1
|
Zhang Q, Wang M, You L, Chen C, Feng J, Song M, Yang K, Liu X, Li G, Liu J. Research progress and application status of organoid in breast cancer subtypes. BIOMOLECULES & BIOMEDICINE 2025; 25:976-985. [PMID: 39720912 PMCID: PMC11984363 DOI: 10.17305/bb.2024.11450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Breast cancer (BC) is a prevalent malignant tumor that poses a significant health risk to women. The complexity of basic BC research and clinical treatment is influenced by multiple factors, including age, fertility, hormone metabolism, molecular subtypes, and tumor grading and staging. Traditional in vitro models often fall short of meeting modern research demands, whereas organoids-an emerging 3D primary culture technology-offer a unique platform that better replicates the tumor microenvironment (TME). Coupled with advances in high-throughput sequencing technologies, organoids have become increasingly valuable in biological and chemical research. Currently, the most widely used organoid model in BC research is the patient-derived organoid (PDO) model, which is generated directly from original tumor tissues. This paper aims to summarize the current status of PDO models across various BC subtypes, highlighting recent advances in genetics, mechanisms of drug resistance, identification of new therapeutic targets, and approaches to personalized treatment. In conclusion, the development of clinical precision medicine urgently requires in vitro models capable of accurately simulating the unique molecular subtypes of patients. This review will examine the challenges and future prospects of organoid models in BC research, offering new insights into the fundamental mechanisms of BC and paving the way for more effective personalized therapies.
Collapse
Affiliation(s)
- Qiuxia Zhang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Min Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Li You
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Chen Chen
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Miao Song
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Kui Yang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Xuexue Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Guangrong Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Sichuan, China
| |
Collapse
|
2
|
Roy NS, Kumari M, Alam K, Bhattacharya A, Kaity S, Kaur K, Ravichandiran V, Roy S. Development of bioengineered 3D patient derived breast cancer organoid model focusing dynamic fibroblast-stem cell reciprocity. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 7:012007. [PMID: 39662055 DOI: 10.1088/2516-1091/ad9dcb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024]
Abstract
Three-dimensional (3D) models, such as tumor spheroids and organoids, are increasingly developed by integrating tissue engineering, regenerative medicine, and personalized therapy strategies. These advanced 3Din-vitromodels are not merely endpoint-driven but also offer the flexibility to be customized or modulated according to specific disease parameters. Unlike traditional 2D monolayer cultures, which inadequately capture the complexities of solid tumors, 3D co-culture systems provide a more accurate representation of the tumor microenvironment. This includes critical interactions with mesenchymal stem/stromal cells (MSCs) and induced pluripotent stem cells (iPSCs), which significantly modulate cancer cell behavior and therapeutic responses. Most of the findings from the co-culture of Michigan Cancer Foundation-7 breast cancer cells and MSC showed the formation of monolayers. Although changes in the plasticity of MSCs and iPSCs caused by other cells and extracellular matrix (ECM) have been extensively researched, the effect of MSCs on cancer stem cell (CSC) aggressiveness is still controversial and contradictory among different research communities. Some researchers have argued that CSCs proliferate more, while others have proposed that cancer spread occurs through dormancy. This highlights the need for further investigation into how these interactions shape cancer aggressiveness. The objective of this review is to explore changes in cancer cell behavior within a 3D microenvironment enriched with MSCs, iPSCs, and ECM components. By describing various MSC and iPSC-derived 3D breast cancer models that replicate tumor biology, we aim to elucidate potential therapeutic targets for breast cancer. A particular focus of this review is the Transwell system, which facilitates understanding how MSCs and iPSCs affect critical processes such as migration, invasion, and angiogenesis. The gradient formed between the two chambers is based on diffusion, as seen in the human body. Once optimized, this Transwell model can serve as a high-throughput screening platform for evaluating various anticancer agents. In the future, primary cell-based and patient-derived 3D organoid models hold promise for advancing personalized medicine and accelerating drug development processes.
Collapse
Affiliation(s)
- Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Mamta Kumari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Department of Anatomy & Regenerative Medicine, Tissue Engineering Research Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| |
Collapse
|
3
|
McDonough E, Barroso M, Ginty F, Corr DT. Modeling intratumor heterogeneity in breast cancer. Biofabrication 2024; 17:10.1088/1758-5090/ad9b50. [PMID: 39642392 PMCID: PMC11740194 DOI: 10.1088/1758-5090/ad9b50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/06/2024] [Indexed: 12/08/2024]
Abstract
Reduced therapy response in breast cancer has been correlated with heterogeneity in biomarker composition, expression level, and spatial distribution of cancer cells within a patient tumor. Thus, there is a need for models to replicate cell-cell, cell-stromal, and cell-microenvironment interactions during cancer progression. Traditional two-dimensional (2D) cell culture models are convenient but cannot adequately represent tumor microenvironment histological organization,in vivo3D spatial/cellular context, and physiological relevance. Recently, three-dimensional (3D)in vitrotumor models have been shown to provide an improved platform for incorporating compositional and spatial heterogeneity and to better mimic the biological characteristics of patient tumors to assess drug response. Advances in 3D bioprinting have allowed the creation of more complex models with improved physiologic representation while controlling for reproducibility and accuracy. This review aims to summarize the advantages and challenges of current 3Din vitromodels for evaluating therapy response in breast cancer, with a particular emphasis on 3D bioprinting, and addresses several key issues for future model development as well as their application to other cancers.
Collapse
Affiliation(s)
- Elizabeth McDonough
- Department of Biomedical Engineering, Rensselaer
Polytechnic Institute, 110 Eighth Street, Troy, New York 12180, United States
- GE HealthCare Technology & Innovation Center, 1
Research Circle, Niskayuna, New York 12309, United States
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany
Medical College, Albany, NY 12208, United States
| | - Fiona Ginty
- GE HealthCare Technology & Innovation Center, 1
Research Circle, Niskayuna, New York 12309, United States
| | - David T. Corr
- Department of Biomedical Engineering, Rensselaer
Polytechnic Institute, 110 Eighth Street, Troy, New York 12180, United States
| |
Collapse
|
4
|
Niazi V, Parseh B. Organoid models of breast cancer in precision medicine and translational research. Mol Biol Rep 2024; 52:2. [PMID: 39570495 DOI: 10.1007/s11033-024-10101-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
One of the most famous and heterogeneous cancers worldwide is breast cancer (BC). Owing to differences in the gene expression profiles and clinical features of distinct BC subtypes, different treatments are prescribed for patients. However, even with more thorough pathological evaluations of tumors than in the past, available treatments do not perform equally well for all individuals. Precision medicine is a new approach that considers the effects of patients' genes, lifestyle, and environment to choose the right treatment for an individual patient. As a powerful tool, the organoid culture system can maintain the morphological and genetic characteristics of patients' tumors. Evidence also shows that organoids have high predictive value for patient treatment. In this review, a variety of BC studies performed on organoid culture systems are evaluated. Additionally, the potential of using organoid models in BC translational research, especially in immunotherapy, drug screening, and precision medicine, has been reported.
Collapse
Affiliation(s)
- Vahid Niazi
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran
- School of Advanced Technologies in Medicine, Golestan University of Medical Science, Shastkola Street, Gorgan, 4918936316, Iran
| | - Benyamin Parseh
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran.
- School of Advanced Technologies in Medicine, Golestan University of Medical Science, Shastkola Street, Gorgan, 4918936316, Iran.
| |
Collapse
|
5
|
Fang T, Xie X, Lu W, Hong Z, Peng W, Zhou J, Wang M, Yao B. Patient-Derived Organoids on a Microarray for Drug Resistance Study in Breast Cancer. Anal Chem 2024; 96:18384-18391. [PMID: 39499082 DOI: 10.1021/acs.analchem.4c02691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Drug resistance is always a challenge in cancer treatment, whether for chemotherapy, targeting, or immunotherapy. Although tumor cell lines are derived from cancer patients, they gradually lost the original characteristics, including heterogeneity and tumor microenvironment (TME), during the long period of in vitro culturing. Therefore, it is urgent to use patient-derived tumor models instead of cancer cell lines to study tumor drug resistance. Herein, we developed a microarray device that serves as a platform for high-throughput and three-dimensional culture of breast cancer patient-derived organoids (BCOs) and investigated their resistance to adriamycin (ADM). Coupled with fluorescence microscopy, this system enabled on-chip drug response monitoring and cell viability assessment without the consumption of a large number of tumor cells. The organoids were divided into a resistant BCO group (RBCO) and a sensitive BCO group (SBCO) according to their half-inhibitory concentration (IC50). Different from cancer cell lines, BCOs demonstrated obvious heterogeneity in drug treatment. Ivermectin (IVM), a broad-spectrum antiparasitic agent approved by the Food and Drug Administration (FDA), was observed to synergistically augment ADM-induced cytotoxicity in organoids. The BCO chip provides a promising platform for investigation of drug resistance and preclinical drug screening based on clinical samples.
Collapse
Affiliation(s)
- Tianyuan Fang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Xinlun Xie
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Wei Lu
- GeneX (Zhejiang) Precision Medicine Co., Ltd, Hangzhou 311100, China
| | - Zichen Hong
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Wenbo Peng
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Jun Zhou
- Department of Breast surgery, The First People's Hospital of Lianyungang, Lianyungang 222002, China
| | - Min Wang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
6
|
Elkordy AA, Hill D, Attia M, Chaw CS. Liposomes and Their Therapeutic Applications in Enhancing Psoriasis and Breast Cancer Treatments. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1760. [PMID: 39513840 PMCID: PMC11547384 DOI: 10.3390/nano14211760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Psoriasis and breast cancer are two examples of diseases where associated inflammatory pathways within the body's immune system are implicated. Psoriasis is a complex, chronic and incurable inflammatory skin disorder that is primarily recognized by thick, scaly plaques on the skin. The most noticeable pathophysiological effect of psoriasis is the abnormal proliferation of keratinocytes. Breast cancer is currently the most diagnosed cancer and the leading cause of cancer-related death among women globally. While treatments targeting the primary tumor have significantly improved, preventing metastasis with systemic treatments is less effective. Nanocarriers such as liposomes and lipid nanoparticles have emerged as promising drug delivery systems for drug targeting and specificity. Advances in technologies and drug combinations have emerged to develop more efficient lipid nanocarriers to include more than one drug in combinational therapy to enhance treatment outcomes and/or relief symptoms for better patients' quality of life. Although there are FDA-approved liposomes with anti-cancer drugs for breast cancer, there are still unmet clinical needs to reduce the side effects associated with those nanomedicines. Hence, combinational nano-therapy may eliminate some of the issues and challenges. Furthermore, there are no nanomedicines yet clinically available for psoriasis. Hence, this review will focus on liposomes encapsulated single and/or combinational therapy to augment treatment outcomes with an emphasis on the effectiveness of combinational therapy within liposomal-based nanoparticulate drug delivery systems to tackle psoriasis and breast cancer. This review will also include an overview of both diseases, challenges in delivering drug therapy and the roles of nanomedicines as well as psoriasis and breast cancer models used for testing therapeutic interventions to pave the way for effective in vivo testing prior to the clinical trials.
Collapse
Affiliation(s)
- Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - David Hill
- School of Nursing and Health Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK;
| | - Mohamed Attia
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - Cheng Shu Chaw
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| |
Collapse
|
7
|
Huang S, Mei Z, Wan A, Zhao M, Qi X. Application and prospect of organoid technology in breast cancer. Front Immunol 2024; 15:1413858. [PMID: 39253075 PMCID: PMC11381393 DOI: 10.3389/fimmu.2024.1413858] [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: 04/08/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Breast cancer is the most common malignant tumor in women. Due to the high heterogeneity of breast cancer cells, traditional in vitro research models still have major limitations. Therefore, it is urgent to establish an experimental model that can accurately simulate the characteristics of human breast cancer. Breast cancer organoid technology emerged as the times required, that is, to construct tissue analogs with organ characteristics by using a patient's tumor tissue through 3D culture in vitro. Since the breast cancer organoid can fully preserve the histology and genetic characteristics of the original tumor, it provides a reliable model for preclinical drug screening, establishment of breast cancer organoid biobanks, research into the mechanisms of tumor development, and determination of cancer targets. It has promoted personalized treatment for clinical breast cancer patients. This article mainly focuses on recent research progress and applications of organoid technology in breast cancer, discussing the current limitations and prospects of breast cancer organoid technology.
Collapse
Affiliation(s)
- Shanlin Huang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zifan Mei
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Andi Wan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Min Zhao
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
8
|
Wang G, Liu T, He WT. Visualization analysis of research hotspots and trends on gastrointestinal tumor organoids. World J Gastrointest Oncol 2024; 16:2826-2841. [PMID: 38994154 PMCID: PMC11236249 DOI: 10.4251/wjgo.v16.i6.2826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/09/2024] [Accepted: 04/19/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Gastrointestinal tumor organoids serve as an effective model for simulating cancer in vitro and have been applied in basic biology and preclinical research. Despite over a decade of development and increasing research achievements in this field, a systematic and comprehensive analysis of the research hotspots and future trends is lacking. AIM To address this problem by employing bibliometric tools to explore the publication years, countries/regions, institutions, journals, authors, keywords, and references in this field. METHODS The literature was collected from Web of Science databases. CiteSpace-6.2R4, a widely used bibliometric analysis software package, was used for institutional analysis and reference burst analysis. VOSviewer 1.6.19 was used for journal co-citation analysis, author co-authorship and co-citation analysis. The 'online platform for bibliometric analysis (https://bibliometric.com/app)' was used to assess the total number of publications and the cooperation relationships between countries. Finally, we employed the bibliometric R software package (version R.4.3.1) in R-studio, for a comprehensive scientific analysis of the literature. RESULTS Our analysis included a total of 1466 publications, revealing a significant yearly increase in articles on the study of gastrointestinal tumor organoids. The United States (n = 393) and Helmholtz Association (n = 93) have emerged as the leading countries and institutions, respectively, in this field, with Hans Clevers and Toshiro Sato being the most contributing authors. The most influential journal in this field is Gastroenterology. The most impactful reference is "Long term expansion of epithelial organs from human colon, adenoma, adenocarcinoma, and Barrett's epithelium". Keywords analysis and citation burst analysis indicate that precision medicine, disease modeling, drug development and screening, and regenerative medicine are the most cutting-edge directions. These focal points were further detailed based on the literature. CONCLUSION This bibliometric study offers an objective and quantitative analysis of the research in this field, which can be considered as an important guide for next scientific research.
Collapse
Affiliation(s)
- Gang Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Tao Liu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou University, Lanzhou 730000, Gansu Province, China
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Wen-Ting He
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Prevention and Treatment and Translational Medicine Engineering Innovation Center of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
- Digestive System Tumor Translational Medicine Engineering Research Center of Gansu Province, Lanzhou University, Lanzhou 730000, Gansu Province, China
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu Province, China
| |
Collapse
|
9
|
Cui Y, Ran R, Da Y, Zhang H, Jiang M, Qi X, Zhang W, Niu L, Zhou Y, Zhou C, Tang X, Wang K, Yan Y, Ren Y, Dong D, Zhou Y, Wang H, Gong J, Hu F, Zhao S, Zhang H, Zhang C, Yang J. The combination of breast cancer PDO and mini-PDX platform for drug screening and individualized treatment. J Cell Mol Med 2024; 28:e18374. [PMID: 38722288 PMCID: PMC11081008 DOI: 10.1111/jcmm.18374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/05/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
The majority of advanced breast cancers exhibit strong aggressiveness, heterogeneity, and drug resistance, and currently, the lack of effective treatment strategies is one of the main challenges that cancer research must face. Therefore, developing a feasible preclinical model to explore tailored treatments for refractory breast cancer is urgently needed. We established organoid biobanks from 17 patients with breast cancer and characterized them by immunohistochemistry (IHC) and next generation sequencing (NGS). In addition, we in the first combination of patient-derived organoids (PDOs) with mini-patient-derived xenografts (Mini-PDXs) for the rapid and precise screening of drug sensitivity. We confirmed that breast cancer organoids are a high-fidelity three-dimension (3D) model in vitro that recapitulates the original tumour's histological and genetic features. In addition, for a heavily pretreated patient with advanced drug-resistant breast cancer, we combined PDO and Mini-PDX models to identify potentially effective combinations of therapeutic agents for this patient who were alpelisib + fulvestrant. In the drug sensitivity experiment of organoids, we observed changes in the PI3K/AKT/mTOR signalling axis and oestrogen receptor (ER) protein expression levels, which further verified the reliability of the screening results. Our study demonstrates that the PDO combined with mini-PDX model offers a rapid and precise drug screening platform that holds promise for personalized medicine, improving patient outcomes and addressing the urgent need for effective therapies in advanced breast cancer.
Collapse
Affiliation(s)
- Yuxin Cui
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Ran Ran
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Yanyan Da
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Center for Molecular Diagnosis and Precision MedicineThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiChina
| | - Huiwen Zhang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Meng Jiang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Xin Qi
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Wei Zhang
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Ligang Niu
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Yuhui Zhou
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Can Zhou
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Xiaojiang Tang
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Ke Wang
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Yu Yan
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Yu Ren
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Danfeng Dong
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Yan Zhou
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Hui Wang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Jin Gong
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Fang Hu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Shidi Zhao
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Huimin Zhang
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| | - Chengsheng Zhang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Center for Molecular Diagnosis and Precision MedicineThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiChina
| | - Jin Yang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiPeople's Republic of China
| |
Collapse
|
10
|
Qu S, Xu R, Yi G, Li Z, Zhang H, Qi S, Huang G. Patient-derived organoids in human cancer: a platform for fundamental research and precision medicine. MOLECULAR BIOMEDICINE 2024; 5:6. [PMID: 38342791 PMCID: PMC10859360 DOI: 10.1186/s43556-023-00165-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 12/08/2023] [Indexed: 02/13/2024] Open
Abstract
Cancer is associated with a high degree of heterogeneity, encompassing both inter- and intra-tumor heterogeneity, along with considerable variability in clinical response to common treatments across patients. Conventional models for tumor research, such as in vitro cell cultures and in vivo animal models, demonstrate significant limitations that fall short of satisfying the research requisites. Patient-derived tumor organoids, which recapitulate the structures, specific functions, molecular characteristics, genomics alterations and expression profiles of primary tumors. They have been efficaciously implemented in illness portrayal, mechanism exploration, high-throughput drug screening and assessment, discovery of innovative therapeutic targets and potential compounds, and customized treatment regimen for cancer patients. In contrast to conventional models, tumor organoids offer an intuitive, dependable, and efficient in vitro research model by conserving the phenotypic, genetic diversity, and mutational attributes of the originating tumor. Nevertheless, the organoid technology also confronts the bottlenecks and challenges, such as how to comprehensively reflect intra-tumor heterogeneity, tumor microenvironment, tumor angiogenesis, reduce research costs, and establish standardized construction processes while retaining reliability. This review extensively examines the use of tumor organoid techniques in fundamental research and precision medicine. It emphasizes the importance of patient-derived tumor organoid biobanks for drug development, screening, safety evaluation, and personalized medicine. Additionally, it evaluates the application of organoid technology as an experimental tumor model to better understand the molecular mechanisms of tumor. The intent of this review is to explicate the significance of tumor organoids in cancer research and to present new avenues for the future of tumor research.
Collapse
Affiliation(s)
- Shanqiang Qu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
| | - Rongyang Xu
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- The First Clinical Medical College of Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
| | - Huayang Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China.
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Nanfang Glioma Center, Guangzhou, 510515, Guangdong, China.
- Institute of Brain disease, Nanfang Hospital, Southern Medical University, Guangzhou Dadao Bei Street 1838, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
11
|
Wang S, Wu W, Lin X, Zhang KM, Wu Q, Luo M, Zhou J. Predictive and prognostic biomarkers of bone metastasis in breast cancer: current status and future directions. Cell Biosci 2023; 13:224. [PMID: 38041134 PMCID: PMC10693103 DOI: 10.1186/s13578-023-01171-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023] Open
Abstract
The most common site of metastasis in breast cancer is the bone, where the balance between osteoclast-mediated bone resorption and osteoblast-mediated bone formation is disrupted. This imbalance causes osteolytic bone metastasis in breast cancer, which leads to bone pain, pathological fractures, spinal cord compression, and other skeletal-related events (SREs). These complications reduce patients' quality of life significantly and have a profound impact on prognosis. In this review, we begin by providing a brief overview of the epidemiology of bone metastasis in breast cancer, including current diagnostic tools, treatment approaches, and existing challenges. Then, we will introduce the pathophysiology of breast cancer bone metastasis (BCBM) and the animal models involved in the study of BCBM. We then come to the focus of this paper: a discussion of several biomarkers that have the potential to provide predictive and prognostic value in the context of BCBM-some of which may be particularly compatible with more comprehensive liquid biopsies. Beyond that, we briefly explore the potential of new technologies such as single-cell sequencing and organoid models, which will improve our understanding of tumor heterogeneity and aid in the development of improved biomarkers. The emerging biomarkers discussed hold promise for future clinical application, aiding in the prevention of BCBM, improving the prognosis of patients, and guiding the implementation of personalized medicine.
Collapse
Affiliation(s)
- Shenkangle Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Wenxin Wu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Xixi Lin
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | | | - QingLiang Wu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Hangzhou Ninth People's Hospital, Hangzhou, 310014, China
| | - Mingpeng Luo
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310014, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
12
|
Nuñez-Olvera SI, Aguilar-Arnal L, Cisneros-Villanueva M, Hidalgo-Miranda A, Marchat LA, Salinas-Vera YM, Ramos-Payán R, Pérez-Plasencia C, Carlos-Reyes Á, Puente-Rivera J, López-Camarillo C. Breast Cancer Cells Reprogram the Oncogenic lncRNAs/mRNAs Coexpression Networks in Three-Dimensional Microenvironment. Cells 2022; 11:3458. [PMID: 36359853 PMCID: PMC9656377 DOI: 10.3390/cells11213458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Organotypic three-dimensional (3D) cell cultures more accurately mimic the characteristics of solid tumors in vivo in comparison with traditional two-dimensional (2D) monolayer cell models. Currently, studies on the regulation of long non-coding RNAs (lncRNAs) have not been explored in breast cancer cells cultured in 3D microenvironments. In the present research, we studied the expression and potential roles of lncRNAs in estrogen receptor-positive luminal B subtype BT-474 breast cancer cells grown over extracellular matrix proteins-enriched 3D cultures. Global expression profiling using DNA microarrays identifies 290 upregulated and 183 downregulated lncRNAs in 3D cultures relative to 2D condition. Using a co-expression analysis approach of lncRNAs and mRNAs pairs expressed in the same experimental conditions, we identify hundreds of regulatory axes modulating genes involved in cancer hallmarks, such as responses to estrogens, cell proliferation, hypoxia, apical junctions, and resistance to endocrine therapy. In addition, we identified 102 lncRNAs/mRNA correlations in 3D cultures, which were similar to those reported in TCGA datasets obtained from luminal B breast cancer patients. Interestingly, we also found a set of mRNAs transcripts co-expressed with LINC00847 and CTD-2566J3.1 lncRNAs, which were predictors of pathologic complete response and overall survival. Finally, both LINC00847 and CTD -2566J3.1 were co-expressed with essential genes for cancer genetic dependencies, such as FOXA1 y GINS2. Our experimental and predictive findings show that co-expressed lncRNAs/mRNAs pairs exhibit a high degree of similarity with those found in luminal B breast cancer patients, suggesting that they could be adequate pre-clinical tools to identify not only biomarkers related to endocrine therapy response and PCR, but to understand the biological behavior of cancer cells in 3D microenvironments.
Collapse
Affiliation(s)
- Stephanie I. Nuñez-Olvera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, ENMyH-Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Yarely M. Salinas-Vera
- Departamento de Bioquímica, Centro de Investigación y Estudios Avanzados, Mexico City 07360, Mexico
| | - Rosalio Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán 80040, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan). Av. San Fernando 22, Col. Sección XVI. Tlalpan, Mexico City 14080, Mexico
- Laboratorio de Genómica, Unidad de Biomedicina, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Ángeles Carlos-Reyes
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias (INER) Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - Jonathan Puente-Rivera
- División de Ciencias de la Salud, Biológicas y Ambientales, Universidad Abierta y a Distancia, Mexico City 03330, Mexico
| | - Cesar López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City 03100, Mexico
| |
Collapse
|
13
|
Derouane F, van Marcke C, Berlière M, Gerday A, Fellah L, Leconte I, Van Bockstal MR, Galant C, Corbet C, Duhoux FP. Predictive Biomarkers of Response to Neoadjuvant Chemotherapy in Breast Cancer: Current and Future Perspectives for Precision Medicine. Cancers (Basel) 2022; 14:3876. [PMID: 36010869 PMCID: PMC9405974 DOI: 10.3390/cancers14163876] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 02/07/2023] Open
Abstract
Pathological complete response (pCR) after neoadjuvant chemotherapy in patients with early breast cancer is correlated with better survival. Meanwhile, an expanding arsenal of post-neoadjuvant treatment strategies have proven beneficial in the absence of pCR, leading to an increased use of neoadjuvant systemic therapy in patients with early breast cancer and the search for predictive biomarkers of response. The better prediction of response to neoadjuvant chemotherapy could enable the escalation or de-escalation of neoadjuvant treatment strategies, with the ultimate goal of improving the clinical management of early breast cancer. Clinico-pathological prognostic factors are currently used to estimate the potential benefit of neoadjuvant systemic treatment but are not accurate enough to allow for personalized response prediction. Other factors have recently been proposed but are not yet implementable in daily clinical practice or remain of limited utility due to the intertumoral heterogeneity of breast cancer. In this review, we describe the current knowledge about predictive factors for response to neoadjuvant chemotherapy in breast cancer patients and highlight the future perspectives that could lead to the better prediction of response, focusing on the current biomarkers used for clinical decision making and the different gene signatures that have recently been proposed for patient stratification and the prediction of response to therapies. We also discuss the intratumoral phenotypic heterogeneity in breast cancers as well as the emerging techniques and relevant pre-clinical models that could integrate this biological factor currently limiting the reliable prediction of response to neoadjuvant systemic therapy.
Collapse
Affiliation(s)
- Françoise Derouane
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Cédric van Marcke
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Martine Berlière
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Department of Gynecology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Gynecology (GYNE), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Amandine Gerday
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Department of Gynecology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Latifa Fellah
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Department of Radiology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Isabelle Leconte
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Department of Radiology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Mieke R. Van Bockstal
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Department of Pathology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Christine Galant
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Department of Pathology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Cyril Corbet
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Francois P. Duhoux
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
14
|
Van Eaton KM, Gustafson DL. Pharmacokinetic and Pharmacodynamic Assessment of Hydroxychloroquine in Breast Cancer. J Pharmacol Exp Ther 2021; 379:331-342. [PMID: 34503992 PMCID: PMC9351720 DOI: 10.1124/jpet.121.000730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Hydroxychloroquine (HCQ) is being tested in a number of human clinical trials to determine the role of autophagy in response to standard anticancer therapies. However, HCQ pharmacodynamic (PD) responses are difficult to assess in patients, and preclinical studies in mouse models are equivocal with regard to HCQ exposure and inhibition of autophagy. Here, pharmacokinetic (PK) assessment of HCQ in non-tumor-bearing mice after intraperitoneal dosing established 60 mg/kg as the human equivalent dose of HCQ in mice. Autophagy inhibition, cell proliferation, and cell death were assessed in two-dimensional (2D) cell culture and three-dimensional tumor organoids in breast cancer. Mice challenged with breast cancer xenografts were then treated with 60 mg/kg HCQ via intraperitoneal dosing, and subsequent PK and PD responses were assessed. Although autophagic flux was significantly inhibited in cells irrespective of autophagy-dependence status, autophagy-dependent tumors had decreased cell proliferation and increased cell death at earlier time points compared with autophagy-independent tumors. Overall, this study shows that 2D cell culture, three-dimensional tumor organoids, and in vivo studies produce similar results, and in vitro studies can be used as surrogates to recapitulate in vivo antitumor responses of HCQ. SIGNIFICANCE STATEMENT: Autophagy-dependent tumors but not autophagy-independent tumors have decreased cell proliferation and increased cell death after single-agent hydroxychloroquine treatment. However, hydroxychloroquine causes decreased autophagic flux regardless of autophagy status, suggesting its clinical efficacy in the context of autophagy inhibition.
Collapse
Affiliation(s)
- Kristen M Van Eaton
- School of Biomedical Engineering (K.M.V.E., D.L.G.), Department of Clinical Sciences (D.L.G.), and Flint Animal Cancer Center (D.L.G.), Colorado State University, Fort Collins, Colorado; and Developmental Therapeutics Program; University of Colorado Cancer Center, Aurora, Colorado (D.L.G.)
| | - Daniel L Gustafson
- School of Biomedical Engineering (K.M.V.E., D.L.G.), Department of Clinical Sciences (D.L.G.), and Flint Animal Cancer Center (D.L.G.), Colorado State University, Fort Collins, Colorado; and Developmental Therapeutics Program; University of Colorado Cancer Center, Aurora, Colorado (D.L.G.)
| |
Collapse
|
15
|
Ebrahimi N, Nasr Esfahani A, Samizade S, Mansouri A, Ghanaatian M, Adelian S, Shadman Manesh V, Hamblin MR. The potential application of organoids in breast cancer research and treatment. Hum Genet 2021; 141:193-208. [PMID: 34713317 DOI: 10.1007/s00439-021-02390-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/16/2021] [Indexed: 12/23/2022]
Abstract
Tumor heterogeneity is a major challenge for breast cancer researchers who have struggled to find effective treatments despite recent advances in oncology. Although the use of 2D cell culture methods in breast cancer research has been effective, it cannot model the heterogeneity of breast cancer as found within the body. The development of 3D culture of tumor cells and breast cancer organoids has provided a new approach in breast cancer research, allowing the identification of biomarkers, study of the interaction of tumor cells with the microenvironment, and for drug screening and discovery. In addition, the possibility of gene editing in organoids, especially using the CRISPR/Cas9 system, is convenient, and has allowed a more detailed study of tumor behavior in models closer to the physiological condition. The present review covers the application of organoids in breast cancer research. The recent use of gene-editing systems to provide insights into therapeutic approaches for breast cancer, is highlighted. The study of organoids and the possibility of gene manipulation may be a step towards the personalized treatment of breast cancer, which has so far remained unattainable due to the high heterogeneity of breast cancer.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Division of Genetics, Department of Cell, Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Alireza Nasr Esfahani
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Setare Samizade
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Atena Mansouri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Ghanaatian
- Department of Microbiology, Islamic Azad University of Jahrom, Jahrom, Fars, Iran
| | - Samaneh Adelian
- Department of Genetics, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vida Shadman Manesh
- Medical Engineering Tissue Engineering, Department of Medical Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Michael R Hamblin
- Faculty of Health Science, Laser Research Centre, University of Johannesburg, Doornfontein, Johannesburg, 2028, South Africa.
| |
Collapse
|
16
|
Ma YS, Yang XL, Xin R, Wu TM, Shi Y, Dan Zhang D, Wang HM, Wang PY, Liu JB, Fu D. The power and the promise of organoid models for cancer precision medicine with next-generation functional diagnostics and pharmaceutical exploitation. Transl Oncol 2021; 14:101126. [PMID: 34020369 PMCID: PMC8144479 DOI: 10.1016/j.tranon.2021.101126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/25/2022] Open
Abstract
As organ-specific three-dimensional cell clusters derived from cancer tissue or cancer-specific stem cells, cancer-derived organoids are organized in the same manner of the cell sorting and spatial lineage restriction in vivo, making them ideal for simulating the characteristics of cancer and the heterogeneity of cancer cells in vivo. Besides the applications as a new in vitro model to study the physiological characteristics of normal tissues and organs, organoids are also used for in vivo cancer cell characterization, anti-cancer drug screening, and precision medicine. However, organoid cultures are not without limitations, i.e., the lack of nerves, blood vessels, and immune cells. As a result, organoids could not fully replicate the characteristics of organs but partially simulate the disease process. This review attempts to provide insights into the organoid models for cancer precision medicine.
Collapse
Affiliation(s)
- Yu-Shui Ma
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China; Cancer Institute, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong 226631, China; International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, National Center for Liver Cancer, the Second Military Medical University, Shanghai 200433, China
| | - Xiao-Li Yang
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rui Xin
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ting-Miao Wu
- Department of Radiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei 230012, China
| | - Yi Shi
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Dan Dan Zhang
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Hui-Min Wang
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Pei-Yao Wang
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ji-Bin Liu
- Cancer Institute, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong 226631, China
| | - Da Fu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China; Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Radiology, The Forth Affiliated Hospital of Anhui Medical University, Hefei 230012, China.
| |
Collapse
|
17
|
Li D, Li L, Chen X, Zhou C, Hao B, Cao Y. Dysregulation of lncRNA-CCRR contributes to brain metastasis of breast cancer by intercellular coupling via regulating connexin 43 expression. J Cell Mol Med 2021; 25:4826-4834. [PMID: 33793070 PMCID: PMC8107087 DOI: 10.1111/jcmm.16455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/19/2021] [Accepted: 03/01/2021] [Indexed: 01/05/2023] Open
Abstract
Cardiac conduction regulatory RNA (CCRR) is down‐regulated in the pathogenesis of heart failure (HF), which accordingly suppresses cardiac conduction while promoting arrhythmogenicity. Meanwhile, CX43 was reported to play a role in the pathogenesis of metastatic breast cancer and melanoma brain colonization. In this study, we studied the role of long non‐coding RNA CCRR and its interaction with CX43 in brain metastasis of breast cancer. Breast cancer patients were grouped according to the metastasis status. Real‐time PCR and IHC assay were used to measure the expression of lncRNA‐CCRR and CX43 in patients. Western blot was conducted to observe the effect of lncRNA‐CCRR on the expression of CX43 in MDA‐MB‐231BR and BT‐474BR cells. Compared with the non‐metastasis group, the mRNA expression of tissue lncRNA‐CCRR, cerebrospinal fluid (CSF) lncRNA‐CCRR, tissue CX43 and tissue protein expression of CX43 were both evidently up‐regulated in metastasis patients, especially in patients with brain metastasis. The expression of lncRNA‐CCRR was positively correlated with the up‐regulated expression of CX43. Moreover, CX43 expression was significantly lower in MDA‐MB‐231WT cells compared with that in MDA‐MB‐231BR cells. Also, the overexpression of lncRNA‐CCRR evidently increased dye transfer rate from astrocytes to MDA‐MB‐231BR/BT‐474BR cells but reduced lncRNA‐CCRR expression and suppressed the transmigration of MDA‐MB‐231BR/BT‐474BR cells in a blood‐brain barrier (BBB) model. In this study, we demonstrated that the presence of lncRNA‐CCRR could up‐regulate the expression of CX43, which promoted gap junction formation in brain metastasis of breast cancer. Accordingly, the communication between breast cancer cells and astrocytes was also promoted.
Collapse
Affiliation(s)
- Deheng Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liangdong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Chen
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Changshuai Zhou
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Hao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiqun Cao
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Organoid As a Novel Technology for Disease Modeling. JOURNAL OF BASIC AND CLINICAL HEALTH SCIENCES 2021. [DOI: 10.30621/jbachs.868837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
19
|
Sumbal J, Budkova Z, Traustadóttir GÁ, Koledova Z. Mammary Organoids and 3D Cell Cultures: Old Dogs with New Tricks. J Mammary Gland Biol Neoplasia 2020; 25:273-288. [PMID: 33210256 DOI: 10.1007/s10911-020-09468-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022] Open
Abstract
3D cell culture methods have been an integral part of and an essential tool for mammary gland and breast cancer research for half a century. In fact, mammary gland researchers, who discovered and deciphered the instructive role of extracellular matrix (ECM) in mammary epithelial cell functional differentiation and morphogenesis, were the pioneers of the 3D cell culture techniques, including organoid cultures. The last decade has brought a tremendous increase in the 3D cell culture techniques, including modifications and innovations of the existing techniques, novel biomaterials and matrices, new technological approaches, and increase in 3D culture complexity, accompanied by several redefinitions of the terms "3D cell culture" and "organoid". In this review, we provide an overview of the 3D cell culture and organoid techniques used in mammary gland biology and breast cancer research. We discuss their advantages, shortcomings and current challenges, highlight the recent progress in reconstructing the complex mammary gland microenvironment in vitro and ex vivo, and identify the missing 3D cell cultures, urgently needed to aid our understanding of mammary gland development, function, physiology, and disease, including breast cancer.
Collapse
Affiliation(s)
- Jakub Sumbal
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Budkova
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Gunnhildur Ásta Traustadóttir
- Stem Cell Research Unit, Biomedical Center, Department of Anatomy, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland.
| | - Zuzana Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
20
|
Yang L, Liu B, Chen H, Gao R, Huang K, Guo Q, Li F, Chen W, He J. Progress in the application of organoids to breast cancer research. J Cell Mol Med 2020; 24:5420-5427. [PMID: 32283573 PMCID: PMC7214171 DOI: 10.1111/jcmm.15216] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 02/13/2020] [Accepted: 03/06/2020] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is the most common cancer diagnosed in women. Breast cancer research is currently based mainly on animal models and traditional cell culture. However, the inherent species gap between humans and animals, as well as differences in organization between organs and cells, limits research advances. The breast cancer organoid can reproduce many of the key features of human breast cancer, thereby providing a new platform for investigating the mechanisms underlying the development, progression, metastasis and drug resistance of breast cancer. The application of organoid technology can also promote drug discovery and the design of individualized treatment strategies. Here, we discuss the latest advances in the use of organoid technology for breast cancer research.
Collapse
Affiliation(s)
- Liping Yang
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Breast Surgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Baoer Liu
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Breast Surgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haodong Chen
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Rui Gao
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Kanghua Huang
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qiuyi Guo
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Feng Li
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Weicai Chen
- Department of Breast Surgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jinsong He
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|