA
| | - Minetta C Liu
- Mayo Clinic, Department of Oncology, Rochester, MN, USA
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Jodi M Carter
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Eric W Klee
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, USA
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Richard M Weinshilboum
- Mayo Clinic, Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA
| | | | - Liewei Wang
- Mayo Clinic, Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA
| | - Fergus J Couch
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Matthew P Goetz
- Mayo Clinic, Department of Oncology, Rochester, MN, USA
- Mayo Clinic, Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN, USA
| | - Krishna R Kalari
- Mayo Clinic, Department of Quantitative Health Sciences, Rochester, MN, USA
| |
Collapse
|
45
|
Bergholtz H, Lien T, Lingaas F, Sørlie T. Comparative analysis of the molecular subtype landscape in canine and human mammary gland tumors. J Mammary Gland Biol Neoplasia 2022; 27:171-183. [PMID: 35932380 PMCID: PMC9433360 DOI: 10.1007/s10911-022-09523-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
Breast cancers in humans belong to one of several intrinsic molecular subtypes each with different tumor biology and different clinical impact. Mammary gland tumors in dogs are proposed as a relevant comparative model for human breast cancer; however, it is still unclear whether the intrinsic molecular subtypes have the same significance in dogs and humans. Using publicly available data, we analyzed gene expression and whole-exome sequencing data from 158 canine mammary gland tumors. We performed molecular subtyping using the PAM50 method followed by subtype-specific comparisons of gene expression characteristics, mutation patterns and copy number profiles between canine tumors and human breast tumors from The Cancer Genome Atlas (TCGA) breast cancer cohort (n = 1097). We found that luminal A canine tumors greatly resemble luminal A human tumors both in gene expression characteristics, mutations and copy number profiles. Also, the basal-like canine and human tumors were relatively similar, with low expression of luminal epithelial markers and high expression of genes involved in cell proliferation. There were, however, distinct differences in immune-related gene expression patterns in basal-like tumors between the two species. Characteristic HER2-enriched and luminal B subtypes were not present in the canine cohort, and we found no tumors with high-level ERBB2 amplifications. Benign and malignant canine tumors displayed similar PAM50 subtype characteristics. Our findings indicate that deeper understanding of the different molecular subtypes in canine mammary gland tumors will further improve the value of canines as comparative models for human breast cancer.
Collapse
Affiliation(s)
- Helga Bergholtz
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Tonje Lien
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Frode Lingaas
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Therese Sørlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
46
|
Singhal SK, Byun JS, Yan T, Yancey R, Caban A, Gil Hernandez S, Bufford S, Hewitt SM, Winfield J, Pradhan JS, Mustkov V, McDonald JA, Pérez-Stable EJ, Napoles AM, Vohra N, De Siervi A, Yates C, Davis MB, Yang M, Tsai YC, Weissman AM, Gardner K. Protein expression of the gp78 E3-ligase predicts poor breast cancer outcome based on race. JCI Insight 2022; 7:157465. [PMID: 35639484 PMCID: PMC9310521 DOI: 10.1172/jci.insight.157465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Women of African ancestry suffer higher rates of breast cancer mortality compared to all other groups in the United States. Though the precise reasons for these disparities remain unclear, many recent studies have implicated a role for differences in tumor biology. Using an epitope-validated antibody against the endoplasmic reticulum-associated degradation (ERAD) E3 ubiquitin ligase, gp78, we show that elevated levels of gp78 in patient breast cancer cells predict poor survival. Moreover, high levels of gp78 are associated with poor outcomes in both ER-positive and ER-negative tumors, and breast cancers expressing elevated amounts of gp78 protein are enriched in gene expression pathways that influence cell cycle, metabolism, receptor-mediated signaling, and cell stress response pathways. In multivariate analysis adjusted for subtype and grade, gp78 protein is an independent predictor of poor outcomes in women of African ancestry. Furthermore, gene expression signatures, derived from patients stratified by gp78 protein expression, are strong predictors of recurrence and pathological complete response in retrospective clinical trial data and share many common features with gene sets previously identified to be overrepresented in breast cancers based on race. These findings implicate a prominent role for gp78 in tumor progression and offer new insights into our understanding of racial differences in breast cancer outcomes.
Collapse
Affiliation(s)
- Sandeep K Singhal
- Department of Pathology, University of North Dakota, Grand Forks, United States of America
| | - Jung S Byun
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Tingfen Yan
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Ryan Yancey
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Ambar Caban
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Sara Gil Hernandez
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Sediqua Bufford
- Masters of Science Biotechnology, Morehouse School of Medicine, Atlanta, United States of America
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, United States of America
| | - Joy Winfield
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Jaya Sarin Pradhan
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Vesco Mustkov
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| | - Jasmine A McDonald
- Department of Epidemiology, Columbia University Medical Center, New York, United States of America
| | - Eliseo J Pérez-Stable
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Anna Maria Napoles
- Intramural Research Program, National Institutes of Minority Health and Health Disparities, Bethesda, United States of America
| | - Nasreen Vohra
- Brody School of Medicine, East Carolina University, Greenville, United States of America
| | - Adriana De Siervi
- Directora del Laboratorio de Oncología Molecular y Nuevos Blancos Terapéut, CONICET, Buenos Aiers, Argentina
| | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, United States of America
| | - Melissa B Davis
- Department of Surgery (Breast Surgery & Oncology), Weill Cornell Medicine, New York, United States of America
| | - Mei Yang
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, Frederick, United States of America
| | - Yien Che Tsai
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, Frederick, United States of America
| | - Allan M Weissman
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, Frederick, United States of America
| | - Kevin Gardner
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, United States of America
| |
Collapse
|
47
|
Weitz P, Wang Y, Kartasalo K, Egevad L, Lindberg J, Grönberg H, Eklund M, Rantalainen M. Transcriptome-wide prediction of prostate cancer gene expression from histopathology images using co-expression based convolutional neural networks. Bioinformatics 2022; 38:3462-3469. [PMID: 35595235 PMCID: PMC9237721 DOI: 10.1093/bioinformatics/btac343] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 03/18/2022] [Accepted: 05/16/2022] [Indexed: 12/15/2022] Open
Abstract
Motivation Molecular phenotyping by gene expression profiling is central in contemporary cancer research and in molecular diagnostics but remains resource intense to implement. Changes in gene expression occurring in tumours cause morphological changes in tissue, which can be observed on the microscopic level. The relationship between morphological patterns and some of the molecular phenotypes can be exploited to predict molecular phenotypes from routine haematoxylin and eosin-stained whole slide images (WSIs) using convolutional neural networks (CNNs). In this study, we propose a new, computationally efficient approach to model relationships between morphology and gene expression. Results We conducted the first transcriptome-wide analysis in prostate cancer, using CNNs to predict bulk RNA-sequencing estimates from WSIs for 370 patients from the TCGA PRAD study. Out of 15 586 protein coding transcripts, 6618 had predicted expression significantly associated with RNA-seq estimates (FDR-adjusted P-value <1×10−4) in a cross-validation and 5419 (81.9%) of these associations were subsequently validated in a held-out test set. We furthermore predicted the prognostic cell-cycle progression score directly from WSIs. These findings suggest that contemporary computer vision models offer an inexpensive and scalable solution for prediction of gene expression phenotypes directly from WSIs, providing opportunity for cost-effective large-scale research studies and molecular diagnostics. Availability and implementation A self-contained example is available from http://github.com/phiwei/prostate_coexpression. Model predictions and metrics are available from doi.org/10.5281/zenodo.4739097. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Philippe Weitz
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Yinxi Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Kimmo Kartasalo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden.,Faculty of Medicine and Health Technology, Tampere University, Tampere, 33100, Finland
| | - Lars Egevad
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden.,Science for Life Laboratory, Stockholm, 17177, Sweden
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| |
Collapse
|
48
|
Association between tumor 18F-fluorodeoxyglucose metabolism and survival in women with estrogen receptor-positive, HER2-negative breast cancer. Sci Rep 2022; 12:7858. [PMID: 35552460 PMCID: PMC9098458 DOI: 10.1038/s41598-022-11603-z] [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: 12/02/2021] [Accepted: 04/26/2022] [Indexed: 11/08/2022] Open
Abstract
We examined whether 18F-fluorodeoxyglucose metabolism is associated with distant relapse-free survival (DRFS) and overall survival (OS) in women with estrogen receptor (ER)-positive, HER2-negative breast cancer. This was a cohort study examining the risk factors for survival that had occurred at the start of the study. A cohort from Asan Medical Center, Korea, recruited between November 2007 and December 2014, was included. Patients received anthracycline-based neoadjuvant chemotherapy. The maximum standardized uptake value (SUV) of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (PET/CT) was measured. The analysis included 466 women. The median (interquartile range) follow-up period without distant metastasis or death was 6.2 (5.3-7.6) years. Multivariable analysis of hazard ratio (95% confidence interval [CI]) showed that the middle and high tertiles of SUV were prognostic for DRFS (2.93, 95% CI 1.62-5.30; P < 0.001) and OS (4.87, 95% CI 1.94-12.26; P < 0.001). The 8-year DRFS rates were 90.7% (95% CI 85.5-96.1%) for those in the low tertile of maximum SUV vs. 73.7% (95% CI 68.0-79.8%) for those in the middle and high tertiles of maximum SUV. 18F-fluorodeoxyglucose PET/CT may assess the risk of distant metastasis and death in ER-positive, HER2-negative patients.
Collapse
|
49
|
Jafarinejad-Farsangi S, Moazzam-Jazi M, Naderi Ghale-Noie Z, Askari N, Miri Karam Z, Mollazadeh S, Hadizadeh M. Investigation of genes and pathways involved in breast cancer subtypes through gene expression meta-analysis. Gene X 2022; 821:146328. [PMID: 35181505 DOI: 10.1016/j.gene.2022.146328] [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: 10/10/2021] [Revised: 01/16/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Molecular-based studies have revealed heterogeneity in Breast cancer BC while also improving classification and treatment. However, efforts are underway to distinguish between distinct subtypes of breast cancer. In this study, the results of several microarray studies were combined to identify genes and pathways specific to each BC subtype. METHODS Meta-analysis of multiple gene expression profile datasets was screened to find differentially expressed genes (DEGs) across subtypes of BC and normal breast tissue samples. Protein-protein interaction network and gene set enrichment analysis were used to identify critical genes and pathways associated with BC subtypes. The differentially expressed genes from meta-analysis was validated using an independent comprehensive breast cancer RNA-sequencing dataset obtained from the Cancer Genome Atlas (TCGA). RESULTS We identified 110 DEGs (13 DEGs in all and 97 DEGs in each subtype) across subtypes of BC. All subtypes had a small set of shared DEGs enriched in the Chemokine receptor bind chemokine pathway. Luminal A specific were enriched in the translational elongation process in mitochondria, and the enhanced process in luminal B subtypes was interferon-alpha/beta signaling. Cell cycle and mitotic DEGs were enriched in the basal-like group. All subtype-specific DEG genes (100%) were successfully validated for Luminal A, Luminal B, ERBB2, and Normal-like. However, the validation percentage for Basal-like group was 77.8%. CONCLUSION Integrating researches such as a meta-analysis of gene expression might be more effective in uncovering subtype-specific DEGs and pathways than a single-study analysis. It would be more beneficial to increase the number of studies that use matched BC subtypes along with GEO profiling approaches to reach a better result regarding DEGs and reduce probable biases. However, achieving 77.8% overlap in basal-specific genes and complete concordance in specific genes related to other subtypes can implicate the strength of our analysis for discovering the subtype-specific genes.
Collapse
Affiliation(s)
- Saeideh Jafarinejad-Farsangi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Maryam Moazzam-Jazi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zari Naderi Ghale-Noie
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Nahid Askari
- Department of Biotechnology, Institute of Sciences and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran.
| | - Zahra Miri Karam
- Student Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Morteza Hadizadeh
- Student Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
50
|
Decision Theory versus Conventional Statistics for Personalized Therapy of Breast Cancer. J Pers Med 2022; 12:jpm12040570. [PMID: 35455687 PMCID: PMC9028435 DOI: 10.3390/jpm12040570] [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: 02/17/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022] Open
Abstract
Estrogen and progesterone receptors being present or not represents one of the most important biomarkers for therapy selection in breast cancer patients. Conventional measurement by immunohistochemistry (IHC) involves errors, and numerous attempts have been made to increase precision by additional information from gene expression. This raises the question of how to fuse information, in particular, if there is disagreement. It is the primary domain of Dempster–Shafer decision theory (DST) to deal with contradicting evidence on the same item (here: receptor status), obtained through different techniques. DST is widely used in technical settings, such as self-driving cars and aviation, and is also promising to deliver significant advantages in medicine. Using data from breast cancer patients already presented in previous work, we focus on comparing DST with classical statistics in this work, to pave the way for its application in medicine. First, we explain how DST not only considers probabilities (a single number per sample), but also incorporates uncertainty in a concept of ‘evidence’ (two numbers per sample). This allows for very powerful displays of patient data in so-called ternary plots, a novel and crucial advantage for medical interpretation. Results are obtained according to conventional statistics (ODDS) and, in parallel, according to DST. Agreement and differences are evaluated, and the particular merits of DST discussed. The presented application demonstrates how decision theory introduces new levels of confidence in diagnoses derived from medical data.
Collapse
|