1
|
Klima S, Hurrell T, Goolam M, Gouws C, Engelbrecht AM, Kaur M, van den Bout I. A new dawn: Vitalising translational oncology research in Africa with the help of advanced cell culture models. Transl Oncol 2025; 56:102391. [PMID: 40228390 PMCID: PMC12017847 DOI: 10.1016/j.tranon.2025.102391] [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: 11/27/2024] [Revised: 03/12/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
The advent of in vitro models such as induced pluripotent stem cells (iPSC) and patient derived (disease) organoids is supporting the development of population and patient specific model systems reflecting human physiology and disease. However, there remains a significant underrepresentation of non-European, especially African model systems. The development of such models should be enthusiastically embraced by Sub-Saharan African countries (SSAC) and middle-income countries (LIMC) to direct their own research focused on the improvement of health of their own populations at a sustainable cost within their respective funding environments. Great care needs to be taken to develop national frameworks to direct, sustainably fund and support such efforts in a way that maximises the output of such models for the investment required. Here, we highlight how advanced culture models can play a role in vitalising local healthcare research by focusing on locally relevant health care questions using appropriate cell culture models. We also provide a potential national platform example that could maximise such output at the lowest cost. This framework presents an opportunity for SSAC and LMIC to base their healthcare research on locally relevant models to ensure that developed health care initiatives and interventions are best suited for the populations they serve and thus represent a reset in global health care research at large.
Collapse
Affiliation(s)
- Stefanie Klima
- Centre for Neuroendocrinology and Department of Immunology, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Tracey Hurrell
- Bioengineering and Integrated Genomics Group, Council for Scientific and Industrial Research, Pretoria, South Africa; Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, South Africa
| | - Mubeen Goolam
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, South Africa; UCT Neuroscience Institute, Cape Town, South Africa
| | - Chrisna Gouws
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), Faculty of Health Sciences, North-West University, South Africa; Desmond Tutu School of Medicine, Faculty of Health Sciences, North-West University, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, South Africa
| | - Iman van den Bout
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, South Africa.
| |
Collapse
|
2
|
Khalili-Tanha G, Radisky ES, Radisky DC, Shoari A. Matrix metalloproteinase-driven epithelial-mesenchymal transition: implications in health and disease. J Transl Med 2025; 23:436. [PMID: 40217300 PMCID: PMC11992850 DOI: 10.1186/s12967-025-06447-w] [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: 02/15/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells, defined by apical-basal polarity and tight intercellular junctions, acquire migratory and invasive properties characteristic of mesenchymal cells. Under normal conditions, EMT directs essential morphogenetic events in embryogenesis and supports tissue repair. When dysregulated, EMT contributes to pathological processes such as organ fibrosis, chronic inflammation, and cancer progression and metastasis. Matrix metalloproteinases (MMPs)-a family of zinc-dependent proteases that degrade structural components of the extracellular matrix-sit at the nexus of this transition by dismantling basement membranes, activating pro-EMT signaling pathways, and cleaving adhesion molecules. When normally regulated, MMPs promote balanced ECM turnover and support the cyclical remodeling necessary for proper development, wound healing, and tissue homeostasis. When abnormally regulated, MMPs drive excessive ECM turnover, thereby promoting EMT-related pathologies, including tumor progression and fibrotic disease. This review provides an integrated overview of the molecular mechanisms by which MMPs both initiate and sustain EMT under physiological and disease conditions. It discusses how MMPs can potentiate EMT through TGF-β and Wnt/β-catenin signaling, disrupt cell-cell junction proteins, and potentiate the action of hypoxia-inducible factors in the tumor microenvironment. It discusses how these pathologic processes remodel tissues during fibrosis, and fuel cancer cell invasion, metastasis, and resistance to therapy. Finally, the review explores emerging therapeutic strategies that selectively target MMPs and EMT, ranging from CRISPR/Cas-mediated interventions to engineered tissue inhibitors of metalloproteinases (TIMPs), and demonstrates how such approaches may suppress pathological EMT without compromising its indispensable roles in normal biology.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
3
|
Yuan X, Rosen JM. Histone acetylation modulators in breast cancer. Breast Cancer Res 2025; 27:49. [PMID: 40165290 PMCID: PMC11959873 DOI: 10.1186/s13058-025-02006-9] [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: 10/30/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Breast cancer is the most prevalent cancer in women worldwide. Aberrant epigenetic reprogramming such as dysregulation of histone acetylation has been associated with the development of breast cancer. Histone acetylation modulators have been targeted as potential treatments for breast cancer. This review comprehensively discusses the roles of these modulators and the effects of their inhibitors on breast cancer. In addition, epigenetic reprogramming not only affects breast cancer cells but also the immunosuppressive myeloid cells, which can facilitate breast cancer progression. Therefore, the review also highlights the roles of these immunosuppressive myeloid cells and summarizes how histone acetylation modulators affect their functions and phenotypes. This review provides insights into histone acetylation modulators as potential therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Xueying Yuan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
4
|
SUN H, XIONG M, XIE S, WEN J. [Epithelial-mesenchymal Transition: Biological Basis and Clinical Prospects
of Lung Cancer Invasion, Metastasis, and Drug Resistance]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2025; 28:155-164. [PMID: 40114492 PMCID: PMC11931237 DOI: 10.3779/j.issn.1009-3419.2025.102.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Indexed: 03/22/2025]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, characterized by high incidence and mortality rates. The primary reasons for treatment failure in lung cancer patients are tumor invasion and drug resistance, particularly resistance to chemotherapeutic agents and epidermal growth factor receptor (EGFR) mutant targeted therapy, which considerably undermine the therapeutic outcomes for those with advanced lung cancer. Epithelial-mesenchymal transition (EMT) serves as a crucial biological process closely associated with physiological or pathological processes such as tissue embryogenesis, organogenesis, wound repair, and tumor invasion. Numerous studies have indicated that EMT, mediated through various signaling pathways, plays a pivotal role in the initiation, progression, and metastasis of lung cancer, while it is also closely associated with drug resistance in lung cancer cells. Therefore, research focusing on the molecular mechanisms and pathophysiology related to EMT can contribute to reversing drug resistance in drug treatment for lung cancer, thereby improving prognosis. This article reviews the progress in research on EMT in the invasion, metastasis, and drug resistance of lung cancer based on relevant domestic and international literature.
Collapse
|
5
|
Ferreira N, Kulkarni A, Agorku D, Midelashvili T, Hardt O, Legler TJ, Ströbel P, Conradi LC, Alves F, Ramos-Gomes F, Markus MA. OrganoIDNet: a deep learning tool for identification of therapeutic effects in PDAC organoid-PBMC co-cultures from time-resolved imaging data. Cell Oncol (Dordr) 2025; 48:101-122. [PMID: 38805131 PMCID: PMC11850476 DOI: 10.1007/s13402-024-00958-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
PURPOSE Pancreatic Ductal Adenocarcinoma (PDAC) remains a challenging disease due to its complex biology and aggressive behavior with an urgent need for efficient therapeutic strategies. To assess therapy response, pre-clinical PDAC organoid-based models in combination with accurate real-time monitoring are required. METHODS We established stable live-imaging organoid/peripheral blood mononuclear cells (PBMCs) co-cultures and introduced OrganoIDNet, a deep-learning-based algorithm, capable of analyzing bright-field images of murine and human patient-derived PDAC organoids acquired with live-cell imaging. We investigated the response to the chemotherapy gemcitabine in PDAC organoids and the PD-L1 inhibitor Atezolizumab, cultured with or without HLA-matched PBMCs over time. Results obtained with OrganoIDNet were validated with the endpoint proliferation assay CellTiter-Glo. RESULTS Live cell imaging in combination with OrganoIDNet accurately detected size-specific drug responses of organoids to gemcitabine over time, showing that large organoids were more prone to cytotoxic effects. This approach also allowed distinguishing between healthy and unhealthy status and measuring eccentricity as organoids' reaction to therapy. Furthermore, imaging of a new organoids/PBMCs sandwich-based co-culture enabled longitudinal analysis of organoid responses to Atezolizumab, showing an increased potency of PBMCs tumor-killing in an organoid-individual manner when Atezolizumab was added. CONCLUSION Optimized PDAC organoid imaging analyzed by OrganoIDNet represents a platform capable of accurately detecting organoid responses to standard PDAC chemotherapy over time. Moreover, organoid/immune cell co-cultures allow monitoring of organoid responses to immunotherapy, offering dynamic insights into treatment behavior within a co-culture setting with PBMCs. This setup holds promise for real-time assessment of immunotherapeutic effects in individual patient-derived PDAC organoids.
Collapse
Affiliation(s)
- Nathalia Ferreira
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ajinkya Kulkarni
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - David Agorku
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Teona Midelashvili
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Olaf Hardt
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tobias J Legler
- Department of Transfusion Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Frauke Alves
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Hematology and Medical Oncology, Department of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Fernanda Ramos-Gomes
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - M Andrea Markus
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
6
|
Łazarczyk M, Skiba D, Mickael ME, Jaskuła K, Nawrocka A, Religa P, Sacharczuk M. Opioid System and Epithelial-Mesenchymal Transition. Pharmaceuticals (Basel) 2025; 18:120. [PMID: 39861181 PMCID: PMC11768736 DOI: 10.3390/ph18010120] [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: 11/30/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Opioids are a challenging class of drugs due to their dual role. They alleviate pain, but also pose a risk of dependency, or trigger constipation, particularly in cancer patients, who require the more potent painkillers in more advanced stages of the disease, closely linked to pain resulting from general inflammation, bone metastases, and primary or secondary tumour outgrowth-related nerve damage. Clinicians' vigilance considering treatment with opioids is necessary, bearing in mind extensive data accumulated over decades that have reported the contribution of opioids to immunosuppression, tumour progression, or impaired tissue regeneration, either following opioid use during surgical tumour resection and post-surgical pain treatment, or as a result of other diseases like diabetes, where chronic wounds healing constitutes a challenge. During last few years, an increasing trend for seeking relationships between opioids and epithelial-mesenchymal transition (EMT) in cancer research can be observed. Transiently lasting EMT is desirable during wound healing, but in cancer, or vital organ fibrogenesis, EMT appears to be an obstacle to overcome, forcing to adjust treatment strategies that would reduce the risk for worsening of the disease outcome and patient prognosis. The same opioid may demonstrate promoting or inhibitory effect on EMT, dependently on various conditions in particular clinical cases. We have summarized current findings on this issue to uncover some rules that govern opioid-mediated EMT induction or repression; however, many aspects still remain to be elucidated.
Collapse
Affiliation(s)
- Marzena Łazarczyk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland
| | - Dominik Skiba
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland
| | - Michel-Edwar Mickael
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland
| | - Kinga Jaskuła
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland
| | - Agata Nawrocka
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland
| | - Piotr Religa
- Department of Medicine, Karolinska Institute, 171 77 Solna, Sweden
| | - Mariusz Sacharczuk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B, 02-091 Warsaw, Poland
| |
Collapse
|
7
|
Frank SA, Yanai I. The origin of novel traits in cancer. Trends Cancer 2024; 10:880-892. [PMID: 39112299 DOI: 10.1016/j.trecan.2024.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 10/11/2024]
Abstract
The traditional view of cancer emphasizes a genes-first process. Novel cancer traits arise by genetic mutations that spread to drive phenotypic change. However, recent data support a phenotypes-first process in which nonheritable cellular variability creates novel traits that later become heritably stabilized by genetic and epigenetic changes. Single-cell measurements reinforce the idea that phenotypes lead genotypes, showing how cancer evolution follows normal developmental plasticity and creates novel traits by recombining parts of different cellular developmental programs. In parallel, studies in evolutionary biology also support a phenotypes-first process driven by developmental plasticity and developmental recombination. These advances in cancer research and evolutionary biology mutually reinforce a revolution in our understanding of how cells and organisms evolve novel traits in response to environmental challenges.
Collapse
Affiliation(s)
- Steven A Frank
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA 92697-2525, USA.
| | - Itai Yanai
- Perlmutter Cancer Center, New York University (NYU) Grossman School of Medicine, New York, NY 10016, USA; Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
8
|
Yuan X, Hao X, Chan HL, Zhao N, Pedroza DA, Liu F, Le K, Smith AJ, Calderon SJ, Lieu N, Soth MJ, Jones P, Zhang XH, Rosen JM. CREB-binding protein/P300 bromodomain inhibition reduces neutrophil accumulation and activates antitumor immunity in triple-negative breast cancer. JCI Insight 2024; 9:e182621. [PMID: 39287984 PMCID: PMC11533985 DOI: 10.1172/jci.insight.182621] [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: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated neutrophils (TANs) have been shown to promote immunosuppression and tumor progression, and a high TAN frequency predicts poor prognosis in triple-negative breast cancer (TNBC). Dysregulation of CREB-binding protein (CBP)/P300 function has been observed with multiple cancer types. The bromodomain (BRD) of CBP/P300 has been shown to regulate its activity. In this study, we found that IACS-70654, a selective CBP/P300 BRD inhibitor, reduced TANs and inhibited the growth of neutrophil-enriched TNBC models. In the bone marrow, CBP/P300 BRD inhibition reduced the tumor-driven abnormal differentiation and proliferation of neutrophil progenitors. Inhibition of CBP/P300 BRD also stimulated the immune response by inducing an IFN response and MHCI expression in tumor cells and increasing tumor-infiltrated cytotoxic T cells. Moreover, IACS-70654 improved the response of a neutrophil-enriched TNBC model to docetaxel and immune checkpoint blockade. This provides a rationale for combining a CBP/P300 BRD inhibitor with standard-of-care therapies in future clinical trials for neutrophil-enriched TNBC.
Collapse
Affiliation(s)
- Xueying Yuan
- Department of Molecular and Cellular Biology and
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Hilda L. Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Na Zhao
- Department of Molecular and Cellular Biology and
| | - Diego A. Pedroza
- Department of Molecular and Cellular Biology and
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Kang Le
- Institute for Applied Cancer Science (IACS), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Nadia Lieu
- Department of Molecular and Cellular Biology and
| | - Michael J. Soth
- Institute for Applied Cancer Science (IACS), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Philip Jones
- Institute for Applied Cancer Science (IACS), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiang H.F. Zhang
- Department of Molecular and Cellular Biology and
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
9
|
Wang Z, Hulikova A, Swietach P. Innovating cancer drug discovery with refined phenotypic screens. Trends Pharmacol Sci 2024; 45:723-738. [PMID: 39013672 DOI: 10.1016/j.tips.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024]
Abstract
Before molecular pathways in cancer were known to a depth that could predict targets, drug development relied on phenotypic screening, where the effectiveness of candidate chemicals is judged from functional readouts without considering the mechanisms of action. The unraveling of tumor-specific pathways has brought targets for molecularly driven drug discovery, but precedents in the field have shown that awareness of pathways does not necessarily predict therapeutic efficacy, and many cancers still lack druggable targets. Phenotypic screening therefore retains a niche in drug development where a targeted approach is not informative. We analyze the unique advantages of phenotypic screens, and how technological advances have improved their discovery power. Notable advances include the use of larger biological panels and refined protocols that address the disease-relevance and increase data content with imaging and omic approaches.
Collapse
Affiliation(s)
- Zhenyi Wang
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK.
| |
Collapse
|
10
|
Huang Y, Liu T, Huang Q, Wang Y. From Organ-on-a-Chip to Human-on-a-Chip: A Review of Research Progress and Latest Applications. ACS Sens 2024; 9:3466-3488. [PMID: 38991227 DOI: 10.1021/acssensors.4c00004] [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: 07/13/2024]
Abstract
Organ-on-a-Chip (OOC) technology, which emulates the physiological environment and functionality of human organs on a microfluidic chip, is undergoing significant technological advancements. Despite its rapid evolution, this technology is also facing notable challenges, such as the lack of vascularization, the development of multiorgan-on-a-chip systems, and the replication of the human body on a single chip. The progress of microfluidic technology has played a crucial role in steering OOC toward mimicking the human microenvironment, including vascularization, microenvironment replication, and the development of multiorgan microphysiological systems. Additionally, advancements in detection, analysis, and organoid imaging technologies have enhanced the functionality and efficiency of Organs-on-Chips (OOCs). In particular, the integration of artificial intelligence has revolutionized organoid imaging, significantly enhancing high-throughput drug screening. Consequently, this review covers the research progress of OOC toward Human-on-a-chip, the integration of sensors in OOCs, and the latest applications of organoid imaging technologies in the biomedical field.
Collapse
Affiliation(s)
- Yisha Huang
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China
| | - Tong Liu
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Huang
- School of Information Engineering, Shanghai Maritime University, Shanghai 201306, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
11
|
Muthuswamy SK, Brugge JS. Organoid Cultures for the Study of Mammary Biology and Breast Cancer: The Promise and Challenges. Cold Spring Harb Perspect Med 2024; 14:a041661. [PMID: 38110241 PMCID: PMC11216180 DOI: 10.1101/cshperspect.a041661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
During the last decade, biomedical research has experienced a resurgence in the use of three-dimensional culture models for studies of normal and cancer biology. This resurgence has been driven by the development of models in which primary cells are grown in tissue-mimicking media and extracellular matrices to create organoid or organotypic cultures that more faithfully replicate the complex architecture and physiology of normal tissues and tumors. In addition, patient-derived tumor organoids preserve the three-dimensional organization and characteristics of the patient tumors ex vivo, becoming excellent preclinical models to supplement studies of tumor xenografts transplanted into immunocompromised mice. In this perspective, we provide an overview of how organoids are being used to investigate normal mammary biology and as preclinical models of breast cancer and discuss improvements that would enhance their utility and relevance to the field.
Collapse
Affiliation(s)
- Senthil K Muthuswamy
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland 20894, USA
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Ludwig Center at Harvard, Harvard Medical School Boston, Boston, Massachusetts 02115, USA
| |
Collapse
|
12
|
Huang K, Li M, Li Q, Chen Z, Zhang Y, Gu Z. Image-based profiling and deep learning reveal morphological heterogeneity of colorectal cancer organoids. Comput Biol Med 2024; 173:108322. [PMID: 38554658 DOI: 10.1016/j.compbiomed.2024.108322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 04/02/2024]
Abstract
Patient-derived organoids have proven to be a highly relevant model for evaluating of disease mechanisms and drug efficacies, as they closely recapitulate in vivo physiology. Colorectal cancer organoids, specifically, exhibit a diverse range of morphologies, which have been analyzed with image-based profiling. However, the relationship between morphological subtypes and functional parameters of the organoids remains underexplored. Here, we identified two distinct morphological subtypes ("cystic" and "solid") across 31360 bright field images using image-based profiling, which correlated differently with viability and apoptosis level of colorectal cancer organoids. Leveraging object detection neural networks, we were able to categorize single organoids achieving higher viability scores as "cystic" than "solid" subtype. Furthermore, a deep generative model was proposed to predict apoptosis intensity based on a apoptosis-featured dataset encompassing over 17000 bright field and matched fluorescent images. Notably, a significant correlation of 0.91 between the predicted value and ground truth was achived, underscoring the feasibility of this generative model as a potential means for assessing organoid functional parameters. The underlying cellular heterogeneity of the organoids, i.e., conserved colonic cell types and rare immune components, was also verified with scRNA sequencing, implying a compromised tumor microenvironment. Additionally, the "cystic" subtype was identified as a relapse phenotype featuring intestinal stem cell signatures, suggesting that this visually discernible relapse phenotype shows potential as a novel biomarker for colorectal cancer diagnosis and prognosis. In summary, our findings demonstrate that the morphological heterogeneity of colorectal cancer organoids explicitly recapitulate the association of phenotypic features and exogenous perturbations through the image-based profiling, providing new insights into disease mechanisms.
Collapse
Affiliation(s)
- Kai Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Mingyue Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Qiwei Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China
| | - Zaozao Chen
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu, 215163, China.
| | - Ying Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
13
|
Yuan X, Hao X, Chan HL, Zhao N, Pedroza DA, Liu F, Le K, Smith AJ, Calderon SJ, Lieu N, Soth MJ, Jones P, Zhang XHF, Rosen JM. CBP/P300 BRD Inhibition Reduces Neutrophil Accumulation and Activates Antitumor Immunity in TNBC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.590983. [PMID: 38712292 PMCID: PMC11071628 DOI: 10.1101/2024.04.25.590983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Tumor-associated neutrophils (TANs) have been shown to promote immunosuppression and tumor progression, and a high TAN frequency predicts poor prognosis in triple-negative breast cancer (TNBC). Dysregulation of CREB binding protein (CBP)/P300 function has been observed with multiple cancer types. The bromodomain (BRD) of CBP/P300 has been shown to regulate its activity. In this study, we found that IACS-70654, a novel and selective CBP/P300 BRD inhibitor, reduced TANs and inhibited the growth of neutrophil-enriched TNBC models. In the bone marrow, CBP/P300 BRD inhibition reduced the tumor-driven abnormal differentiation and proliferation of neutrophil progenitors. Inhibition of CBP/P300 BRD also stimulated the immune response by inducing an IFN response and MHCI expression in tumor cells and increasing tumor-infiltrated CTLs. Moreover, IACS-70654 improved the response of a neutrophil-enriched TNBC model to docetaxel and immune checkpoint blockade. This provides a rationale for combining a CBP/P300 BRD inhibitor with standard-of-care therapies in future clinical trials for neutrophil-enriched TNBC.
Collapse
Affiliation(s)
- Xueying Yuan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Na Zhao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Diego A Pedroza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Kang Le
- Institute for Applied Cancer Science (IACS), the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex J Smith
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Sebastian J Calderon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nadia Lieu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Michael J Soth
- Institute for Applied Cancer Science (IACS), the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip Jones
- Institute for Applied Cancer Science (IACS), the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Hong T, Xing J. Data- and theory-driven approaches for understanding paths of epithelial-mesenchymal transition. Genesis 2024; 62:e23591. [PMID: 38553870 PMCID: PMC11017362 DOI: 10.1002/dvg.23591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/16/2024] [Accepted: 03/16/2024] [Indexed: 04/02/2024]
Abstract
Reversible transitions between epithelial and mesenchymal cell states are a crucial form of epithelial plasticity for development and disease progression. Recent experimental data and mechanistic models showed multiple intermediate epithelial-mesenchymal transition (EMT) states as well as trajectories of EMT underpinned by complex gene regulatory networks. In this review, we summarize recent progress in quantifying EMT and characterizing EMT paths with computational methods and quantitative experiments including omics-level measurements. We provide perspectives on how these studies can help relating fundamental cell biology to physiological and pathological outcomes of EMT.
Collapse
Affiliation(s)
- Tian Hong
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, Knoxville TN, USA
| | - Jianhua Xing
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Ma X, Wang Q, Li G, Li H, Xu S, Pang D. Cancer organoids: A platform in basic and translational research. Genes Dis 2024; 11:614-632. [PMID: 37692477 PMCID: PMC10491878 DOI: 10.1016/j.gendis.2023.02.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 02/16/2023] [Indexed: 09/12/2023] Open
Abstract
An accumulation of previous work has established organoids as good preclinical models of human tumors, facilitating translation from basic research to clinical practice. They are changing the paradigm of preclinical cancer research because they can recapitulate the heterogeneity and pathophysiology of human cancers and more closely approximate the complex tissue environment and structure found in clinical tumors than in vitro cell lines and animal models. However, the potential applications of cancer organoids remain to be comprehensively summarized. In the review, we firstly describe what is currently known about cancer organoid culture and then discuss in depth the basic mechanisms, including tumorigenesis and tumor metastasis, and describe recent advances in patient-derived tumor organoids (PDOs) for drug screening and immunological studies. Finally, the present challenges faced by organoid technology in clinical practice and its prospects are discussed. This review highlights that organoids may offer a novel therapeutic strategy for cancer research.
Collapse
Affiliation(s)
- Xin Ma
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Qin Wang
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150086, China
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Guozheng Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Hui Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Shouping Xu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150086, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
- Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang 150086, China
| |
Collapse
|
16
|
Soragni C, Queiroz K, Ng CP, Stok A, Olivier T, Tzagkaraki D, Heijmans J, Suijker J, de Ruiter SPM, Olczyk A, Bokkers M, Schavemaker F, Trietsch SJ, Lanz HL, Vulto P, Joore J. Phenotypic screening in Organ-on-a-Chip systems: a 1537 kinase inhibitor library screen on a 3D angiogenesis assay. Angiogenesis 2024; 27:37-49. [PMID: 37493987 PMCID: PMC10881651 DOI: 10.1007/s10456-023-09888-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023]
Abstract
Modern drug development increasingly requires comprehensive models that can be utilized in the earliest stages of compound and target discovery. Here we report a phenotypic screening exercise in a high-throughput Organ-on-a-Chip setup. We assessed the inhibitory effect of 1537 protein kinase inhibitors in an angiogenesis assay. Over 4000 micro-vessels were grown under perfusion flow in microfluidic chips, exposed to a cocktail of pro-angiogenic factors and subsequently exposed to the respective kinase inhibitors. Efficacy of compounds was evaluated by reduced angiogenic sprouting, whereas reduced integrity of the main micro-vessel was taken as a measure for toxicity. The screen yielded 53 hits with high anti-angiogenicity and low toxicity, of which 44 were previously unassociated with angiogenic pathways. This study demonstrates that Organ-on-a-Chip models can be screened in high numbers to identify novel compounds and targets. This will ultimately reduce bias in early-stage drug development and increases probability to identify first in class compounds and targets for today's intractable diseases.
Collapse
Affiliation(s)
- Camilla Soragni
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
- Department of Cardiology, Maastricht University, Maastricht, The Netherlands
| | - Karla Queiroz
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Chee Ping Ng
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Arthur Stok
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Thomas Olivier
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Dora Tzagkaraki
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Jeroen Heijmans
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Johnny Suijker
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | | | | | - Marleen Bokkers
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | | | | | | | - Paul Vulto
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Jos Joore
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands.
| |
Collapse
|
17
|
Wu W, Peng Y, Xu M, Yan T, Zhang D, Chen Y, Mei K, Chen Q, Wang X, Qiao Z, Wang C, Wu S, Zhang Q. Deep-Learning-Based Nanomechanical Vibration for Rapid and Label-Free Assay of Epithelial Mesenchymal Transition. ACS NANO 2024; 18:3480-3496. [PMID: 38169507 DOI: 10.1021/acsnano.3c10811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Cancer is a profound danger to our life and health. The classification and related studies of epithelial and mesenchymal phenotypes of cancer cells are key scientific questions in cancer research. Here, we investigated cancer cell colonies from a mechanical perspective and developed an assay for classifying epithelial/mesenchymal cancer cell colonies using the biomechanical fingerprint in the form of "nanovibration" in combination with deep learning. The classification method requires only 1 s of vibration data and has a classification accuracy of nearly 92.5%. The method has also been validated for the screening of anticancer drugs. Compared with traditional methods, the method has the advantages of being nondestructive, label-free, and highly sensitive. Furthermore, we proposed a perspective that subcellular structure influences the amplitude and spectrum of nanovibrations and demonstrated it using experiments and numerical simulation. These findings allow internal changes in the cell colony to be manifested by nanovibrations. This work provides a perspective and an ancillary method for cancer cell phenotype diagnosis and promotes the study of biomechanical mechanisms of cancer progression.
Collapse
Affiliation(s)
- Wenjie Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Yongpei Peng
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Mengjun Xu
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Tianhao Yan
- Department of Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, People's Republic of China
| | - Duo Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Ye Chen
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Kainan Mei
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Qiubo Chen
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Xiapeng Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Zihan Qiao
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Chen Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Shangquan Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | - Qingchuan Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| |
Collapse
|
18
|
Adewunmi O, Shen Y, Zhang XHF, Rosen JM. Targeted Inhibition of lncRNA Malat1 Alters the Tumor Immune Microenvironment in Preclinical Syngeneic Mouse Models of Triple-Negative Breast Cancer. Cancer Immunol Res 2023; 11:1462-1479. [PMID: 37603945 PMCID: PMC10618655 DOI: 10.1158/2326-6066.cir-23-0045] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/18/2023] [Accepted: 08/17/2023] [Indexed: 08/23/2023]
Abstract
Long noncoding RNAs (lncRNA) play an important role in gene regulation in both normal tissues and cancer. Targeting lncRNAs is a promising therapeutic approach that has become feasible through the development of gapmer antisense oligonucleotides (ASO). Metastasis-associated lung adenocarcinoma transcript (Malat1) is an abundant lncRNA whose expression is upregulated in several cancers. Although Malat1 increases the migratory and invasive properties of tumor cells, its role in the tumor microenvironment (TME) is still not well defined. We explored the connection between Malat1 and the tumor immune microenvironment (TIME) using several immune-competent preclinical syngeneic Tp53-null triple-negative breast cancer (TNBC) mouse models that mimic the heterogeneity and immunosuppressive TME found in human breast cancer. Using a Malat1 ASO, we were able to knockdown Malat1 RNA expression resulting in a delay in primary tumor growth, decreased proliferation, and increased apoptosis. In addition, immunophenotyping of tumor-infiltrating lymphocytes revealed that Malat1 inhibition altered the TIME, with a decrease in immunosuppressive tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) as well as an increase in cytotoxic CD8+ T cells. Malat1 depletion in tumor cells, TAMs, and MDSCs decreased immunosuppressive cytokine/chemokine secretion whereas Malat1 inhibition in T cells increased inflammatory secretions and T-cell proliferation. Combination of a Malat1 ASO with chemotherapy or immune checkpoint blockade (ICB) improved the treatment responses in a preclinical model. These studies highlight the immunostimulatory effects of Malat1 inhibition in TNBC, the benefit of a Malat1 ASO therapeutic, and its potential use in combination with chemotherapies and immunotherapies.
Collapse
Affiliation(s)
- Oluwatoyosi Adewunmi
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas
| | - Yichao Shen
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Xiang H.-F. Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Lester and Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jeffrey M. Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
19
|
Voutsadakis IA. Molecular Characteristics and Therapeutic Vulnerabilities of Claudin-low Breast Cancers Derived from Cell Line Models. Cancer Genomics Proteomics 2023; 20:539-555. [PMID: 37889067 PMCID: PMC10614063 DOI: 10.21873/cgp.20404] [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: 06/25/2023] [Revised: 08/16/2023] [Accepted: 08/28/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND/AIM Breast cancers constitute heterogeneous tumor groups and their categorization in subtypes based on the expression of the estrogen (ER), progesterone (PR) and HER2 receptors has advanced therapeutics. Claudin-low breast cancer has been proposed as an additional subtype which is mostly ER, PR and HER2 negative, but its identification has not led to corresponding specific treatments yet. MATERIALS AND METHODS Breast cancer cell lines from the Cancer Cell Line Encyclopedia (CCLE) were assessed for mRNA suppression of claudins and mRNA expression of ER and ERBB2 (the gene encoding HER2). The set of identified claudin-low cell lines were compared with representative ER-/ERBB2- cell lines for associated molecular alterations, gene dependencies through CRISPR and microRNA arrays and in vitro drug sensitivities using the Genomics of Drug Sensitivity in Cancer (GDSC) project. RESULTS Claudin-low cell lines display up-regulation of mRNA expression of epithelial to mesenchymal transition (EMT) regulators. Methylation sensitive genes are down-regulated in claudin-low lines compared with other cell lines, without associated up-regulation of DNA methyltransferases. Dependency screen microarrays reveal dependencies of claudin-low cell lines on components of the cytoskeleton but no consistent dependencies in known oncogenes or tumor suppressors. Potential drug sensitivities revealed in the drug screens included sensitivities to WNT pathway modulators, tyrosine kinase cascade inhibitors and BET inhibitors. On the other hand, claudin-low cell lines showed resistance to deacetylase inhibitors. CONCLUSION Claudin-low cell line models duplicate features of claudin-low breast cancers and may serve as guides for identification of drugs worth exploring for further development.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste Marie, ON, Canada;
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
20
|
Volmer LL, Önder CE, Volz B, Singh AR, Brucker SY, Engler T, Hartkopf AD, Koch A. Microfluidic Isolation of Disseminated Tumor Cells from the Bone Marrow of Breast Cancer Patients. Int J Mol Sci 2023; 24:13930. [PMID: 37762233 PMCID: PMC10531360 DOI: 10.3390/ijms241813930] [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: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Disseminated tumor cells (DTCs) in the bone marrow (BM) of breast cancer (BC) patients are putative precursors of metastatic disease, and their presence is associated with an adverse clinical outcome. To achieve the personalization of therapy on a clinical routine level, the characterization of DTCs and in vitro drug testing on DTCs are of great interest. Therefore, biobanking methods, as well as novel approaches to DTC isolation, need to be developed. In this study, we established a protocol for the biobanking of BM samples and evaluated a microfluidic-based separation system (Parsortix®) for the enrichment of cryopreserved DTCs. We were able to successfully isolate viable DTCs after the prior cryopreservation of BM samples. We calculated a significant increase of up to 90-fold in harvested DTCs with the proposed method compared to the current standard techniques, opening up new analysis possibilities for DTCs. Our advanced method further presents options for 3D DTC cultures, enabling the individualized testing of targeted therapies for BC patients. In conclusion, we present a novel approach for DTC enrichment, with possibilities for future clinical implications.
Collapse
Affiliation(s)
- Léa L. Volmer
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Cansu E. Önder
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Barbara Volz
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Anjali R. Singh
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Sara Y. Brucker
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Tobias Engler
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Andreas D. Hartkopf
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - André Koch
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
21
|
Lei J, Pan Y, Gao R, He B, Wang Z, Lei X, Zhang Z, Yang N, Yan M. Rutaecarpine induces the differentiation of triple-negative breast cancer cells through inhibiting fumarate hydratase. J Transl Med 2023; 21:553. [PMID: 37592347 PMCID: PMC10436383 DOI: 10.1186/s12967-023-04396-w] [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: 05/10/2023] [Accepted: 07/29/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the most aggressive human cancers and has poor prognosis. Approximately 80% of TNBC cases belong to the molecular basal-like subtype, which can be exploited therapeutically by inducing differentiation. However, the strategies for inducing the differentiation of TNBC remain underexplored. METHODS A three-dimensional (3D) morphological screening model based on a natural compound library was used to identify possible candidate compounds that can induce TNBC cell differentiation. The efficacy of rutaecarpine was verified using assays: RT-qPCR, RNA-seq, flow cytometry, immunofluorescence, SCENITH and label-free LC-MS/MS. The direct targets of rutaecarpine were identified through drug affinity responsive target stability (DARTS) assay. A xenograft mice model was also constructed to confirm the effect of rutaecarpine in vivo. RESULTS We identified that rutaecarpine, an indolopyridoquinazolinone, induces luminal differentiation of basal TNBC cells in both 3D spheroids and in vivo mice models. Mechanistically, rutaecarpine treatment leads to global metabolic stress and elevated ROS in 3D cultured TNBC cells. Moreover, NAC, a scavenger of ROS, impedes rutaecarpine-induced differentiation of TNBC cells in 3D culture. Finally, we identified fumarate hydratase (FH) as the direct interacting target of rutaecarpine. The inhibition of FH and the knockdown of FH consistently induced the differentiation of TNBC cells in 3D culture. CONCLUSIONS Our results provide a platform for differentiation therapy drug discovery using 3D culture models and identify rutaecarpine as a potential compound for TNBC treatment.
Collapse
Affiliation(s)
- Jie Lei
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yujia Pan
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116023, China
| | - Rui Gao
- Department of Medical Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 510275, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xinxing Lei
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zijian Zhang
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China.
| | - Min Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
22
|
Reisenauer KN, Aroujo J, Tao Y, Ranganathan S, Romo D, Taube JH. Therapeutic vulnerabilities of cancer stem cells and effects of natural products. Nat Prod Rep 2023; 40:1432-1456. [PMID: 37103550 PMCID: PMC10524555 DOI: 10.1039/d3np00002h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Covering: 1995 to 2022Tumors possess both genetic and phenotypic heterogeneity leading to the survival of subpopulations post-treatment. The term cancer stem cells (CSCs) describes a subpopulation that is resistant to many types of chemotherapy and which also possess enhanced migratory and anchorage-independent growth capabilities. These cells are enriched in residual tumor material post-treatment and can serve as the seed for future tumor re-growth, at both primary and metastatic sites. Elimination of CSCs is a key goal in enhancing cancer treatment and may be aided by application of natural products in conjunction with conventional treatments. In this review, we highlight molecular features of CSCs and discuss synthesis, structure-activity relationships, derivatization, and effects of six natural products with anti-CSC activity.
Collapse
Affiliation(s)
| | - Jaquelin Aroujo
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | - Yongfeng Tao
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | | | - Daniel Romo
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | - Joseph H Taube
- Department of Biology, Baylor University, Waco, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
23
|
Li C, Holman JB, Shi Z, Qiu B, Ding W. On-chip modeling of tumor evolution: Advances, challenges and opportunities. Mater Today Bio 2023; 21:100724. [PMID: 37483380 PMCID: PMC10359640 DOI: 10.1016/j.mtbio.2023.100724] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Tumor evolution is the accumulation of various tumor cell behaviors from tumorigenesis to tumor metastasis and is regulated by the tumor microenvironment (TME). However, the mechanism of solid tumor progression has not been completely elucidated, and thus, the development of tumor therapy is still limited. Recently, Tumor chips constructed by culturing tumor cells and stromal cells on microfluidic chips have demonstrated great potential in modeling solid tumors and visualizing tumor cell behaviors to exploit tumor progression. Herein, we review the methods of developing engineered solid tumors on microfluidic chips in terms of tumor types, cell resources and patterns, the extracellular matrix and the components of the TME, and summarize the recent advances of microfluidic chips in demonstrating tumor cell behaviors, including proliferation, epithelial-to-mesenchymal transition, migration, intravasation, extravasation and immune escape of tumor cells. We also outline the combination of tumor organoids and microfluidic chips to elaborate tumor organoid-on-a-chip platforms, as well as the practical limitations that must be overcome.
Collapse
Affiliation(s)
- Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Joseph Benjamin Holman
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Weiping Ding
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| |
Collapse
|
24
|
Singh S, Saini H, Sharma A, Gupta S, Huddar VG, Tripathi R. Breast cancer: miRNAs monitoring chemoresistance and systemic therapy. Front Oncol 2023; 13:1155254. [PMID: 37397377 PMCID: PMC10312137 DOI: 10.3389/fonc.2023.1155254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
With a high mortality rate that accounts for millions of cancer-related deaths each year, breast cancer is the second most common malignancy in women. Chemotherapy has significant potential in the prevention and spreading of breast cancer; however, drug resistance often hinders therapy in breast cancer patients. The identification and the use of novel molecular biomarkers, which can predict response to chemotherapy, might lead to tailoring breast cancer treatment. In this context, accumulating research has reported microRNAs (miRNAs) as potential biomarkers for early cancer detection, and are conducive to designing a more specific treatment plan by helping analyze drug resistance and sensitivity in breast cancer treatment. In this review, miRNAs are discussed in two alternative ways-as tumor suppressors to be used in miRNA replacement therapy to reduce oncogenesis and as oncomirs to lessen the translation of the target miRNA. Different miRNAs like miR-638, miR-17, miR-20b, miR-342, miR-484, miR-21, miR-24, miR-27, miR-23 and miR-200 are involved in the regulation of chemoresistance through diverse genetic targets. For instance, tumor-suppressing miRNAs like miR-342, miR-16, miR-214, and miR-128 and tumor-promoting miRNAs like miR101 and miR-106-25 cluster regulate the cell cycle, apoptosis, epithelial to mesenchymal transition and other pathways to impart breast cancer drug resistance. Hence, in this review, we have discussed the significance of miRNA biomarkers that could assist in providing novel therapeutic targets to overcome potential chemotherapy resistance to systemic therapy and further facilitate the design of tailored therapy for enhanced efficacy against breast cancer.
Collapse
Affiliation(s)
- Shivam Singh
- Department of Radiation Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Heena Saini
- Integrated translational Molecular Biology laboratory, Department of Rog Nidan and Vikriti vigyan (Pathology), All India Institute of Ayurveda (AIIA), New Delhi, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhash Gupta
- Department of Radiation Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - V. G. Huddar
- Department of Kaya Chikitsa (Internal Medicine), All India Institute of Ayurveda (AIIA), New Delhi, India
| | - Richa Tripathi
- Integrated translational Molecular Biology laboratory, Department of Rog Nidan and Vikriti vigyan (Pathology), All India Institute of Ayurveda (AIIA), New Delhi, India
| |
Collapse
|
25
|
Gu H, Shi R, Xu C, Lv W, Hu X, Xu C, Pan Y, He X, Wu A, Li J. EGFR-Targeted Liposomes Combined with Ginsenoside Rh2 Inhibit Triple-Negative Breast Cancer Growth and Metastasis. Bioconjug Chem 2023. [PMID: 37235785 DOI: 10.1021/acs.bioconjchem.3c00207] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Triple-negative breast cancer (TNBC) remains the most challenging breast cancer subtype due to its lack of targeted therapies and poor prognosis. In order to treat patients with these tumors, efforts have been made to explore feasible targets. Epidermal growth factor receptor (EGFR)-targeted therapy is currently in clinical trials and regarded to be a promising treatment strategy. In this study, an EGFR-targeting nanoliposome (LTL@Rh2@Lipo-GE11) using ginsenoside Rh2 as a wall material was developed, in which GE11 was used as the EGFR-binding peptide to deliver more ginsenoside Rh2 and luteolin into TNBC. In comparison to non-targeted liposomes (Rh2@Lipo and LTL@Rh2@Lipo), the nanoliposomes LTL@Rh2@Lipo-GE11 demonstrated a high specificity to MDA-MB-231 cells that expressed a high level of EGFR both in vitro and in vivo, contributing to the strong inhibitory effects on the growth and migration of TNBC. These results suggest that LTL@Rh2@Lipo-GE11 is a prospective candidate for targeted therapy of TNBC, with a remarkable capability to inhibit tumor development and metastasis.
Collapse
Affiliation(s)
- Haiyan Gu
- Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Rui Shi
- Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China
| | - Chen Xu
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Wenhao Lv
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Xueyin Hu
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Canxin Xu
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Yuanbo Pan
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Xiahong He
- Laboratory for Forest Resources Conservation and Utilization in the Southwest Mountains of China, Ministry of Education, International Ecological Forestry Research Center of Kunming, Southwest Forestry University, Kunming 650224, China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Juan Li
- Cixi Institute of Biomedical Engineering, Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| |
Collapse
|
26
|
Van Hemelryk A, Erkens-Schulze S, Lim L, de Ridder CMA, Stuurman DC, Jenster GW, van Royen ME, van Weerden WM. Viability Analysis and High-Content Live-Cell Imaging for Drug Testing in Prostate Cancer Xenograft-Derived Organoids. Cells 2023; 12:1377. [PMID: 37408211 DOI: 10.3390/cells12101377] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 07/07/2023] Open
Abstract
Tumor organoids have been pushed forward as advanced model systems for in vitro oncology drug testing, with the eventual goal to direct personalized cancer treatments. However, drug testing efforts suffer from a large variation in experimental conditions for organoid culturing and organoid treatment. Moreover, most drug tests are restricted to whole-well viability as the sole read-out, thereby losing important information about key biological aspects that might be impacted due to the use of administered drugs. These bulk read-outs also discard potential inter-organoid heterogeneity in drug responses. To tackle these issues, we developed a systematic approach for processing organoids from prostate cancer (PCa) patient-derived xenografts (PDXs) for viability-based drug testing and identified essential conditions and quality checks for consistent results. In addition, we generated an imaging-based drug testing procedure using high-content fluorescence microscopy in living PCa organoids to detect various modalities of cell death. Individual organoids and cell nuclei in organoids were segmented and quantified using a dye combination of Hoechst 33342, propidium iodide and Caspase 3/7 Green, allowing the identification of cytostatic and cytotoxic treatment effects. Our procedures provide important insights into the mechanistic actions of tested drugs. Moreover, these methods can be adapted for tumor organoids originating from other cancer types to increase organoid-based drug test validity, and ultimately, accelerate clinical implementation.
Collapse
Affiliation(s)
- Annelies Van Hemelryk
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Sigrun Erkens-Schulze
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Lifani Lim
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Corrina M A de Ridder
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Debra C Stuurman
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Guido W Jenster
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Wytske M van Weerden
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
27
|
Lian B, Chen X, Shen K. Inhibition of histone deacetylases attenuates tumor progression and improves immunotherapy in breast cancer. Front Immunol 2023; 14:1164514. [PMID: 36969235 PMCID: PMC10034161 DOI: 10.3389/fimmu.2023.1164514] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Breast cancer is one of the common malignancies with poor prognosis worldwide. The treatment of breast cancer patients includes surgery, radiation, hormone therapy, chemotherapy, targeted drug therapy and immunotherapy. In recent years, immunotherapy has potentiated the survival of certain breast cancer patients; however, primary resistance or acquired resistance attenuate the therapeutic outcomes. Histone acetyltransferases induce histone acetylation on lysine residues, which can be reversed by histone deacetylases (HDACs). Dysregulation of HDACs via mutation and abnormal expression contributes to tumorigenesis and tumor progression. Numerous HDAC inhibitors have been developed and exhibited the potent anti-tumor activity in a variety of cancers, including breast cancer. HDAC inhibitors ameliorated immunotherapeutic efficacy in cancer patients. In this review, we discuss the anti-tumor activity of HDAC inhibitors in breast cancer, including dacinostat, belinostat, abexinostat, mocetinotat, panobinostat, romidepsin, entinostat, vorinostat, pracinostat, tubastatin A, trichostatin A, and tucidinostat. Moreover, we uncover the mechanisms of HDAC inhibitors in improving immunotherapy in breast cancer. Furthermore, we highlight that HDAC inhibitors might be potent agents to potentiate immunotherapy in breast cancer.
Collapse
Affiliation(s)
| | | | - Kunwei Shen
- *Correspondence: Xiaosong Chen, ; Kunwei Shen,
| |
Collapse
|
28
|
Chai C, Ji P, Xu H, Tang H, Wang Z, Zhang H, Zhou W. Targeting cancer drug resistance utilizing organoid technology. Biomed Pharmacother 2023; 158:114098. [PMID: 36528918 DOI: 10.1016/j.biopha.2022.114098] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer organoids generated from 3D in vitro cell cultures have contributed to the study of drug resistance. Maintenance of genomic and transcriptomic similarity between organoids and parental cancer allows organoids to have the ability of accurate prediction in drug resistance testing. Protocols of establishing therapy-sensitive and therapy-resistant organoids are concluded in two aspects, which are generated directly from respective patients' cancer and by induction of anti-cancer drug. Genomic and transcriptomic analyses and gene editing have been applied to organoid studies to identify key targets in drug resistance and FGFR3, KHDRBS3, lnc-RP11-536 K7.3 and FBN1 were found to be key targets. Furthermore, mechanisms contributing to resistance have been identified, including metabolic adaptation, activation of DNA damage response, defects in apoptosis, reduced cellular senescence, cellular plasticity, subpopulation interactions and gene fusions. Additionally, cancer stem cells (CSCs) have been verified to be involved in drug resistance utilizing organoid technology. Reversal of drug resistance can be achieved by targeting key genes and CSCs in cancer organoids. In this review, we summarize applications of organoids to cancer drug resistance research, indicating prospects and limitations.
Collapse
Affiliation(s)
- Changpeng Chai
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China; The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Pengfei Ji
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Hao Xu
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Huan Tang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Zhengfeng Wang
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Hui Zhang
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Wence Zhou
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou 730000, Gansu, China; The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| |
Collapse
|
29
|
Castaneda M, den Hollander P, Kuburich NA, Rosen JM, Mani SA. Mechanisms of cancer metastasis. Semin Cancer Biol 2022; 87:17-31. [PMID: 36354098 DOI: 10.1016/j.semcancer.2022.10.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/10/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
Abstract
Metastatic cancer is almost always terminal, and more than 90% of cancer deaths result from metastatic disease. Combating cancer metastasis and post-therapeutic recurrence successfully requires understanding each step of metastatic progression. This review describes the current state of knowledge of the etiology and mechanism of cancer progression from primary tumor growth to the formation of new tumors in other parts of the body. Open questions, avenues for future research, and therapeutic approaches with the potential to prevent or inhibit metastasis through personalization to each patient's mutation and/or immune profile are also highlighted.
Collapse
Affiliation(s)
- Maria Castaneda
- Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Petra den Hollander
- Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology and Lab Medicine, Brown University, Providence, RI 02912, USA; Legoretta Cancer Center, Brown University, Providence, RI 021912, USA
| | - Nick A Kuburich
- Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology and Lab Medicine, Brown University, Providence, RI 02912, USA; Legoretta Cancer Center, Brown University, Providence, RI 021912, USA
| | - Jeffrey M Rosen
- Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Sendurai A Mani
- Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology and Lab Medicine, Brown University, Providence, RI 02912, USA; Legoretta Cancer Center, Brown University, Providence, RI 021912, USA.
| |
Collapse
|
30
|
Shozu K, Kaneko S, Shinkai N, Dozen A, Kosuge H, Nakakido M, Machino H, Takasawa K, Asada K, Komatsu M, Tsumoto K, Ohnuma SI, Hamamoto R. Repression of the PRELP gene is relieved by histone deacetylase inhibitors through acetylation of histone H2B lysine 5 in bladder cancer. Clin Epigenetics 2022; 14:147. [PMID: 36371227 PMCID: PMC9656081 DOI: 10.1186/s13148-022-01370-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Proline/arginine-rich end leucine-rich repeat protein (PRELP) is a member of the small leucine-rich proteoglycan family of extracellular matrix proteins, which is markedly suppressed in the majority of early-stage epithelial cancers and plays a role in regulating the epithelial-mesenchymal transition by altering cell-cell adhesion. Although PRELP is an important factor in the development and progression of bladder cancer, the mechanism of PRELP gene repression remains unclear. RESULTS Here, we show that repression of PRELP mRNA expression in bladder cancer cells is alleviated by HDAC inhibitors (HDACi) through histone acetylation. Using ChIP-qPCR analysis, we found that acetylation of lysine residue 5 of histone H2B in the PRELP gene promoter region is a marker for the de-repression of PRELP expression. CONCLUSIONS These results suggest a mechanism through which HDACi may partially regulate the function of PRELP to suppress the development and progression of bladder cancer. Some HDACi are already in clinical use, and the findings of this study provide a mechanistic basis for further investigation of HDACi-based therapeutic strategies.
Collapse
Affiliation(s)
- Kanto Shozu
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.267346.20000 0001 2171 836XDepartment of Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | - Syuzo Kaneko
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.509456.bRIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Tokyo, Japan
| | - Norio Shinkai
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.509456.bRIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Tokyo, Japan ,grid.265073.50000 0001 1014 9130Department of NCC Cancer Science, Biomedical Science and Engineering Track, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ai Dozen
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan
| | - Hirofumi Kosuge
- grid.26999.3d0000 0001 2151 536XSchool of Engineering, The University of Tokyo, Tokyo, Japan
| | - Makoto Nakakido
- grid.26999.3d0000 0001 2151 536XSchool of Engineering, The University of Tokyo, Tokyo, Japan
| | - Hidenori Machino
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.509456.bRIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Tokyo, Japan
| | - Ken Takasawa
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.509456.bRIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Tokyo, Japan
| | - Ken Asada
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.509456.bRIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Tokyo, Japan
| | - Masaaki Komatsu
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.509456.bRIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Tokyo, Japan
| | - Kouhei Tsumoto
- grid.26999.3d0000 0001 2151 536XSchool of Engineering, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichi Ohnuma
- grid.83440.3b0000000121901201UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL UK ,grid.5335.00000000121885934Department of Oncology, The Hutchison/MRC Research Center, University of Cambridge, Hills Road, Cambridge, CB2 2XZ UK
| | - Ryuji Hamamoto
- grid.272242.30000 0001 2168 5385Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045 Japan ,grid.509456.bRIKEN Center for Advanced Intelligence Project, Cancer Translational Research Team, Tokyo, Japan
| |
Collapse
|
31
|
Advances in the Biology, Detection Techniques, and Clinical Applications of Circulating Tumor Cells. JOURNAL OF ONCOLOGY 2022; 2022:7149686. [PMID: 36090904 PMCID: PMC9462976 DOI: 10.1155/2022/7149686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/12/2022] [Accepted: 08/02/2022] [Indexed: 12/01/2022]
Abstract
Circulating tumor cells (CTCs) play a crucial role in tumor recurrence and metastasis, and their early detection has shown remarkable benefits in clinical theranostics. However, CTCs are extremely rare, thus detecting them in the blood is very challenging. New CTC detection techniques are continuously being developed, enabling deeper analysis of CTC biology and potential clinical application. This article reviews current CTC detection techniques and their clinical application. CTCs have provided, and will continue to provide, important insights into the process of metastasis, which could lead to development of new therapies for different cancers.
Collapse
|
32
|
Huang Z, Zhang Z, Zhou C, Liu L, Huang C. Epithelial–mesenchymal transition: The history, regulatory mechanism, and cancer therapeutic opportunities. MedComm (Beijing) 2022; 3:e144. [PMID: 35601657 PMCID: PMC9115588 DOI: 10.1002/mco2.144] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a program wherein epithelial cells lose their junctions and polarity while acquiring mesenchymal properties and invasive ability. Originally defined as an embryogenesis event, EMT has been recognized as a crucial process in tumor progression. During EMT, cell–cell junctions and cell–matrix attachments are disrupted, and the cytoskeleton is remodeled to enhance mobility of cells. This transition of phenotype is largely driven by a group of key transcription factors, typically Snail, Twist, and ZEB, through epigenetic repression of epithelial markers, transcriptional activation of matrix metalloproteinases, and reorganization of cytoskeleton. Mechanistically, EMT is orchestrated by multiple pathways, especially those involved in embryogenesis such as TGFβ, Wnt, Hedgehog, and Hippo, suggesting EMT as an intrinsic link between embryonic development and cancer progression. In addition, redox signaling has also emerged as critical EMT modulator. EMT confers cancer cells with increased metastatic potential and drug resistant capacity, which accounts for tumor recurrence in most clinic cases. Thus, targeting EMT can be a therapeutic option providing a chance of cure for cancer patients. Here, we introduce a brief history of EMT and summarize recent advances in understanding EMT mechanisms, as well as highlighting the therapeutic opportunities by targeting EMT in cancer treatment.
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Chengwei Zhou
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Lin Liu
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| |
Collapse
|
33
|
Tazawa H, Shigeyasu K, Noma K, Kagawa S, Sakurai F, Mizuguchi H, Kobayashi H, Imamura T, Fujiwara T. Tumor‐targeted fluorescence labeling systems for cancer diagnosis and treatment. Cancer Sci 2022; 113:1919-1929. [PMID: 35398956 PMCID: PMC9207361 DOI: 10.1111/cas.15369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/25/2022] [Accepted: 04/02/2022] [Indexed: 11/28/2022] Open
Abstract
Conventional imaging techniques are available for clinical identification of tumor sites. However, detecting metastatic tumor cells that are spreading from primary tumor sites using conventional imaging techniques remains difficult. In contrast, fluorescence‐based labeling systems are useful tools for detecting tumor cells at the single‐cell level in cancer research. The ability to detect fluorescent‐labeled tumor cells enables investigations of the biodistribution of tumor cells for the diagnosis and treatment of cancer. For example, the presence of fluorescent tumor cells in the peripheral blood of cancer patients is a predictive biomarker for early diagnosis of distant metastasis. The elimination of fluorescent tumor cells without damaging normal tissues is ideal for minimally invasive treatment of cancer. To capture fluorescent tumor cells within normal tissues, however, tumor‐specific activated target molecules are needed. This review focuses on recent advances in tumor‐targeted fluorescence labeling systems, in which indirect reporter labeling using tumor‐specific promoters is applied to fluorescence labeling of tumor cells for the diagnosis and treatment of cancer. Telomerase promoter‐dependent fluorescence labeling using replication‐competent viral vectors produces fluorescent proteins that can be used to detect and eliminate telomerase‐positive tumor cells. Tissue‐specific promoter‐dependent fluorescence labeling enables identification of specific tumor cells. Vimentin promoter‐dependent fluorescence labeling is a useful tool for identifying tumor cells that undergo epithelial–mesenchymal transition (EMT). The evaluation of tumor cells undergoing EMT is important for accurately assessing metastatic potential. Thus, tumor‐targeted fluorescence labeling systems represent novel platforms that enable the capture of tumor cells for the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Hiroshi Tazawa
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
- Center for Innovative Clinical Medicine Okayama University Hospital Okayama Japan
| | - Kunitoshi Shigeyasu
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
- Minimally Invasive Therapy Center Okayama University Hospital Okayama Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology Graduate School of Pharmaceutical Sciences Osaka University Osaka Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology Graduate School of Pharmaceutical Sciences Osaka University Osaka Japan
| | - Hisataka Kobayashi
- Molecular Imaging Branch Center for Cancer Research National Cancer Institute National Institutes of Health Bethesda MD USA
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis Ehime University Graduate School of Medicine Ehime Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
| |
Collapse
|
34
|
Powell RT, Moussalli MJ, Guo L, Bae G, Singh P, Stephan C, Shureiqi I, Davies PJ. deepOrganoid: A brightfield cell viability model for screening matrix-embedded organoids. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:175-184. [PMID: 35314378 DOI: 10.1016/j.slasd.2022.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
High-throughput viability screens are commonly used in the identification and development of chemotherapeutic drugs. These systems rely on the fidelity of the cellular model systems to recapitulate the drug response that occurs in vivo. In recent years, there has been an expansion in the utilization of patient-derived materials as well as advanced cell culture techniques, such as multi-cellular tumor organoids, to further enhance the translational relevance of cellular model systems. Simple quantitative analysis remains a challenge, primarily due to the difficulties of robust image segmentation in heterogenous 3D cultures. However, explicit segmentation is not required with the advancement of deep learning, and it can be used for both continuous (regression) or categorical classification problems. Deep learning approaches are additionally benefited by being fully data-driven and highly automatable, thus they can be established and run with minimal to no user-defined parameters. In this article, we describe the development and implementation of a regressive deep learning model trained on brightfield images of patient-derived organoids and use the terminal viability readout (CellTiter-Glo) as training labels. Ultimately, this has led to the generation of a non-invasive and label-free tool to evaluate changes in organoid viability.
Collapse
Affiliation(s)
- Reid T Powell
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States.
| | - Micheline J Moussalli
- Department of Gastrointestinal Medical Oncology, UT MDACC, Houston, TX, United States
| | - Lei Guo
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Goeun Bae
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Pankaj Singh
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Clifford Stephan
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Imad Shureiqi
- Department of Gastrointestinal Medical Oncology, UT MDACC, Houston, TX, United States
| | - Peter J Davies
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| |
Collapse
|
35
|
Raue R, Frank AC, Fuhrmann DC, de la Cruz-Ojeda P, Rösser S, Bauer R, Cardamone G, Weigert A, Syed SN, Schmid T, Brüne B. MicroRNA-200c Attenuates the Tumor-Infiltrating Capacity of Macrophages. BIOLOGY 2022; 11:biology11030349. [PMID: 35336722 PMCID: PMC8945044 DOI: 10.3390/biology11030349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022]
Abstract
Simple Summary The tumor microenvironment determines the prognosis and outcome for cancer patients. Herein, tumor-associated macrophages are not only highly abundant, but also play a crucial role in shaping a tumor-supporting microenvironment. Both their recruitment to the tumor as well as their functional polarization toward a pro-tumorigenic phenotype are mediated by tumor-derived factors including microRNAs. However, the impact of most microRNAs on the tumor cell-macrophage crosstalk remains to be elucidated. Thus, we reached out to investigate the role of hsa-miR-200c-3p (miR-200c) in tumor cell–macrophage interactions, as it was shown to be differentially expressed during cancer progression and metastasis. miR-200c was highly expressed in MCF7 breast tumor cells compared to macrophages. Furthermore, we identified a CD36-dependent uptake of miR-200c, derived from apoptotic tumor cells, into macrophages. In macrophages, elevated miR-200c levels reduced the expression of numerous migration-associated mRNAs, consequently reducing the capacity of macrophages to infiltrate into tumor spheroids. Finally, a distinct signature of miR-200c-repressed, predicted targets was identified, which strongly correlated with tumor infiltration. Targeting the miR-200c transfer from dying tumor cells to macrophages might therefore provide the opportunity to specifically modulate tumor-associated macrophage recruitment. Abstract Macrophages constitute a major part of the tumor-infiltrating immune cells. Within the tumor microenvironment, they acquire an alternatively activated, tumor-supporting phenotype. Factors released by tumor cells are crucial for the recruitment of tumor-associated macrophages. In the present project, we aimed to understand the role of hsa-miR-200c-3p (miR-200c) in the interplay between tumor cells and macrophages. To this end, we employed a coculture system of MCF7 breast tumor cells and primary human macrophages and observed the transfer of miR-200c from apoptotic tumor cells to macrophages, which required intact CD36 receptor in macrophages. We further comprehensively determined miR-200c targets in macrophages by mRNA-sequencing and identified numerous migration-associated mRNAs to be downregulated by miR-200c. Consequently, miR-200c attenuated macrophage infiltration into 3-dimensional tumor spheroids. miR-200c-mediated reduction in infiltration further correlated with a miR-200c migration signature comprised of the four miR-200c-repressed, predicted targets PPM1F, RAB11FIB2, RDX, and MSN.
Collapse
Affiliation(s)
- Rebecca Raue
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
| | - Ann-Christin Frank
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
| | - Dominik C. Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
| | - Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain;
| | - Silvia Rösser
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
| | - Rebekka Bauer
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
| | - Giulia Cardamone
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
| | - Shahzad Nawaz Syed
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
- Correspondence:
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (R.R.); (A.-C.F.); (D.C.F.); (S.R.); (R.B.); (G.C.); (A.W.); (S.N.S.); (B.B.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, 60596 Frankfurt, Germany
| |
Collapse
|
36
|
Huang PS, Liao CJ, Huang YH, Yeh CT, Chen CY, Tang HC, Chang CC, Lin KH. Functional and Clinical Significance of Dysregulated microRNAs in Liver Cancer. Cancers (Basel) 2021; 13:5361. [PMID: 34771525 PMCID: PMC8582514 DOI: 10.3390/cancers13215361] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 11/27/2022] Open
Abstract
Liver cancer is the leading cause of cancer-related mortality in the world. This mainly reflects the lack of early diagnosis tools and effective treatment methods. MicroRNAs (miRNAs) are a class of non-transcribed RNAs, some of which play important regulatory roles in liver cancer. Here, we discuss microRNAs with key impacts on liver cancer, such as miR-122, miR-21, miR-214, and miR-199. These microRNAs participate in various physiological regulatory pathways of liver cancer cells, and their modulation can have non-negligible effects in the treatment of liver cancer. We discuss whether these microRNAs can be used for better clinical diagnosis and/or drug development. With the advent of novel technologies, fast, inexpensive, and non-invasive RNA-based biomarker research has become a new mainstream approach. However, the clinical application of microRNA-based markers has been limited by the high sequence similarity among them and the potential for off-target problems. Therefore, researchers particularly value microRNAs that are specific to or have special functions in liver cancer. These include miR-122, which is specifically expressed in the liver, and miR-34, which is necessary for the replication of the hepatitis C virus in liver cancer. Clinical treatment drugs have been developed based on miR-34 and miR-122 (MRX34 and Miravirsen, respectively), but their side effects have not yet been overcome. Future research is needed to address these weaknesses and establish a feasible microRNA-based treatment strategy for liver cancer.
Collapse
Affiliation(s)
- Po-Shuan Huang
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (P.-S.H.); (C.-J.L.)
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Chia-Jung Liao
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (P.-S.H.); (C.-J.L.)
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Ya-Hui Huang
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (Y.-H.H.); (C.-T.Y.)
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (Y.-H.H.); (C.-T.Y.)
| | - Cheng-Yi Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Hui-Chi Tang
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Cheng-Chih Chang
- Department of General Surgery, Chang Gung Memorial Hospital at Chia-yi, Chia-yi 613, Taiwan
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (P.-S.H.); (C.-J.L.)
- Department of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (Y.-H.H.); (C.-T.Y.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| |
Collapse
|