1
|
Schalck A, Tran T, Li J, Sei E, Bai S, Hu M, Lin J, Bright SJ, Reddick S, Yang F, Batra H, Contreras A, Raso MG, Stauder MC, Hoffman KE, Reddy JP, Nead KT, Smith BD, Sawakuchi GO, Woodward WA, Watowich SS, Litton JK, Bedrosian I, Mittendorf EA, Le-Petross H, Navin NE, Shaitelman SF. The impact of breast radiotherapy on the tumor genome and immune ecosystem. Cell Rep 2025; 44:115703. [PMID: 40378044 DOI: 10.1016/j.celrep.2025.115703] [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: 05/02/2024] [Revised: 02/17/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
Radiotherapy is a pillar of breast cancer treatment; however, it remains unclear how radiotherapy modulates the tumor microenvironment. We investigated this question in a cohort of 20 patients with estrogen-receptor positive (ER+) breast tumors who received neoadjuvant radiotherapy. Tumor biopsies were collected before and 7 days postradiation. Single-cell DNA sequencing (scDNA-seq) and scRNA-seq were conducted on 8 and 11 patients, respectively, at these two time points. The scRNA data showed increased infiltration of naive-like CD4 T cells and an early, activated CD8 T cell population following radiotherapy. Radiotherapy also eliminated existing cytotoxic T cells and resulted in myeloid cell increases. In tumor cells, the scDNA-seq data showed a high genomic selection of subclones in half of the patients with high ER expression, while the remaining number had low genomic selection and an interferon response. Collectively, these data provide insight into the impact of radiotherapy in ER+ breast cancer patients.
Collapse
Affiliation(s)
- Aislyn Schalck
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biological Sciences, University of Texas, Houston, TX 770303, USA
| | - Tuan Tran
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianzhuo Li
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emi Sei
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shanshan Bai
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Hu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jerome Lin
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott J Bright
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samuel Reddick
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Fei Yang
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Janssen China Research & Development, Johnson&Johnson, Shanghai 201210, China
| | - Harsh Batra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alejandro Contreras
- Department of Anatomical Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael C Stauder
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Karen E Hoffman
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jay P Reddy
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kevin T Nead
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Benjamin D Smith
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel O Sawakuchi
- Graduate School of Biological Sciences, University of Texas, Houston, TX 770303, USA; Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wendy A Woodward
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Huong Le-Petross
- Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas E Navin
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biological Sciences, University of Texas, Houston, TX 770303, USA; Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Simona F Shaitelman
- Department of Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Pabba A, Zels G, De Schepper M, Geukens T, Van Baelen K, Maetens M, Leduc S, Nguyen HL, Mahdami A, Van Cauwenberge J, Borremans K, Izci H, Hatse S, Neven P, Wildiers H, Biganzoli E, Van Den Bogaert W, Richard F, Floris G, Desmedt C. Stromal Tumor-Infiltrating Lymphocytes in Hormone Receptor-Positive/HER2 Negative Metastatic Breast Cancer. Mod Pathol 2025; 38:100650. [PMID: 39522641 DOI: 10.1016/j.modpat.2024.100650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The immune landscape of hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer (HR+/HER2- mBC), the most common subtype of BC, remains understudied. This is mainly because of reduced sample acquisition opportunities from metastases as compared with primary tumors. In this study, we explored stromal tumor-infiltrating lymphocytes (sTIL) in metastatic samples collected through our post-mortem tissue donation program UZ/KU Leuven Post-mortem Tissue Donation program to Enhance Research (NCT04531696). sTIL were scored as a continuous parameter according to the international guidelines on 427 metastases and 38 primary untreated tumors acquired from 20 patients with HR+/HER2- mBC. Estrogen receptor (ER) status was evaluated on 362 metastases with a cutoff value for positivity set at 1% according to the American Society of Clinical Oncology/College of American Pathologists guidelines. Our analyses show that 54% and 15% of metastases had sTIL levels of ≥1% and ≥5%, respectively. sTIL levels tended to be lower in metastases as compared with their respective primary tumors (estimate, -2.83; 95% CI, -5.77 to 0.11; P = .07). sTIL levels were lower in metastases from invasive lobular carcinoma than in metastases from invasive breast carcinoma of no special type (estimate, -1.67; 95% CI, -2.35 to -0.98; P < .001). A loss of ER expression was observed in 14% of all metastases, yet a negative ER status was not significantly associated with increased sTIL levels. Finally, sTIL levels were significantly higher in lung and axillary lymph node metastases compared with all metastases. Although these analyses were conducted on multiple metastases obtained at the end of life after several lines of treatment, the data provide novel and valuable insights into the state of immune infiltration in patients with HR+/HER2- mBC.
Collapse
Affiliation(s)
- Anirudh Pabba
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Gitte Zels
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Pathology, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Maxim De Schepper
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Pathology, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Tatjana Geukens
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Oncology, General Medical Oncology Unit, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Karen Van Baelen
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Oncology, Gynecological Oncology Unit, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Marion Maetens
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sophia Leduc
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ha-Linh Nguyen
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Amena Mahdami
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Josephine Van Cauwenberge
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Oncology, Gynecological Oncology Unit, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Kristien Borremans
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Oncology, Gynecological Oncology Unit, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Hava Izci
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sigrid Hatse
- Department of Oncology, Laboratory of Experimental Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Oncology, Gynecological Oncology Unit, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Hans Wildiers
- Department of Oncology, General Medical Oncology Unit, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | - Elia Biganzoli
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Biomedical and Clinical Sciences, Unit of Medical Statistics, Biometry and Epidemiology, Ospedale "L. Sacco" LITA campus-Università degli Studi di Milano, Milan, Italy; Data Science and Research Center, Ospedale "L. Sacco" LITA campus-Università degli Studi di Milano, Milan, Italy
| | | | - François Richard
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Giuseppe Floris
- Department of Pathology, Universitair Ziekenhuis Leuven, Leuven, Belgium; Department of Imaging and Pathology, Laboratory for Translational Cell and Tissue Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Christine Desmedt
- Department of Oncology, Laboratory for Translational Breast Cancer Research, Katholieke Universiteit Leuven, Leuven, Belgium.
| |
Collapse
|
3
|
Merloni F, Palleschi M, Gianni C, Sirico M, Serra R, Casadei C, Sarti S, Cecconetto L, Di Menna G, Mariotti M, Maltoni R, Montanari D, Romeo A, De Giorgi U. Local treatment for oligoprogressive metastatic sites of breast cancer: efficacy, toxicities and future perspectives. Clin Exp Metastasis 2024; 41:863-875. [PMID: 39312051 PMCID: PMC11606987 DOI: 10.1007/s10585-024-10312-3] [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: 07/09/2024] [Accepted: 09/08/2024] [Indexed: 11/05/2024]
Abstract
Metastatic breast cancer (MBC) is still an incurable disease, which eventually develops resistance mechanisms against systemic therapies. While most patients experience widespread disease progression during systemic treatment (ST), in some cases, progression may occur at a limited number of metastatic sites. Evidence from other malignancies suggests that local treatment with stereotactic ablative radiotherapy (SABR) of oligoprogressive disease (OPD) may allow effective disease control without the need to modify ST. Available evidence regarding local treatment of oligoprogressive breast cancer is limited, mostly consisting of retrospective studies. The only randomized data come from the randomized CURB trial, which enrolled patients with oligoprogressive disease, including both small cell lung cancer and breast cancer patients, and did not show a survival benefit from local treatment in the latter group. However, local treatment of oligoprogressive MBC is still considered in clinical practice, especially to delay the switch to more toxic STs. This review aims to identify patients who may benefit from this approach based on the current available knowledge, focusing also on the potential risks associated with the combination of radiotherapy (RT) and ST, as well as on possible future scenarios.
Collapse
Affiliation(s)
- Filippo Merloni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy.
| | - Michela Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Riccardo Serra
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Samanta Sarti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Lorenzo Cecconetto
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Giandomenico Di Menna
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Marita Mariotti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Roberta Maltoni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Daniela Montanari
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Antonino Romeo
- Radiotherapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via P.Maroncelli 40, 47014, Meldola, Italy
| |
Collapse
|
4
|
Zhang LK, Li Y, Zhai L, Tang Y, Jiao Y, Mei Y, Yang R, You R, Yin L, Ni H, Ge J, Guan YQ. Natural Phycocyanin/Paclitaxel Micelle Delivery of Therapeutic P53 to Activate Apoptosis for HER2 or ER Positive Breast Cancer Therapy. ACS Biomater Sci Eng 2024; 10:6995-7004. [PMID: 39390952 DOI: 10.1021/acsbiomaterials.4c00756] [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: 10/12/2024]
Abstract
The P53 gene is commonly mutated in breast cancer, protein based the gene as anticancer drugs could provide efficient and stable advantages by restoring the function of the wild-type P53 protein. In this study, we describe the creation and utilization of a micelle composed by natural phycocyanin and paclitaxel and grafting anti-HER2 (PPH), which effectively packages and transports recombinant P53 protein with anti-ER (PE), resulting in a new entity designated as PE@PPH, to address localization obstacles and modify cellular tropism to the cell membrane or nucleus. The results indicate that PE@PPH has strong antitumor properties, even at low doses of PTX both in vitro and in vivo. These findings suggest that PE@PPH could be an enhancing micelle for delivering therapeutic proteins and promoting protein functional recovery, particularly in addressing the challenges posed by tumor heterogeneity in breast cancer.
Collapse
Affiliation(s)
- Ling-Kun Zhang
- School of Life Science, South China Normal University, Guangzhou 510631, China
- School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Yuan Li
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Limin Zhai
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yunzhi Tang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yuxuan Jiao
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yitong Mei
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Runcai Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Rong You
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - He Ni
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou 510631, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
5
|
Papalexis P, Georgakopoulou VE, Drossos PV, Thymara E, Nonni A, Lazaris AC, Zografos GC, Spandidos DA, Kavantzas N, Thomopoulou GE. Precision medicine in breast cancer (Review). Mol Clin Oncol 2024; 21:78. [PMID: 39246849 PMCID: PMC11375768 DOI: 10.3892/mco.2024.2776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Precision medicine in breast cancer is a revolutionary approach that customizes diagnosis and treatment based on individual and tumor characteristics, departing from the traditional one-size-fits-all approach. Breast cancer is diverse, with various subtypes driven by distinct genetic mutations. Understanding this diversity is crucial for tailored treatment strategies that target specific vulnerabilities in each tumor. Genetic testing, particularly for mutations in breast cancer gene (BRCA) DNA repair-associated genes, helps assess hereditary risks and influences treatment decisions. Molecular subtyping guides personalized treatments, such as hormonal therapies for receptor-positive tumors and human epidermal growth factor receptor 2 (HER2)-targeted treatments. Targeted therapies, including those for HER2-positive and hormone receptor-positive breast cancers, offer more effective and precise interventions. Immunotherapy, especially checkpoint inhibitors, shows promise, particularly in certain subtypes such as triple-negative breast cancer, with ongoing research aiming to broaden its effectiveness. Integration of big data and artificial intelligence enhances personalized treatment strategies, while liquid biopsies provide real-time insights into tumor dynamics, aiding in treatment monitoring and modification. Challenges persist, including accessibility and tumor complexity, but emerging technologies and precision prevention offer hope for improved outcomes. Ultimately, precision medicine aims to optimize treatment efficacy, minimize adverse effects and enhance the quality of life for patients with breast cancer.
Collapse
Affiliation(s)
- Petros Papalexis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | | | - Panagiotis V Drossos
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | - Eirini Thymara
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Aphrodite Nonni
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Andreas C Lazaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George C Zografos
- Department of Propedeutic Surgery, Hippokration Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Nikolaos Kavantzas
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgia Eleni Thomopoulou
- Cytopathology Department, 'Attikon' University General Hospital, School of Medicine, National and Kapodistrian University of Athens, 12461 Athens, Greece
| |
Collapse
|
6
|
Wang TC, Sawhney S, Morgan D, Bennett RL, Rashmi R, Estecio MR, Brock A, Singh I, Baer CF, Licht JD, Lele TP. Genetic variation drives cancer cell adaptation to ECM stiffness. Proc Natl Acad Sci U S A 2024; 121:e2403062121. [PMID: 39302966 PMCID: PMC11441511 DOI: 10.1073/pnas.2403062121] [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: 02/14/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
The progression of many solid tumors is accompanied by temporal and spatial changes in the stiffness of the extracellular matrix (ECM). Cancer cells adapt to soft and stiff ECM through mechanisms that are not fully understood. It is well known that there is significant genetic heterogeneity from cell to cell in tumors, but how ECM stiffness as a parameter might interact with that genetic variation is not known. Here, we employed experimental evolution to study the response of genetically variable and clonal populations of tumor cells to variable ECM stiffness. Proliferation rates of genetically variable populations cultured on soft ECM increased over a period of several weeks, whereas clonal populations did not evolve. Tracking of DNA barcoded cell lineages revealed that soft ECM consistently selected for the same few variants. These data provide evidence that ECM stiffness exerts natural selection on genetically variable tumor populations. Soft-selected cells were highly migratory, with enriched oncogenic signatures and unusual behaviors such as spreading and traction force generation on ECMs with stiffness as low as 1 kPa. Rho-regulated cell spreading was found to be the directly selected trait, with yes-associated protein 1 translocation to the nucleus mediating fitness on soft ECM. Overall, these data show that genetic variation can drive cancer cell adaptation to ECM stiffness.
Collapse
Affiliation(s)
- Ting-Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX77843
| | - Suchitaa Sawhney
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
| | - Daylin Morgan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX78712
| | - Richard L. Bennett
- Division of Hematology and Oncology, University of Florida Health Cancer Center, Gainesville, FL32610
| | - Richa Rashmi
- Department of Cell Biology and Genetics, Texas A&M University, Bryan, TX77807
| | - Marcos R. Estecio
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX78712
| | - Irtisha Singh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
- Department of Cell Biology and Genetics, Texas A&M University, Bryan, TX77807
| | - Charles F. Baer
- Department of Biology, University of Florida, Gainesville, FL32611
| | - Jonathan D. Licht
- Division of Hematology and Oncology, University of Florida Health Cancer Center, Gainesville, FL32610
| | - Tanmay P. Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX77843
- Department of Biomedical Engineering, Texas A&M University, College Station, TX77843
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX77030
| |
Collapse
|
7
|
Duan N, Hua Y, Yan X, He Y, Zeng T, Gong J, Fu Z, Li W, Yin Y. Unveiling Alterations of Epigenetic Modifications and Chromatin Architecture Leading to Lipid Metabolic Reprogramming during the Evolutionary Trastuzumab Adaptation of HER2-Positive Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309424. [PMID: 38460162 PMCID: PMC11095153 DOI: 10.1002/advs.202309424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/08/2024] [Indexed: 03/11/2024]
Abstract
Secondary trastuzumab resistance represents an evolutionary adaptation of HER2-positive breast cancer during anti-HER2 treatment. Most current studies have tended to prioritize HER2 and its associated signaling pathways, often overlooking broader but seemingly less relevant cellular processes, along with their associated genetic and epigenetic mechanisms. Here, transcriptome data is not only characterized but also examined epigenomic and 3D genome architecture information in both trastuzumab-sensitive and secondary-resistant breast cancer cells. The findings reveal that the global metabolic reprogramming associated with trastuzumab resistance may stem from genome-wide alterations in both histone modifications and chromatin structure. Specifically, the transcriptional activities of key genes involved in lipid metabolism appear to be regulated by variant promoter H3K27me3 and H3K4me3 modifications, as well as promoter-enhancer interactions. These discoveries offer valuable insights into how cancer cells adapt to anti-tumor drugs and have the potential to impact future diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Ningjun Duan
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Yijia Hua
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Xueqi Yan
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Yaozhou He
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Tianyu Zeng
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Jue Gong
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Ziyi Fu
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Wei Li
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| | - Yongmei Yin
- Department of oncologyFirst affiliation hospital of Nanjing medical universityNanjing210029China
| |
Collapse
|
8
|
Li X, Zhu Y, Yi J, Deng Y, Lei B, Ren H. Adoptive cell immunotherapy for breast cancer: harnessing the power of immune cells. J Leukoc Biol 2024; 115:866-881. [PMID: 37949484 DOI: 10.1093/jleuko/qiad144] [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: 08/15/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023] Open
Abstract
Breast cancer is the most prevalent malignant neoplasm worldwide, necessitating the development of novel therapeutic strategies owing to the limitations posed by conventional treatment modalities. Immunotherapy is an innovative approach that has demonstrated significant efficacy in modulating a patient's innate immune system to combat tumor cells. In the era of precision medicine, adoptive immunotherapy for breast cancer has garnered widespread attention as an emerging treatment strategy, primarily encompassing cellular therapies such as tumor-infiltrating lymphocyte therapy, chimeric antigen receptor T/natural killer/M cell therapy, T cell receptor gene-engineered T cell therapy, lymphokine-activated killer cell therapy, cytokine-induced killer cell therapy, natural killer cell therapy, and γδ T cell therapy, among others. This treatment paradigm is based on the principles of immune memory and antigen specificity, involving the collection, processing, and expansion of the patient's immune cells, followed by their reintroduction into the patient's body to activate the immune system and prevent tumor recurrence and metastasis. Currently, multiple clinical trials are assessing the feasibility, effectiveness, and safety of adoptive immunotherapy in breast cancer. However, this therapeutic approach faces challenges associated with tumor heterogeneity, immune evasion, and treatment safety. This review comprehensively summarizes the latest advancements in adoptive immunotherapy for breast cancer and discusses future research directions and prospects, offering valuable guidance and insights into breast cancer immunotherapy.
Collapse
Affiliation(s)
- Xue Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - Yunan Zhu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - Jinfeng Yi
- Department of Pathology, Harbin Medical University, 157 Baojian Road, Harbin 150081, Heilongjiang, China
| | - Yuhan Deng
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - Bo Lei
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| | - He Ren
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150076, Heilongjiang, China
| |
Collapse
|
9
|
Balmaña J, Fasching PA, Couch FJ, Delaloge S, Labidi-Galy I, O'Shaughnessy J, Park YH, Eisen AF, You B, Bourgeois H, Gonçalves A, Kemp Z, Swampillai A, Jankowski T, Sohn JH, Poddubskaya E, Mukhametshina G, Aksoy S, Timcheva CV, Park-Simon TW, Antón-Torres A, John E, Baria K, Gibson I, Gelmon KA. Clinical effectiveness and safety of olaparib in BRCA-mutated, HER2-negative metastatic breast cancer in a real-world setting: final analysis of LUCY. Breast Cancer Res Treat 2024; 204:237-248. [PMID: 38112922 PMCID: PMC10948524 DOI: 10.1007/s10549-023-07165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/23/2023] [Indexed: 12/21/2023]
Abstract
PURPOSE The interim analysis of the phase IIIb LUCY trial demonstrated the clinical effectiveness of olaparib in patients with germline BRCA-mutated (gBRCAm), human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (mBC), with median progression-free survival (PFS) of 8.11 months, which was similar to that in the olaparib arm of the phase III OlympiAD trial (7.03 months). This prespecified analysis provides final overall survival (OS) and safety data. METHODS The open-label, single-arm LUCY trial of olaparib (300 mg, twice daily) enrolled adults with gBRCAm or somatic BRCA-mutated (sBRCAm), HER2-negative mBC. Patients had previously received a taxane or anthracycline for neoadjuvant/adjuvant or metastatic disease and up to two lines of chemotherapy for mBC. RESULTS Of 563 patients screened, 256 (gBRCAm, n = 253; sBRCAm, n = 3) were enrolled. In the gBRCAm cohort, median investigator-assessed PFS (primary endpoint) was 8.18 months and median OS was 24.94 months. Olaparib was clinically effective in all prespecified subgroups: hormone receptor status, previous chemotherapy for mBC, previous platinum-based chemotherapy (including by line of therapy), and previous cyclin-dependent kinase 4/6 inhibitor use. The most frequent treatment-emergent adverse events (TEAEs) were nausea (55.3%) and anemia (39.2%). Few patients (6.3%) discontinued olaparib owing to a TEAE. No deaths associated with AEs occurred during the study treatment or 30-day follow-up. CONCLUSION The LUCY patient population reflects a real-world population in line with the licensed indication of olaparib in mBC. These findings support the clinical effectiveness and safety of olaparib in patients with gBRCAm, HER2-negative mBC. CLINICAL TRIAL REGISTRATION Clinical trials registration number: NCT03286842.
Collapse
Affiliation(s)
- Judith Balmaña
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Suzette Delaloge
- Breast Cancer Unit, Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Intidhar Labidi-Galy
- Department of Oncology, Geneva University Hospital, Department of Medicine, Division of Oncology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology and US Oncology, Dallas, TX, USA
| | - Yeon Hee Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Andrea F Eisen
- Division of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Benoit You
- Department of Medical Oncology, Hospices Civils of Lyon Cancer Institute, Centre for Therapeutic Investigation in Oncology and Haematology of Lyon, Lyon Sud Hospital Centre, Lyon, France
- Faculty of Medicine of Lyon Sud, Claude Bernard Lyon 1 University, Lyon, France
- GINECO-GINEGEPS, Paris, France
| | - Hughes Bourgeois
- Medical Oncology Department, Victor Hugo Clinic-Jean Bernard Center, Le Mans, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
- Cancer Research Center of Marseille, Aix-Marseille University, French National Centre for Scientific Research, National Institute for Health and Medical Research, Marseille, France
| | - Zoe Kemp
- Breast Cancer Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Angela Swampillai
- Department of Clinical Oncology, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
- Breast Cancer Now Research Unit, Guy's Hospital, King's College London, London, UK
| | - Tomasz Jankowski
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Joo Hyuk Sohn
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | - Sercan Aksoy
- Medical Oncology Department, Hacettepe University Cancer Institute, Ankara, Turkey
| | | | | | - Antonio Antón-Torres
- Department of Medical Oncology, Miguel Servet University Hospital and Aragon Health Research Institute, Zaragoza, Spain
| | | | | | | | - Karen A Gelmon
- Department of Medical Oncology, BC Cancer, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
10
|
Li P, Li J, Tong X, Xiao Z, Diao W, Zhong C, Zhou J, Wu W. Global research trends and prospects related to tumor microenvironment within Triple Negative Breast Cancer: a bibliometric analysis. Front Immunol 2023; 14:1261290. [PMID: 38111580 PMCID: PMC10725926 DOI: 10.3389/fimmu.2023.1261290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
Background and aims The tumor microenvironment (TME) has pivotal parts within multiple tumor models of onset/progression, such as triple-negative breast cancer (TNBC). This bibliometric analysis was developed to explore trends and research niches revolving around TME in TNBC. Methods Web of Science Core Collection was queried for identifying studies linked with TME in TNBC, after which the VOSviewer, CiteSpace, and R software programs were used to conduct bibliometric analyses and to generate corresponding visualizations. Results In total, this study included 1,604 studies published from 2005-2023. The USA and China exhibited the highest numbers of citations, and the research institutions with the greatest output in this field included Harvard University, the University of Texas System, and Fudan University. Ying Wang from Sun Yat-Sen University was the most published and most cited author in this space. The highest number of articles were published in Cancer, while the greatest co-citation number was evident in Breast Cancer Research. Important keywords related to this research topic included metastasis, tumor-infiltrating lymphocytes, immunotherapy, chemotherapy, and nanoparticles. In particular, pembrolizumab, immunotherapy, nanoparticles, combination treatment, and biomarkers were topics of marked interest in recent reports. Conclusion The TME in TNBC is an area of rapidly growing and evolving research interest, with extensive global collaboration helping to drive this field forward. Antitumor therapies targeting the TME in TNBC patients represent an emerging topic of future research, providing opportunities for translational findings. The results of this analysis may provide additional guidance for work focused on the TME in TNBC.
Collapse
Affiliation(s)
- Peiting Li
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jun Li
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaofei Tong
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenyang Xiao
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wuliang Diao
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chi Zhong
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jianda Zhou
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wu
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Yu J, Wang H, Shen W, Zhou Y, Cui J, Li H, Gao B. Hsa_circ_0007823 Overexpression Suppresses the Progression of Triple-Negative Breast Cancer via Regulating miR-182-5p-FOXO1 Axis. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:695-708. [PMID: 37873520 PMCID: PMC10590585 DOI: 10.2147/bctt.s417547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023]
Abstract
Background This study aimed to analyze the specific expression of hsa_circ_0007823 in triple-negative breast cancer (TNBC) and explore the roles and related molecular mechanisms of hsa_circ_0007823 in TNBC. Materials and Methods Relative hsa_circ_0007823 levels in TNBC tissues and cell lines were examined by reverse transcription-quantitative polymerase chain reaction. The value of hsa_circ_0007823 levels was evaluated in patients' clinicopathological characteristics and prognostic prediction. A dual-luciferase reporter assay was used to determine the relationship between hsa_circ_0007823, miR-182-5p, and FOXO1. The effect of circ_0007823 overexpression on the growth of TNBC cells was investigated in vitro and in vivo. Results Lower levels of hsa_circ_0007823 were found in TNBC tissues and cell lines and were closely associated with lymph node metastasis, poorer overall and disease-free survival rates. MiR-182-5p was significantly up-regulated, whereas FOXO1 was down-regulated in TNBC cell lines. The miR-182-5p inhibition up-regulated FOXO1 in TNBC cells. Dual-luciferase reporter assays showed that hsa_circ_0007823, miR-182-5p, and FOXO1 interacted with each other. Overexpression of circ_0007823 significantly inhibited the viability, migration, and invasion of TNBC cell lines, but promoted apoptosis. In vivo experiments showed that circ_0007823 overexpression inhibited tumor growth and down-regulated miR-182-5p and up-regulated FOXO1. Conclusion Hsa_circ_0007823 overexpression could suppress the growth, invasion, and migration of TNBC cells, and inhibit tumor growth by regulating miR-182-5p/FOXO1.
Collapse
Affiliation(s)
- Jinling Yu
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, People’s Republic of China
| | - Haofeng Wang
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, People’s Republic of China
| | - Weida Shen
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, People’s Republic of China
| | - Yingzi Zhou
- Department of Pathology, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, People’s Republic of China
| | - Jing Cui
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, People’s Republic of China
| | - Haichuan Li
- Department of Laboratory, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, People’s Republic of China
| | - Beimin Gao
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, People’s Republic of China
| |
Collapse
|
12
|
Andrade JR, Gallagher AD, Maharaj J, McClelland SE. Disentangling the roles of aneuploidy, chromosomal instability and tumour heterogeneity in developing resistance to cancer therapies. Chromosome Res 2023; 31:28. [PMID: 37721639 PMCID: PMC10506951 DOI: 10.1007/s10577-023-09737-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/26/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
Aneuploidy is defined as the cellular state of having a number of chromosomes that deviates from a multiple of the normal haploid chromosome number of a given organism. Aneuploidy can be present in a static state: Down syndrome individuals stably maintain an extra copy of chromosome 21 in their cells. In cancer cells, however, aneuploidy is usually present in combination with chromosomal instability (CIN) which leads to a continual generation of new chromosomal alterations and the development of intratumour heterogeneity (ITH). The prevalence of cells with specific chromosomal alterations is further shaped by evolutionary selection, for example, during the administration of cancer therapies. Aneuploidy, CIN and ITH have each been individually associated with poor prognosis in cancer, and a wealth of evidence suggests they contribute, either alone or in combination, to cancer therapy resistance by providing a reservoir of potential resistant states, or the ability to rapidly evolve resistance. A full understanding of the contribution and interplay between aneuploidy, CIN and ITH is required to tackle therapy resistance in cancer patients. However, these characteristics often co-occur and are intrinsically linked, presenting a major challenge to defining their individual contributions. Moreover, their accurate measurement in both experimental and clinical settings is a technical hurdle. Here, we attempt to deconstruct the contribution of the individual and combined roles of aneuploidy, CIN and ITH to therapy resistance in cancer, and outline emerging approaches to measure and disentangle their roles as a step towards integrating these principles into cancer therapeutic strategy.
Collapse
Affiliation(s)
- Joana Reis Andrade
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M6BQ, England
| | - Annie Dinky Gallagher
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M6BQ, England
| | - Jovanna Maharaj
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M6BQ, England
| | | |
Collapse
|
13
|
Martino F, Lupi M, Giraudo E, Lanzetti L. Breast cancers as ecosystems: a metabolic perspective. Cell Mol Life Sci 2023; 80:244. [PMID: 37561190 PMCID: PMC10415483 DOI: 10.1007/s00018-023-04902-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer and one of the major causes of cancer death. Despite enormous progress in its management, both from the therapeutic and early diagnosis viewpoints, still around 700,000 patients succumb to the disease each year, worldwide. Late recurrency is the major problem in BC, with many patients developing distant metastases several years after the successful eradication of the primary tumor. This is linked to the phenomenon of metastatic dormancy, a still mysterious trait of the natural history of BC, and of several other types of cancer, by which metastatic cells remain dormant for long periods of time before becoming reactivated to initiate the clinical metastatic disease. In recent years, it has become clear that cancers are best understood if studied as ecosystems in which the impact of non-cancer-cell-autonomous events-dependent on complex interaction between the cancer and its environment, both local and systemic-plays a paramount role, probably as significant as the cell-autonomous alterations occurring in the cancer cell. In adopting this perspective, a metabolic vision of the cancer ecosystem is bound to improve our understanding of the natural history of cancer, across space and time. In BC, many metabolic pathways are coopted into the cancer ecosystem, to serve the anabolic and energy demands of the cancer. Their study is shedding new light on the most critical aspect of BC management, of metastatic dissemination, and that of the related phenomenon of dormancy and fostering the application of the knowledge to the development of metabolic therapies.
Collapse
Affiliation(s)
- Flavia Martino
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Mariadomenica Lupi
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Enrico Giraudo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Science and Drug Technology, University of Torino, Turin, Italy
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Turin, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
| |
Collapse
|
14
|
Orrapin S, Thongkumkoon P, Udomruk S, Moonmuang S, Sutthitthasakul S, Yongpitakwattana P, Pruksakorn D, Chaiyawat P. Deciphering the Biology of Circulating Tumor Cells through Single-Cell RNA Sequencing: Implications for Precision Medicine in Cancer. Int J Mol Sci 2023; 24:12337. [PMID: 37569711 PMCID: PMC10418766 DOI: 10.3390/ijms241512337] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Circulating tumor cells (CTCs) hold unique biological characteristics that directly involve them in hematogenous dissemination. Studying CTCs systematically is technically challenging due to their extreme rarity and heterogeneity and the lack of specific markers to specify metastasis-initiating CTCs. With cutting-edge technology, single-cell RNA sequencing (scRNA-seq) provides insights into the biology of metastatic processes driven by CTCs. Transcriptomics analysis of single CTCs can decipher tumor heterogeneity and phenotypic plasticity for exploring promising novel therapeutic targets. The integrated approach provides a perspective on the mechanisms underlying tumor development and interrogates CTCs interactions with other blood cell types, particularly those of the immune system. This review aims to comprehensively describe the current study on CTC transcriptomic analysis through scRNA-seq technology. We emphasize the workflow for scRNA-seq analysis of CTCs, including enrichment, single cell isolation, and bioinformatic tools applied for this purpose. Furthermore, we elucidated the translational knowledge from the transcriptomic profile of individual CTCs and the biology of cancer metastasis for developing effective therapeutics through targeting key pathways in CTCs.
Collapse
Affiliation(s)
- Santhasiri Orrapin
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Patcharawadee Thongkumkoon
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Sasimol Udomruk
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| | - Sutpirat Moonmuang
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Songphon Sutthitthasakul
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Petlada Yongpitakwattana
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
| | - Dumnoensun Pruksakorn
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| | - Parunya Chaiyawat
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.O.); (P.T.); (S.U.); (S.M.); (S.S.); (P.Y.); (D.P.)
- Musculoskeletal Science and Translational Research (MSTR) Center, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand
| |
Collapse
|
15
|
Kluska M, Piastowska-Ciesielska AW, Tokarz P. Cell Cycle Status Influences Resistance to Apoptosis Induced by Oxidative Stress in Human Breast Cancer Cells, Which Is Accompanied by Modulation of Autophagy. Curr Issues Mol Biol 2023; 45:6325-6338. [PMID: 37623218 PMCID: PMC10453102 DOI: 10.3390/cimb45080399] [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: 06/19/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023] Open
Abstract
Cancer cells are characterised by uncontrolled cell proliferation; however, some of them can temporarily arrest their cell cycle at the G0 or G1 phase, which could contribute to tumour heterogeneity and drug resistance. The cell cycle status plays a critical role in chemosensitivity; however, the influence of G0- and G1-arrest has not been elucidated. To study the cell cycle arrest-mediated resistance, we used MCF-7 cells and generated three populations of cells: (1) cells arrested in the G0-like phase, (2) cells that resumed the cell cycle after the G0-like phase and (3) cells arrested in early G1 with a history of G0-like arrest. We observed that both the G0-like- and the G1-arrested cells acquired resistance to apoptosis induced by oxidative stress, accompanied by a decreased intracellular reactive oxygen species and DNA damage. This effect was associated with increased autophagy, likely facilitating their survival at DNA damage insult. The cell cycle reinitiation restored a sensitivity to oxidative stress typical for cells with a non-modulated cell cycle, with a concomitant decrease in autophagy. Our results support the need for further research on the resistance of G0- and G1-arrested cancer cells to DNA-damaging agents and present autophagy as a candidate for targeting in anticancer treatment.
Collapse
Affiliation(s)
- Magdalena Kluska
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | | | - Paulina Tokarz
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
16
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
17
|
Singh B, Sarli VN, Milligan RD, Kinne HE, Shamsnia A, Washburn LJ, Addanki S, Lucci A. Sensitization of Resistant Cells with a BET Bromodomain Inhibitor in a Cell Culture Model of Deep Intrinsic Resistance in Breast Cancer. Cancers (Basel) 2023; 15:cancers15072036. [PMID: 37046697 PMCID: PMC10093448 DOI: 10.3390/cancers15072036] [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: 02/17/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
We treated highly metabolically adaptable (SUM149-MA) triple-negative inflammatory breast cancer cells and their control parental SUM149-Luc cell line with JQ1 for long periods to determine its efficacy at inhibiting therapy-resistant cells. After 20 days of treatment with 1-2 µM of JQ1, which killed majority of cells in the parental cell line, a large number of SUM149-MA cells survived, consistent with their pan-resistant nature. Interestingly, though, the JQ1 treatment sensitized resistant cancer cells in both the SUM149-MA and SUM149-Luc cell lines to subsequent treatment with doxorubicin and paclitaxel. To measure JQ1-mediated sensitization of resistant cancer cells, we first eradicated approximately 99% of relatively chemotherapy-sensitive cancer cells in culture dishes by long treatments with doxorubicin or paclitaxel, and then analyzed the remaining resistant cells for survival and growth into colonies. In addition, combination, rather than sequential, treatment with JQ1 and doxorubicin was also effective in overcoming resistance. Notably, Western blotting showed that JQ1-treated cancer cells had significantly lower levels of PD-L1 protein than did untreated cells, indicating that JQ1 treatment may reduce tumor-mediated immune suppression and improve the response to immunotherapy targeting PD-L1. Finally, JQ1 treatment with a low 62.5 nM dose sensitized another resistant cell line, FC-IBC02-MA, to treatment with doxorubicin and paclitaxel.
Collapse
Affiliation(s)
- Balraj Singh
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vanessa N Sarli
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan D Milligan
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hannah E Kinne
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anna Shamsnia
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Laura J Washburn
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sridevi Addanki
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anthony Lucci
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Doxorubicin and Cisplatin Modulate miR-21, miR-106, miR-126, miR-155 and miR-199 Levels in MCF7, MDA-MB-231 and SK-BR-3 Cells That Makes Them Potential Elements of the DNA-Damaging Drug Treatment Response Monitoring in Breast Cancer Cells—A Preliminary Study. Genes (Basel) 2023; 14:genes14030702. [PMID: 36980974 PMCID: PMC10048428 DOI: 10.3390/genes14030702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
One of the most innovative medical trends is personalized therapy, based on simple and reproducible methods that detect unique features of cancer cells. One of the good prognostic and diagnostic markers may be the miRNA family. Our work aimed to evaluate changes in selected miRNA levels in various breast cancer cell lines (MCF7, MDA-MB-231, SK-BR-3) treated with doxorubicin or cisplatin. The selection was based on literature data regarding the most commonly altered miRNAs in breast cancer (21-3p, 21-5p, 106a-5p, 126-3p, 126-5p, 155-3p, 155-5p, 199b-3p, 199b-5p, 335-3p, 335-5p). qPCR assessment revealed significant differences in the basal levels of some miRNAs in respective cell lines, with the most striking difference in miR-106a-5p, miR-335-5p and miR-335-3p—all of them were lowest in MCF7, while miR-153p was not detected in SK-BR-3. Additionally, different alterations of selected miRNAs were observed depending on the cell line and the drug. However, regardless of these variables, 21-3p/-5p, 106a, 126-3p, 155-3p and 199b-3p miRNAs were shown to respond either to doxorubicin or to cisplatin treatment. These miRNAs seem to be good candidates for markers of breast cancer cell response to doxorubicin or cisplatin. Especially since some earlier reports suggested their role in affecting pathways and expression of genes associated with the DNA-damage response. However, it must be emphasized that the preliminary study shows effects that may be highly related to the applied drug itself and its concentration. Thus, further examination, including human samples, is required.
Collapse
|
19
|
Chu J, Li Y, He M, Zhang H, Yang L, Yang M, Liu J, Cui C, Hong L, Hu X, Zhou L, Li T, Li C, Fan H, Jiang G, Lang T. Zinc finger and SCAN domain containing 1, ZSCAN1, is a novel stemness-related tumor suppressor and transcriptional repressor in breast cancer targeting TAZ. Front Oncol 2023; 13:1041688. [PMID: 36923432 PMCID: PMC10009259 DOI: 10.3389/fonc.2023.1041688] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023] Open
Abstract
INTRODUCTION Cancer stem cells (CSCs) targeted therapy holds the potential for improving cancer management; identification of stemness-related genes in CSCs is necessary for its development. METHODS The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets were used for survival analysis. ZSCAN1 correlated genes was identified by Spearman correlation analysis. Breast cancer stem-like cells (BCSLCs) were isolated by sorting CD44+CD24- cells from suspension cultured breast cancer (BC) spheroids. The sphere-forming capacity and sphere- and tumor-initiating capacities were determined by sphere formation and limiting dilution assays. The relative gene expression was determined by qRT-PCR, western blot. Lentivirus system was used for gene manipulation. Nuclear run-on assay was employed to examine the levels of nascent mRNAs. DNA pull-down and Chromatin immunoprecipitation (ChIP) assays were used for determining the interaction between protein and target DNA fragments. Luciferase reporter assay was used for evaluating the activity of the promoter. RESULTS AND DISCUSSION ZSCAN1 is aberrantly suppressed in BC, and this suppression indicates a bad prognosis. Ectopic expression of ZSCAN1 inhibited the proliferation, clonogenicity, and tumorigenicity of BC cells. ZSCAN1-overexpressing BCSLCs exhibited weakened stemness properties. Normal human mammary epithelial (HMLE) cells with ZSCAN1 depletion exhibited enhanced stemness properties. Mechanistic studies showed that ZSCAN1 directly binds to -951 ~ -925bp region of WWTR1 (encodes TAZ) promoter, inhibits WWTR1 transcription, thereby inhibiting the stemness of BCSCs. Our work thus revealed ZSCAN1 as a novel stemness-related tumor suppressor and transcriptional repressor in BC.
Collapse
Affiliation(s)
- Jian Chu
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yunzhe Li
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Misi He
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Hui Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lingling Yang
- School of Medicine, Chongqing University, Chongqing, China
| | - Muyao Yang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Jingshu Liu
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chenxi Cui
- School of Medicine, Chongqing University, Chongqing, China
| | - Liquan Hong
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xingchi Hu
- Department of General Surgery, Yancheng City No.1 People’s Hospital, Yancheng, Jiangsu, China
| | - Lei Zhou
- School of Optometry, Department of Applied Biology and Chemical Technology, Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
| | - Tangya Li
- Department of General Surgery, Yancheng City No.1 People’s Hospital, Yancheng, Jiangsu, China
| | - Changchun Li
- Department of General Surgery, Yancheng City No.1 People’s Hospital, Yancheng, Jiangsu, China
| | - Huiwen Fan
- Department of General Surgery, Yancheng City No.1 People’s Hospital, Yancheng, Jiangsu, China
| | - Guoqin Jiang
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tingyuan Lang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Zhang Y, Li G, Bian W, Bai Y, He S, Liu Y, Liu H, Liu J. Value of genomics- and radiomics-based machine learning models in the identification of breast cancer molecular subtypes: a systematic review and meta-analysis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1394. [PMID: 36660694 PMCID: PMC9843333 DOI: 10.21037/atm-22-5986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/20/2022] [Indexed: 01/01/2023]
Abstract
Background In the era of precision therapy, early classification of breast cancer (BRCA) molecular subtypes has clinical significance for disease management and prognosis. We explored the accuracy of machine learning (ML) models for early classification of BRCA molecular subtypes through a systematic review of the literature currently available. Methods We retrieved relevant studies published in PubMed, EMBASE, Cochrane, and Web of Science until 15 April 2022. A prediction model risk of bias assessment tool (PROBAST) was applied for the assessment of risk of bias of a genomics-based ML model, and the Radiomics Quality Score (RQS) was simultaneously used to evaluate the quality of this radiomics-based ML model. A random effects model was adopted to analyze the predictive accuracy of genomics-based ML and radiomics-based ML for Luminal A, Luminal B, Basal-like or triple-negative breast cancer (TNBC), and human epidermal growth factor receptor 2 (HER2). The PROSPERO of our study was prospectively registered (CRD42022333611). Results Of the 38 studies were selected for analysis, 14 ML models were based on gene-transcriptomic, with only 4 external validations; and 43 ML models were based on radiomics, with only 14 external validations. Meta-analysis results showed that c-statistic values of the ML based on radiomics for the identification of BRCA molecular subtypes Luminal A, Luminal B, Basal-like or TNBC, and HER2 were 0.76 [95% confidence interval (CI): 0.60-0.96], 0.78 (95% CI: 0.69-0.87), 0.89 (95% CI: 0.83-0.91), and 0.83 (95% CI: 0.81-0.86), respectively. The c-statistic values of ML based on the gene-transcriptomic analysis cohort for the identification of the previously described BRCA molecular subtypes were 0.96 (95% CI: 0.93-0.99), 0.96 (95% CI: 0.93-0.99), 0.98 (95% CI: 0.95-1.00), and 0.97 (95% CI: 0.96-0.98) respectively. Additionally, the sensitivity of the ML model based on radiomics for each molecular subtype ranged from 0.79 to 0.85, while the sensitivity of the ML model based on gene-transcriptomic was between 0.92 and 0.99. Conclusions Both radiomics and gene transcriptomics produced ideal effects on BRCA molecular subtype prediction. Compared with radiomics, gene transcriptomics yielded better prediction results, but radiomics was simpler and more convenient from a clinical point of view.
Collapse
Affiliation(s)
- Yiwen Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Guofeng Li
- Department of Traditional Chinese Medicine Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Wenqing Bian
- Intensive Care Unit, Zibo Maternal and Child Health Hospital, Zibo, China
| | - Yuzhuo Bai
- Department of Traditional Chinese Medicine Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Shuangyan He
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yulian Liu
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| | - Huan Liu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jiaqi Liu
- Department of Breast Thyroid Surgery, Zibo Central Hospital, Zibo, China
| |
Collapse
|
21
|
Lowe L, LaValley JW, Felsher DW. Tackling heterogeneity in treatment-resistant breast cancer using a broad-spectrum therapeutic approach. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:917-925. [PMID: 36627896 PMCID: PMC9771755 DOI: 10.20517/cdr.2022.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/29/2022] [Accepted: 08/02/2022] [Indexed: 06/17/2023]
Abstract
Tumor heterogeneity can contribute to the development of therapeutic resistance in cancer, including advanced breast cancers. The object of the Halifax project was to identify new treatments that would address mechanisms of therapeutic resistance through tumor heterogeneity by uncovering combinations of therapeutics that could target the hallmarks of cancer rather than focusing on individual gene products. A taskforce of 180 cancer researchers, used molecular profiling to highlight key targets responsible for each of the hallmarks of cancer and then find existing therapeutic agents that could be used to reach those targets with limited toxicity. In many cases, natural health products and re-purposed pharmaceuticals were identified as potential agents. Hence, by combining the molecular profiling of tumors with therapeutics that target the hallmark features of cancer, the heterogeneity of advanced-stage breast cancers can be addressed.
Collapse
Affiliation(s)
- Leroy Lowe
- Getting to Know Cancer (NGO), Truro, Nova Scotia B2N 1X5, Canada
| | | | - Dean W. Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, CA CCSR 1105, USA
| |
Collapse
|
22
|
Dankó T, Petővári G, Raffay R, Sztankovics D, Moldvai D, Vetlényi E, Krencz I, Rókusz A, Sipos K, Visnovitz T, Pápay J, Sebestyén A. Characterisation of 3D Bioprinted Human Breast Cancer Model for In Vitro Drug and Metabolic Targeting. Int J Mol Sci 2022; 23:ijms23137444. [PMID: 35806452 PMCID: PMC9267600 DOI: 10.3390/ijms23137444] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023] Open
Abstract
Monolayer cultures, the less standard three-dimensional (3D) culturing systems, and xenografts are the main tools used in current basic and drug development studies of cancer research. The aim of biofabrication is to design and construct a more representative in vivo 3D environment, replacing two-dimensional (2D) cell cultures. Here, we aim to provide a complex comparative analysis of 2D and 3D spheroid culturing, and 3D bioprinted and xenografted breast cancer models. We established a protocol to produce alginate-based hydrogel bioink for 3D bioprinting and the long-term culturing of tumour cells in vitro. Cell proliferation and tumourigenicity were assessed with various tests. Additionally, the results of rapamycin, doxycycline and doxorubicin monotreatments and combinations were also compared. The sensitivity and protein expression profile of 3D bioprinted tissue-mimetic scaffolds showed the highest similarity to the less drug-sensitive xenograft models. Several metabolic protein expressions were examined, and the in situ tissue heterogeneity representing the characteristics of human breast cancers was also verified in 3D bioprinted and cultured tissue-mimetic structures. Our results provide additional steps in the direction of representing in vivo 3D situations in in vitro studies. Future use of these models could help to reduce the number of animal experiments and increase the success rate of clinical phase trials.
Collapse
Affiliation(s)
- Titanilla Dankó
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Gábor Petővári
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Regina Raffay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Enikő Vetlényi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - András Rókusz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Krisztina Sipos
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Tamás Visnovitz
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary;
- Department of Plant Physiology and Molecular Plant Biology, ELTE Eötvös Loránd University, Pázmány Péter Sétány 1/c, 1117 Budapest, Hungary
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (T.D.); (G.P.); (R.R.); (D.S.); (D.M.); (E.V.); (I.K.); (A.R.); (K.S.); (J.P.)
- Correspondence: or
| |
Collapse
|
23
|
Singh B, Sarli VN, Lucci A. Sensitization of Resistant Breast Cancer Cells with a Jumonji Family Histone Demethylase Inhibitor. Cancers (Basel) 2022; 14:cancers14112631. [PMID: 35681611 PMCID: PMC9179491 DOI: 10.3390/cancers14112631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 01/12/2023] Open
Abstract
Simple Summary Using a cell culture model of resistant breast cancer cells with the phenotype that is often responsible for the early relapse of triple-negative breast cancer, namely, the persistence of these cells in reversible quiescence under a variety of challenges, we found that reprogramming the epigenome by treatment with JIB-04, a small-molecule inhibitor of Jumonji-family histone demethylases, sensitized resistant cells. We used this model of deep intrinsic resistance featuring many molecular mechanisms of achieving this phenotype to perform lengthy evaluations of less cytotoxic doses of JIB-04. We found that resistant cells derived from triple-negative inflammatory breast cancer cell lines were either much more sensitive to JIB-04 than the parental cell line or altered by the treatment such that they became sensitive to the chemotherapeutic drugs paclitaxel and doxorubicin. Notably, JIB-04 exposure increased PD-L1 expression in cancer cells, which means that JIB-04 may have clinical applications in improving the responses of triple-negative breast cancer to anti-PD-L1 therapy. Abstract In the present study, we evaluated JIB-04, a small-molecule epigenetic inhibitor initially discovered to inhibit cancer growth, to determine its ability to affect deep intrinsic resistance in a breast cancer model. The model was based on a function-based approach to the selection of cancer cells in a cell culture that can survive a variety of challenges in prolonged, but reversible, quiescence. These resistant cancer cells possessed a variety of mechanisms, including modifications of the epigenome and transcriptome, for generating a high degree of cellular heterogeneity. We found that long pretreatment with JIB-04 sensitized resistant triple-negative inflammatory breast cancer cells and their parental cell line SUM149 to the chemotherapeutic drugs doxorubicin and paclitaxel. Resistant cancer cells derived from another inflammatory breast cancer cell line, FC-IBC02, were considerably more sensitive to JIB-04 than the parental cell line. Investigating a mechanism of sensitization, we found that JIB-04 exposure increased the expression of PD-L1 in resistant cells, suggesting that JIB-04 may also sensitize resistant breast cancer cells to anti-PD-L1 immune therapy. Finally, these results support the usefulness of a cell culture-based experimental strategy for evaluating anticancer agents, such as JIB-04, that may halt cancer evolution and prevent the development of cancer resistance to currently used therapies.
Collapse
Affiliation(s)
- Balraj Singh
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (B.S.); (A.L.)
| | - Vanessa N. Sarli
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anthony Lucci
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (B.S.); (A.L.)
| |
Collapse
|
24
|
Choo JRE, Jan YH, Ow SGW, Wong A, Lee MX, Ngoi N, Yadav K, Lim JSJ, Lim SE, Chan CW, Hartman M, Tang SW, Goh BC, Tan HL, Chong WQ, Yvonne ALE, Chan GHJ, Chen SJ, Tan KT, Lee SC. Serial Tumor Molecular Profiling of Newly Diagnosed HER2-Negative Breast Cancers During Chemotherapy in Combination with Angiogenesis Inhibitors. Target Oncol 2022; 17:355-368. [PMID: 35699834 PMCID: PMC9217774 DOI: 10.1007/s11523-022-00886-x] [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] [Accepted: 04/26/2022] [Indexed: 11/30/2022]
Abstract
Background Breast cancers are heterogeneous with variable clinical courses and treatment responses. Objective We sought to evaluate dynamic changes in the molecular landscape of HER2-negative tumors treated with chemotherapy and anti-angiogenic agents. Patients and Methods Newly diagnosed HER2-negative breast cancer patients received low-dose sunitinib or bevacizumab prior to four 2-weekly cycles of dose-dense doxorubicin and cyclophosphamide. Tumor biopsies were obtained at baseline, after 2 weeks and after 8 weeks of chemotherapy. Next-generation sequencing was performed to assess for single nucleotide variants (SNVs) and copy number alterations (CNAs) of 440 cancer-related genes (ACTOnco®). Observed genomic changes were correlated with the Miller-Payne histological response to treatment. Results Thirty-four patients received sunitinib and 18 received bevacizumab. In total, 77% were hormone receptor positive (HER2−/HR+) and 23% were triple negative breast cancers (TNBC). New therapy-induced mutations were infrequent, occurring only in 13%, and appeared early after a single cycle of treatment. Seventy-two percent developed changes in the variant allele frequency (VAF) of pathogenic SNVs; the majority (51%) of these changes occurred early at 2 weeks and were sustained for 8 weeks. Changes in VAF of SNVs were most commonly seen in the PI3K/mTOR/AKT pathway; 13% developed changes in pathogenic mutations, which potentially confer sensitivity to PIK3CA inhibitors. Tumors with poor Miller-Payne response to treatment were less likely to experience changes in VAF of SNVs compared with those with good response (50% [7/14] vs 15% [4/24] had no changes observed at any timepoint, p = 0.029). Conclusions Serial molecular profiling identifies early therapy-induced genomic alterations, which may guide future selection of targeted therapies in breast cancer patients who progress after standard chemotherapy. Clinical trial registration ClinicalTrials.gov: NCT02790580 (first posted June 6, 2016). Supplementary Information The online version contains supplementary material available at 10.1007/s11523-022-00886-x.
Collapse
Affiliation(s)
- Joan R E Choo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | | | - Samuel G W Ow
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Andrea Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Matilda Xinwei Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Natalie Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Kritika Yadav
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Joline S J Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Siew Eng Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Ching Wan Chan
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Mikael Hartman
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Siau Wei Tang
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore.,Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Hon Lyn Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Wan Qin Chong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Ang Li En Yvonne
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | - Gloria H J Chan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore
| | | | | | - Soo Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS) National University Health System, 1E Lower Kent Ridge Road, Singapore, 119228, Singapore. .,Cancer Science Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
25
|
Ahn J, Park WC, Yoon CI, Paik PS, Cho MK, Yoo TK. The Radiological Response Rate Pattern Is Associated With Recurrence Free Survival in Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy. J Breast Cancer 2022; 25:106-116. [PMID: 35506579 PMCID: PMC9065354 DOI: 10.4048/jbc.2022.25.e19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 09/24/2021] [Accepted: 04/21/2022] [Indexed: 11/30/2022] Open
Abstract
PURPOSE The aim of this study was to evaluate the radiological response rate patterns during neoadjuvant chemotherapy (NAC) in patients with breast cancer. METHODS Patients who underwent NAC with two specific chemotherapy regimens (doxorubicin with cyclophosphamide or doxorubicin with docetaxel) and who underwent a response evaluation every two cycles were included in the study. The initial response ratio was defined as the ratio of the largest tumor diameter at diagnosis to that after two cycles of NAC. The latter response ratio was defined as the ratio between the tumor size after two cycles and that after four cycles of NAC. The radiological response rate pattern was divided into three groups: the fast-to-slow response group (F-S group, initial response ratio > latter response ratio + 20%), slow-to-fast response group (S-F group, latter response ratio > initial response ratio + 20%), and constant response group (less than 20% difference between the initial and latter response ratios). RESULTS In total, 177 patients were included in the analysis. Forty-two (23.9%) patients were categorized into the F-S group, 26 (14.8%) into the S-F group, and 108 (61.2%) into the constant group. Clinicopathologic factors did not differ according to radiologic response rate patterns. The median follow-up period was 50 months (range, 3-112) months. In the univariate analysis, the F-S group had a significantly worse recurrence-free survival than the S-F and constant groups (hazard ratio [HR], 3.63; 95% confidence interval [CI], 1.05-12.46; p = 0.041). The F-S group also presented with significantly worse survival than the S-F group in the multivariate analysis (HR, 3.45; 95% CI, 1.00-11.89; p = 0.049). CONCLUSION The F-S group had a poorer survival rate than the S-F group. Radiological response rate patterns may be useful for accurate prognostic assessments, especially when considering post-neoadjuvant therapy.
Collapse
Affiliation(s)
- Juneyoung Ahn
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woo-Chan Park
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang Ik Yoon
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Pill Sun Paik
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Kyung Cho
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Tae-Kyung Yoo
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|