1
|
Liu H, Tao T, Gan Z, Xie Y, Wang Y, Yang Y, Zhang X, Li X, Qin J. Organoid in droplet: Production of uniform pancreatic cancer organoids from single cells. Mater Today Bio 2025; 32:101765. [PMID: 40270893 PMCID: PMC12017920 DOI: 10.1016/j.mtbio.2025.101765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/06/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
Cancer organoids have improved our understanding of recapitulating the histology, genotypes, and drug response of patient tumors for personalized medicine. However, the existing cancer organoids are typically grown in animal-derived matrices (e.g., Matrigel), which suffer from poor reproducibility and low throughput due to uncontrollable origin of seed cells, undefined matrix, and manual manipulation. Here, we report a new strategy to massively generate uniform pancreatic cancer organoids (PCOs) in a droplet system from single cells. This system is composed of all-in-water fluids that allow to mildly encapsulate single tumor cell into isolated droplet, which subsequently proliferate and self-assemble into organoids, resembling the initial state of a tumor in the body. This high-throughput method can produce thousands of organoids in a single batch. The droplets can serve as templates for synthesizing defined microgels with proper stiffness similar to that of native tumors, facilitating functional expressions of PCOs. These organoids exhibit superior uniformity and controllability in terms of size and morphologies compared with organoids cultured in manually dropped Matrigel, due mainly to the controllable number of initiating cells and defined microgels. In addition, the established organoids maintain the key biomarkers of pancreatic tumor (e.g., KRT7, KRT19 and SOX9) and higher expression of genes associated with drug metabolism confirmed by RNA-seq and PCR analysis. Furthermore, they show distinguishing responses to four clinically used drugs in a reproducible manner in automatic pipetting workstation, indicating the feasibility of the proposed method in high-throughput drug testing. The established strategy has integrated the formation, 3D cultures, and analysis of PCOs derived from single cells in a whole system, which may provide a novel platform for advancing organoids research with standardized procedure in translational applications.
Collapse
Affiliation(s)
- Haitao Liu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Tingting Tao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhongqiao Gan
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingying Xie
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqing Wang
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Yizhao Yang
- Imperial College London, White City Campus, London, W12 0BZ, UK
| | - Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Xianliang Li
- Department of HBP Surgery, Beijing Chao Yang Hospital, the Capital Medical University, Beijing, 100020, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| |
Collapse
|
2
|
Wu Y, Zhang F, Du F, Huang J, Wei S. Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review). Mol Med Rep 2025; 31:140. [PMID: 40183402 PMCID: PMC11976518 DOI: 10.3892/mmr.2025.13505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Malignant tumors notably decrease life expectancy. Despite advances in cancer diagnosis and treatment, the mechanisms underlying tumorigenesis, progression and drug resistance have not been fully elucidated. An emerging method to study tumors is tumor organoids, which are a three‑dimensional miniature structure. These retain the patient‑specific tumor heterogeneity while demonstrating the histological, genetic and molecular features of original tumors. Compared with conventional cancer cell lines and animal models, patient‑derived tumor organoids are more advanced at physiological and clinical levels. Their synergistic combination with other technologies, such as organ‑on‑a‑chip, 3D‑bioprinting, tissue‑engineered cell scaffolds and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR‑associated protein 9, may overcome limitations of the conventional 3D organoid culture and result in the development of more appropriate model systems that preserve the complex tumor stroma, inter‑organ and intra‑organ communications. The present review summarizes the evolution of tumor organoids and their combination with advanced technologies, as well as the application of tumor organoids in basic and clinical research.
Collapse
Affiliation(s)
- Ying Wu
- Department of Obstetrics and Gynecology, The 920th Hospital of Joint Logistics Support Force, Kunming, Yunnan 650032, P.R. China
| | - Fan Zhang
- Department of Comprehensive Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| | - Furong Du
- Department of Medicine, Kingbio Medical Co., Ltd., Chongqing 401123, P.R. China
| | - Juan Huang
- Department of Breast Surgery and Multidisciplinary Breast Cancer Center, Clinical Research Center of Breast Cancer in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shuqing Wei
- Department of Comprehensive Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| |
Collapse
|
3
|
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
|
4
|
Schwerd‐Kleine P, Würth R, Cheytan T, Michel L, Thewes V, Gutjahr E, Seker‐Cin H, Kazdal D, Neuberth S, Thiel V, Schwickert J, Vorberg T, Wischhusen J, Stenzinger A, Zapatka M, Lichter P, Schneeweiss A, Trumpp A, Sprick MR. Biopsy-derived organoids in personalised early breast cancer care: Challenges of tumour purity and normal cell overgrowth cap their practical utility. Int J Cancer 2025; 156:2200-2209. [PMID: 40022208 PMCID: PMC11970545 DOI: 10.1002/ijc.35386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 12/05/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
The ability to establish organoids composed exclusively of tumour rather than healthy cells is essential for their implementation into clinical practice. Organoids have recently emerged as a powerful tool to expand patient material in culture and generate modifiable 3D models derived from humans or animal models. For translational research, they enable the creation of model systems for an ever-increasing number of cell types and diseases. And in personalised medicine, they potentially allow for functional drug testing with high predictive power in certain settings. We found that using biopsy material from untreated, early-stage primary breast cancer patients poses significant challenges for consistently culturing tumour cells as organoids. Specifically, we observed frequent outgrowth of genetically normal, non-cancerous epithelial cells. We analysed >100 biopsy samples from early-stage breast cancer and present our large collection of >70 organoid lines. We also show methods of assessing successful tumour cell culture in a time, and cost-efficient manner, proving a high rate (>85%) of normal cell overgrowth in early-stage breast cancer organoids. Finally, we show a number of successful attempts to culture cancer organoids from mastectomy-derived tissue of advanced, metastatic breast cancer. We conclude that the usefulness of organoids from early breast cancer for translational research and personalised medicine, especially guidance of adjuvant or post-surgical maintenance therapy, is strongly limited by the low success rate of culturing cancerous cells under organoid conditions.
Collapse
Affiliation(s)
- Paul Schwerd‐Kleine
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Roberto Würth
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| | - Tasneem Cheytan
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Laura Michel
- Division of Gynecologic OncologyNational Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Verena Thewes
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Ewgenija Gutjahr
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Institute of Pathology, Heidelberg University HospitalHeidelbergGermany
| | - Huriye Seker‐Cin
- Institute of Pathology, Heidelberg University HospitalHeidelbergGermany
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University HospitalHeidelbergGermany
| | - Sarah‐Jane Neuberth
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Vera Thiel
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Jonas Schwickert
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Tim Vorberg
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Jennifer Wischhusen
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| | | | - Marc Zapatka
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Peter Lichter
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Andreas Schneeweiss
- Division of Gynecologic OncologyNational Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Andreas Trumpp
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| | - Martin R. Sprick
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| |
Collapse
|
5
|
Shao F, Huang X, Ma Z, Li L, Qi C. Differences in chemotherapeutic drug sensitivity before and after patient-derived tumor organoid construction. Toxicol Appl Pharmacol 2025; 499:117340. [PMID: 40228674 DOI: 10.1016/j.taap.2025.117340] [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: 11/04/2024] [Revised: 03/19/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Patient-derived organoids (PDOs) have emerged as promising preclinical models for various tumor types. This study aimed to optimize the process of constructing 3D organoid models and facilitate the development of personalized therapies for gastric and colon cancers. METHODS Tumor tissues were divided into two parts: one part was dissociated into a single-cell suspension, and the other part was used to culture tumor organoids. RNA sequencing (RNA-seq) was performed on both tumor cells and cultured organoids. Four chemotherapeutic agents-Oxaliplatin (L-OHP), Gemcitabine (GEM), 5-Fluorouracil (5-FU), and Paclitaxel (PTX)-were utilized to assess cytotoxicity and proliferation in both organoids and freshly isolated tumor cells, then the effects of these agents were evaluated. RESULTS Organoids were successfully established from both surgically resected and biopsy-derived tumor tissues. Phenotypic analysis indicated that the organoids retained the histological features and expression profiles of the original tumors. Notably, the morphological characteristics of the organoids remained stable across passages, demonstrating robust growth over time. Differentially expressed genes were identified in both gastric and colon cancer PDOs. GO and KEGG pathway analyses revealed similar gene enrichment in gastric and colon PDOs. Both gastric and colon cancer PDOs exhibited increased significant sensitivity to PTX and 5-FU compared to freshly isolated cancer cells. Furthermore, the expression of most stemness-related genes was reduced after organoid culture. CONCLUSIONS We successfully established organoid models that demonstrated robust growth and heightened drug sensitivity compared to freshly isolated tumor cells. These findings suggest that caution should be exercised when interpreting drug sensitivity results from organoid-based assays.
Collapse
Affiliation(s)
- Fang Shao
- Laboratory of Oncology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China; Largescale Equipment Platform, Changzhou Medical Center, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Xin Huang
- Laboratory of Oncology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Zhihong Ma
- TCM Key Laboratory Cultivation Base of Zhejiang Province for the Development and Clinical Transformation of Immunomodulatory Drugs, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Liqin Li
- TCM Key Laboratory Cultivation Base of Zhejiang Province for the Development and Clinical Transformation of Immunomodulatory Drugs, Huzhou Central Hospital, Huzhou, Zhejiang, China.
| | - Chunjian Qi
- Laboratory of Oncology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China.
| |
Collapse
|
6
|
Kumar D, Gupta S, Gupta V, Tanwar R, Chandel A. Engineering the Future of Regenerative Medicines in Gut Health with Stem Cell-Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10893-w. [PMID: 40380985 DOI: 10.1007/s12015-025-10893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
The advent of intestinal organoids, three-dimensional structures derived from stem cells, has significantly advanced the field of biology by providing robust in vitro models that closely mimic the architecture and functionality of the human intestine. These organoids, generated from induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), or adult stem cells, possess remarkable capabilities for self-renewal, differentiation into diverse intestinal cell types, and functional recapitulation of physiological processes, including nutrient absorption, epithelial barrier integrity, and host-microbe interactions. The utility of intestinal organoids has been extensively demonstrated in disease modeling, drug screening, and personalized medicine. Notable examples include iPSC-derived organoids, which have been effectively employed to model enteric infections, and ESC-derived organoids, which have provided critical insights into fetal intestinal development. Patient-derived organoids have emerged as powerful tools for investigating personalized therapeutics and regenerative interventions for conditions such as inflammatory bowel disease (IBD), cystic fibrosis, and colorectal cancer. Preclinical studies involving transplantation of human intestinal organoids into murine models have shown promising outcomes, including functional integration, epithelial restoration, and immune system interactions. Despite these advancements, several challenges persist, particularly in achieving reproducibility, scalability, and maturation of organoids, which hinder their widespread clinical translation. Addressing these limitations requires the establishment of standardized protocols for organoid generation, culture, storage, and analysis to ensure reproducibility and comparability of findings across studies. Nevertheless, intestinal organoids hold immense promise for transforming our understanding of gastrointestinal pathophysiology, enhancing drug development pipelines, and advancing personalized medicine. By bridging the gap between preclinical research and clinical applications, these organoids represent a paradigm shift in the exploration of novel therapeutic strategies and the investigation of gut-associated diseases.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India.
| | - Sonia Gupta
- Swami Devi Dyal Group of Professional Institute, Panchkula, India
| | - Vrinda Gupta
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Rajni Tanwar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Anchal Chandel
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| |
Collapse
|
7
|
Liu L, Wang H, Chen R, Song Y, Wei W, Baek D, Gillin M, Kurabayashi K, Chen W. Cancer-on-a-chip for precision cancer medicine. LAB ON A CHIP 2025. [PMID: 40376718 DOI: 10.1039/d4lc01043d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Many cancer therapies fail in clinical trials despite showing potent efficacy in preclinical studies. One of the key reasons is the adopted preclinical models cannot recapitulate the complex tumor microenvironment (TME) and reflect the heterogeneity and patient specificity in human cancer. Cancer-on-a-chip (CoC) microphysiological systems can closely mimic the complex anatomical features and microenvironment interactions in an actual tumor, enabling more accurate disease modeling and therapy testing. This review article concisely summarizes and highlights the state-of-the-art progresses in CoC development for modeling critical TME compartments including the tumor vasculature, stromal and immune niche, as well as its applications in therapying screening. Current dilemma in cancer therapy development demonstrates that future preclinical models should reflect patient specific pathophysiology and heterogeneity with high accuracy and enable high-throughput screening for anticancer drug discovery and development. Therefore, CoC should be evolved as well. We explore future directions and discuss the pathway to develop the next generation of CoC models for precision cancer medicine, such as patient-derived chip, organoids-on-a-chip, and multi-organs-on-a-chip with high fidelity. We also discuss how the integration of sensors and microenvironmental control modules can provide a more comprehensive investigation of disease mechanisms and therapies. Next, we outline the roadmap of future standardization and translation of CoC technology toward real-world applications in pharmaceutical development and clinical settings for precision cancer medicine and the practical challenges and ethical concerns. Finally, we overview how applying advanced artificial intelligence tools and computational models could exploit CoC-derived data and augment the analytical ability of CoC.
Collapse
Affiliation(s)
- Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - Huishu Wang
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - Ruiqi Chen
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Yujing Song
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - William Wei
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - David Baek
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Mahan Gillin
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Katsuo Kurabayashi
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
8
|
Wang Q, Yuan F, Zuo X, Li M. Breakthroughs and challenges of organoid models for assessing cancer immunotherapy: a cutting-edge tool for advancing personalised treatments. Cell Death Discov 2025; 11:222. [PMID: 40335487 PMCID: PMC12059183 DOI: 10.1038/s41420-025-02505-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Organoid models are powerful tools for evaluating cancer immunotherapy that provide a more accurate representation of the tumour microenvironment (TME) and immune responses than traditional models. This review focuses on the latest advancements in organoid technologies, including immune cell co-culture, 3D bioprinting, and microfluidic systems, which enhance the modelling of TME and facilitate the assessment of immune therapies such as immune checkpoint inhibitors (ICIs), CAR-T therapies, and oncolytic viruses. Although these models have great potential in personalised cancer treatment, challenges persist in immune cell diversity, long-term culture stability, and reproducibility. Future developments integrating artificial intelligence (AI), multi-omics, and high-throughput platforms are expected to improve the predictive power of organoid models and accelerate the clinical translation of immunotherapy.
Collapse
Affiliation(s)
- Qian Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210009, Jiangsu, PR China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, Jiangsu, PR China
| | - Fangwei Yuan
- Department of Thoracic Surgery, Lian Shui County People's Hospital, Huaian, 223400, Jiangsu, PR China
| | - Xianglin Zuo
- Biobank of Jiangsu Cancer Hospital (Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University), Nanjing, 210000, Jiangsu, PR China.
| | - Ming Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210009, Jiangsu, PR China.
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, Jiangsu, PR China.
| |
Collapse
|
9
|
Liu Y, Ma Y, Zhou B, Bian B, Zhou Y, Chen S, Zhang P, Shen L, Chen H. Clofoctol impairs the stemness of gastric cancer and induces TNF-mediated necroptosis by directly binding to RanBP2. Cell Mol Life Sci 2025; 82:194. [PMID: 40325218 PMCID: PMC12052660 DOI: 10.1007/s00018-025-05723-8] [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: 12/04/2024] [Revised: 04/10/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
Gastric cancer stem cells (GCSCs) play a crucial role in the initiation, progression, recurrence and therapeutic resistance, contributing to a poor prognosis. Consequently, GCSCs are deemed to be a potential therapeutic target for gastric cancer (GC). Although β-catenin is a well-recognized therapeutic target for GC and several inhibitors have demonstrated potent anti-tumor effects, there is a dearth of therapeutic agents targeting β-catenin for clinical therapy. In this study, we carried out high-throughput screening of clinically approved drugs to identify effective inhibitors of β-catenin. The results revealed that the antibiotic drug, clofoctol (CFT) effectively reduced the β-catenin level, attenuated stemness traits both in vitro and in vivo, and induced necroptosis of GCSCs. Further analyzing of downstream genes and targeted proteins, we found that CFT inhibited GCSCs viability by binding to the SUMO E3 ligase RanBP2, thereby suppressing the SerpinE1/β-catenin axis and activating TNF-mediated necroptosis. These results indicate that CFT may exert potent therapeutic effects against GC by targeting β-catenin and inhibiting the viability of GCSCs.
Collapse
Affiliation(s)
- Yi Liu
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhui Ma
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingqian Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Bingxian Bian
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shiyu Chen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Peng Zhang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hui Chen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Abate-Shen C, Politi K. The Evolution of Mouse Models of Cancer: Past, Present, and Future. Cold Spring Harb Perspect Med 2025; 15:a041736. [PMID: 38772706 PMCID: PMC12047742 DOI: 10.1101/cshperspect.a041736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
In the nearly 50 years since the original models of cancer first hit the stage, mouse models have become a major contributor to virtually all aspects of cancer research, and these have evolved well beyond simple transgenic or xenograft models to encompass a wide range of more complex models. As the sophistication of mouse models has increased, an explosion of new technologies has expanded the potential to both further develop and apply these models to address major challenges in cancer research. In the current era, cancer modeling has expanded to include nongermline genetically engineered mouse models (GEMMs), patient-derived models, organoids, and adaptations of the models better suited for cancer immunology research. New technologies that have transformed the field include the application of CRISPR-Cas9-mediated genome editing, in vivo imaging, and single-cell analysis to cancer modeling. Here, we provide a historical perspective on the evolution of mouse models of cancer, focusing on how far we have come in a relatively short time and how new technologies will shape the future development of mouse models of cancer.
Collapse
Affiliation(s)
- Cory Abate-Shen
- Departments of Molecular Pharmacology and Therapeutics, Urology, Pathology and Cell Biology, Medicine, and Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Katerina Politi
- Departments of Pathology and Internal Medicine (Section of Medical Oncology) and Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06405, USA
| |
Collapse
|
11
|
Zhou JM, Dai WX, Wang RJ, Xu WQ, Xiang Z, Wang YX, Zhang T, Zhao YM, Wang L, Mao AR. Organoid modeling identifies USP3-AS1 as a novel promoter in colorectal cancer liver metastasis through increasing glucose-driven histone lactylation. Acta Pharmacol Sin 2025; 46:1404-1418. [PMID: 39837984 PMCID: PMC12032002 DOI: 10.1038/s41401-024-01465-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025]
Abstract
Dysregulation of long non-coding RNAs (lncRNAs) is common in colorectal cancer liver metastasis (CRLM). Emerging evidence links lncRNAs to multiple stages of metastasis from initial migration to colonization of distant organs. In this study we investigated the role of lncRNAs in metabolic reprogramming during CRLM using patient-derived organoid (PDO) models. We established five pairs of PDOs from primary tumors and matched liver metastatic lesions, followed by microarray analysis. We found that USP3-AS1 was significantly upregulated in CRLM-derived PDOs compared to primary tumors. High level of USP3-AS1 was positively associated with postoperative liver metastasis and negatively correlated with the prognosis of colorectal cancer (CRC) patients. Overexpression of USP3-AS1 significantly enhanced both sphere formation efficiency and liver metastasis in PDOs. Gene set enrichment analysis revealed that USP3-AS1 upregulation significantly enriched glycolysis and MYC signaling pathways. Metabolomics analysis confirmed that USP3-AS1 promoted glycolysis in PDOs, whereas glycolysis inhibition partially attenuated the effects of USP3-AS1 overexpression on PDO growth and liver metastasis. We revealed that USP3-AS1 stabilized MYC via post-translational deubiquitination, thereby promoting glycolysis. We demonstrated that USP3-AS1 increased the stability of USP3 mRNA, resulting in higher USP3 protein expression. The elevated USP3 protein then interacted with MYC and promoted its stability by deubiquitination. The USP3-AS1-MYC-glycolysis regulatory axis modulated liver metastasis by promoting H3K18 lactylation and CDC27 expression in CRC. In conclusion, USP3-AS1 is a novel promoter of CRLM by inducing histone lactylation.
Collapse
Affiliation(s)
- Jia-Min Zhou
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wei-Xing Dai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Colorectal Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Ren-Jie Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Colorectal Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Wei-Qi Xu
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhen Xiang
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Xiu Wang
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ti Zhang
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Ming Zhao
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lu Wang
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - An-Rong Mao
- Department of Hepatic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Li S, Ren J, Wu J, Xia Z, Li Y, Li C, Cao W. Establishment and molecular characterisation of patient-derived organoids for primary central nervous system lymphoma. Leukemia 2025; 39:1169-1183. [PMID: 40102628 DOI: 10.1038/s41375-025-02562-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/20/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
Primary central nervous system lymphoma (PCNSL) exhibits substantial intratumoural and intertumoural heterogeneity, complicating the development of effective treatment methods. Existing in vitro models fail to simulate the cellular and mutational diversity of native tumours and require prolonged generation times. Therefore, we developed a culture method for patient-derived PCNSL organoids (CLOs) and evaluated the organoids through extensive molecular characterisation, histopathological analysis, single-nucleus RNA sequencing, bulk RNA sequencing and whole-exome sequencing. These CLOs accurately mimicked the histological attributes, gene expression landscapes and mutational profiles of their original tumours. Single-nucleus RNA sequencing also revealed that CLOs maintained cell-type heterogeneity and the molecular signatures of their original tumours. CLOs were generated within 2 weeks, demonstrating rapid development and reliability. Therapeutic profiling was performed on three selected CLOs treated with four standard drugs. The CLOs exhibited specific sensitivity to methotrexate, and resistance to dexamethasone, ibrutinib and rituximab, suggesting that CLOs may be valuable tools for reflecting drug sensitivities. Taken together, these results emphasise that CLOs effectively emulate the key characteristics of PCNSL, increasing the understanding of the genetic landscape of this complex disease. CLOs provide a rapid and reliable platform for exploring individualised treatment strategies, potentially accelerating the transition of research findings to clinical practice.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| | - Jun Ren
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianing Wu
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zuguang Xia
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yingzhu Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chengxun Li
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.
| | - Wenjun Cao
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Guo Z, Liu J, Zhang X, Ma Y, Wang Y, Li P, Huang R, Li Z. Precision treatment guided by patient-derived organoids-based drug testing for locally advanced thyroid cancer: a single arm, phase 2 study. Endocrine 2025:10.1007/s12020-025-04240-9. [PMID: 40304938 DOI: 10.1007/s12020-025-04240-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE Locally advanced thyroid cancer (LATC) presents significant surgical challenges, with a high risk of incomplete resection and poor prognosis. Patient-derived organoids (PDOs) are a powerful tool to assess drug sensitivity at an individual level and to suggest new treatment options or re-challenge. This study aimed to evaluate the method's feasibility and efficacy as applied to patients with LATC. METHODS In this single arm, phase 2 study, we enrolled 75 patients with LATC. Biopsies from the primary tumor or metastatic site were cultured using organoid models. Sensitivity testing was performed by using PDOs with a panel of drugs with proven activity in phase II or III trials. At the discretion of the investigator considering toxicity, the drug with the highest relative activity was offered. The primary endpoint was the objective response rate (ORR). RESULTS Fifty-five patients received at least one dose of recommended drug and the primary endpoint, objective response was met in 18 patients with an overall ORR as 32.7% (95% CI 20.7-46.7). Based on the pre-defined subgroups of different histological subtypes, the ORR for patients with differentiated thyroid cancer, medullary thyroid cancer, anaplastic thyroid cancer were 32.6%, (95% CI 19.1-48.5), 16.7% (95% CI 0.4-64.1) and 50% (95% CI 11.8-88.2), respectively. The R0/R1 resection rate was 34.5% (19/55). CONCLUSIONS This study is the first to validate the feasibility of PDOs and in vitro sensitivity testing for LATC. PDO-based neoadjuvant therapy holds promise in improving prognosis and providing surgical opportunities for these patients. TRIAL REGISTRATION The study was registered at ClinicalTrials.gov (NCT06482086) on 06/25/2024.
Collapse
Affiliation(s)
- Ziliang Guo
- Division of Thyroid Surgery, Department of General Surgery; Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610000, China
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Jiaye Liu
- Division of Thyroid Surgery, Department of General Surgery; Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610000, China
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Xinyue Zhang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610000, China
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yu Ma
- Division of Thyroid Surgery, Department of General Surgery; Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yichao Wang
- Division of Thyroid Surgery, Department of General Surgery; Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Peng Li
- Department of thyroid surgery, Peking University Shenzhen hospital, Shenzhen, Guangdong, 518036, China
| | - Rui Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610000, China.
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery; Laboratory of Thyroid and Parathyroid Diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
14
|
Liu P, Zhou S, Zhou Z, Jin Z, Chen W, Li Z, Xu J, Chen F, Li Y, Wen Y, Zhang S, Zhang C, Li B, Zhao J, Chen H. Discovery and antitumor evaluation of a mitochondria-targeting ruthenium complex for effective cancer therapy. Cancer Lett 2025; 616:217582. [PMID: 40021041 DOI: 10.1016/j.canlet.2025.217582] [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: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Ruthenium-based metallodrugs have garnered attention as a promising alternative for anticancer therapy, aiming to overcome chemoresistance and severe side effects linked to platinum-based drugs. However, ruthenium complexes tested in clinical trials to date have yielded unsatisfactory results. This study synthesized a positively charged ruthenium complex (Ru-2) that effectively penetrated cancer cells and exhibited superior cytotoxicity to cisplatin in vitro against cancer cell lines and organoids. Ru-2 selectively targeted mitochondria, disrupting their function by depolarizing mitochondrial membrane potential, elevating reactive oxygen species production, and impairing both oxidative phosphorylation and the tricarboxylic acid cycle. Furthermore, Ru-2 triggered endoplasmic reticulum (ER) stress and apoptosis. Integrative transcriptomic and proteomic analyses, performed using RNA sequencing and mass spectrometry, identified key molecular changes in cancer cells treated with Ru-2. For enhanced in vivo application, we developed a transferrin-based nanomedicine formulation, TF/Ru-2, incorporating Ru-2 into transferrin. In vivo studies demonstrated that both Ru-2 and TF/Ru-2 exhibited superior antitumor efficacy and improved biosafety compared to cisplatin. This study presents a novel ruthenium complex and a transferrin-based drug delivery platform with significant potential for future cancer therapies.
Collapse
Affiliation(s)
- Peng Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Shangbo Zhou
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Zihan Jin
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Wei Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Zihang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Jiaqi Xu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Feng Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - You Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Yingfei Wen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Shiqiang Zhang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China; Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Binbin Li
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Hengxing Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China; Clinical Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
15
|
Rodríguez-Martínez A, Giraldo-Ruiz L, Ramos MC, Luque I, Ribeiro D, Postigo-Corrales F, Alburquerque-González B, Montoro-García S, Arroyo-Rodríguez AB, Conesa-Zamora P, Hurtado AM, Luengo-Gil G, Pérez-Sánchez H. Discovery of Z1362873773: a novel fascin inhibitor from a large chemical library for colorectal cancer. Sci Rep 2025; 15:14906. [PMID: 40295602 PMCID: PMC12037858 DOI: 10.1038/s41598-025-96457-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Metastasis is one of the leading causes of cancer-related death worldwide. Fascin, a protein that bundles actin filaments to produce protrusions in cancer cells, plays a significant role in the enhancement of cell migration. This protein has been shown that the overexpression of this protein is related to the appearance of different types of cancer, such as colorectal cancer. In this study, we conducted in silico screening of the Enamine library, a compound library with a broad chemical space. Using a ligand-based virtual screening approach based on the pharmacophore model of G2, we identified the predicted inhibitors. First, these compounds were validated by physicochemical analysis. Differential scanning calorimetry (DSF) was used to study the binding between the predicted compounds and fascin protein, followed by an F-actin bundling assay to determine which compounds inhibited the bundling function of fascin. Z1362873773, which exhibited binding to fascin and inhibited F-actin bundling, was further tested in cell cultures to assess its effects on cancer cell viability and migration as well as in organoid models to evaluate potential cytotoxicity. Finally, we established a protocol that can be applied to discover anti-fascin agents from diverse compound libraries. A new molecule has been identified with considerable fascin inhibitory and migration-arresting capacity, which may lead to the development of new therapies to treat cancer.
Collapse
Affiliation(s)
- Alejandro Rodríguez-Martínez
- Structural Bioinformatics and High Performance Computing Research Group (BIO-HPC), UCAM Universidad Católica de Murcia (UCAM), HiTech Innovation Hub, Murcia, 30107, Spain
- Health Sciences PhD Program, Universidad Católica de Murcia UCAM, Campus de los Jerónimos nº135, Guadalupe, Murcia, 30107, Spain
| | - Lucía Giraldo-Ruiz
- Departamento de Química Física, Instituto de Biotecnología y Unidad de Excelencia de Química aplicada a Biomedicina y Medioambiente, Universidad de Granada, Campus Fuentenueva s/n, Granada, 18071, Spain
| | - María C Ramos
- Fundación MEDINA, PTS Health Sciences Technology Park, Avenida del Conocimiento 34, Granada, 18016, Spain
| | - Irene Luque
- Departamento de Química Física, Instituto de Biotecnología y Unidad de Excelencia de Química aplicada a Biomedicina y Medioambiente, Universidad de Granada, Campus Fuentenueva s/n, Granada, 18071, Spain
| | - Diogo Ribeiro
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Fátima Postigo-Corrales
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Begoña Alburquerque-González
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Silvia Montoro-García
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Ana Belén Arroyo-Rodríguez
- Molecular Pathology and Pharmacogenetics Research Group, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital General Universitario Santa Lucía, Cartagena, 30202, Spain
| | - Pablo Conesa-Zamora
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
- Molecular Pathology and Pharmacogenetics Research Group, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital General Universitario Santa Lucía, Cartagena, 30202, Spain
| | - Ana María Hurtado
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Ginés Luengo-Gil
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain.
- Molecular Pathology and Pharmacogenetics Research Group, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital General Universitario Santa Lucía, Cartagena, 30202, Spain.
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High Performance Computing Research Group (BIO-HPC), UCAM Universidad Católica de Murcia (UCAM), HiTech Innovation Hub, Murcia, 30107, Spain.
| |
Collapse
|
16
|
Kan L, Yu Y, Wang Y, Shi L, Fan T, Chen H, Ren C. The application of organoids in investigating immune evasion in the microenvironment of gastric cancer and screening novel drug candidates. Mol Cancer 2025; 24:125. [PMID: 40287758 PMCID: PMC12032790 DOI: 10.1186/s12943-025-02328-4] [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: 12/09/2024] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric cancer (GC) is a prevalent digestive system tumor, the fifth most diagnosed cancer worldwide, and a leading cause of cancer deaths. GC is distinguished by its pronounced heterogeneity and a dynamically evolving tumor microenvironment (TME). The lack of accurate disease models complicates the understanding of its mechanisms and impedes the discovery of novel drugs. A growing body of evidence suggests that GC organoids, developed using organoid culture technology, preserve the genetic, phenotypic, and behavioral characteristics. GC organoids hold significant potential for predicting treatment responses in individual patients. This review provides a comprehensive overview of the current clinical treatment strategies for GC, as well as the history, construction and clinical applications of organoids. The focus is on the role of organoids in simulating the TME to explore mechanisms of immune evasion and intratumoral microbiota in GC, as well as their applications in guiding clinical drug therapy and facilitating novel drug screening. Furthermore, we summarize the limitations of GC organoid models and underscore the need for continued technological advancements to benefit both basic and translational oncological research.
Collapse
Affiliation(s)
- Liuyue Kan
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Ying Yu
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yaxue Wang
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Lei Shi
- Department of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China
| | - Tingyuan Fan
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Hui Chen
- Department of Geriatrics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China.
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| | - Chuanli Ren
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Department of Laboratory Medicine, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China.
- The Yangzhou Clinical Medical College of Xuzhou Medical University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| |
Collapse
|
17
|
Park YG, Kim S, Min S, Kim E, Kim D, Cho YH, Kim S, Joo H, Jeong I, Lim JA, Lee S, Cho SW, Park JU. Soft 3D Bioelectrodes for Intraorganoid Signal Monitoring in Cardiac Models. NANO LETTERS 2025; 25:6481-6490. [PMID: 40200576 DOI: 10.1021/acs.nanolett.5c00069] [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: 04/10/2025]
Abstract
Continuous monitoring of physiological activities within the internal regions of three-dimensional (3D) organoids holds significant promise for advancing organoid-based research. However, conventional methods are constrained to capturing signals from the peripheral surfaces of organoids, limiting insights into internal dynamics. Here, we present a soft 3D bioelectrode platform for continuous intraorganoid signal monitoring. These bioelectrodes, formed via 3D printing of liquid metal, are designed with customizable geometric parameters, including height and diameter, to adapt to various organoid structures. The tissue-comparable softness of the electrodes minimizes damage to cardiac organoids, ensuring a stable interface for reliable signal recording even under dynamic deformations caused by rhythmic contractions or displacements in aqueous environments. The array configuration enables simultaneous electrocardiogram (ECG) recordings from 32 organoids. Demonstrating real-time monitoring of drug-induced ECG responses, this scalable platform highlights its potential for high-throughput drug screening.
Collapse
Affiliation(s)
- Young-Geun Park
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sumin Kim
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Department of MetaBioHealth, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Enji Kim
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Dayeon Kim
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Seoul 03722, Republic of Korea
- Soft Hybrid Materials Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Nanoscience and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Yo Han Cho
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Suran Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Inhea Jeong
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jung Ah Lim
- Soft Hybrid Materials Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Nanoscience and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Sangmin Lee
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jang-Ung Park
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
18
|
Zhao Y, Zhang L, Xia L, E H, Wang T, Lu H, Chen H, She Y, Tang H, Wu J, Zhao D, Chen C. A METTL3-NFE2L3 axis mediates tumor stemness and progression in lung adenocarcinoma. SCIENCE ADVANCES 2025; 11:eadt7682. [PMID: 40249818 PMCID: PMC12007586 DOI: 10.1126/sciadv.adt7682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/14/2025] [Indexed: 04/20/2025]
Abstract
The progression of lung adenocarcinoma is primarily driven by cancer stem cells (CSCs), which have self-renewal capabilities and confer resistance to therapies, including neoadjuvant treatments combining chemotherapy and immune checkpoint inhibitors. In this study, we identified that OV6+ tumor cells exhibit stem-like characteristics and are notably enriched in patients with non-major pathological response, closely associated with resistance to combination therapies. Hypoxia and HIF1α were found to drive the formation of OV6+ CSCs. METTL3, a methyltransferase, was revealed as a critical regulator of OV6+ CSCs by stabilizing NFE2L3 messenger RNA via an N6-methyladenosine-dependent manner, thereby up-regulating NFE2L3 and activating the intrinsic WNT signaling pathway essential for maintaining stemness. OV6+ tumor cells promoted M2 macrophage infiltration and the formation of an immunosuppressive tumor microenvironment (TME). Targeting METTL3 effectively eliminated OV6+ CSCs and suppressed tumor progression. Moreover, the combination of STM2457 with cisplatin overcame chemoresistance, remodeled the TME, and provided promising insights for enhancing the efficacy of neoadjuvant combination therapies.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lang Xia
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haoran E
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huinan Lu
- Peking University Yangtze Center of Future Health Technology, Wuxi, Jiangsu, China
| | - Hezhong Chen
- Department of Thoracic Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Tang
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
19
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
20
|
Calibasi-Kocal G, Sever T, Canda AE, Kadioglu LE, Ates H, Basbinar Y, Ellidokuz E. Impact of enzymatic isolation on the propagation efficiency of patient-derived colorectal cancer organoids. Sci Rep 2025; 15:13452. [PMID: 40251201 PMCID: PMC12008271 DOI: 10.1038/s41598-025-97650-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
Cancer organoids are three-dimensional in vitro models that closely replicate the genetic, phenotypic, and heterogeneity characteristics of original tumors, making them valuable tools in cancer research. However, the lack of standardized protocols limits their broader application. This study evaluates the role of enzymatic isolation in generating patient-derived organoids (PDOs) from colorectal cancer tissues by comparing four enzymatic methods: TrypLE, Trypsin-EDTA (T/E), Collagenase, and Hyaluronidase. Colorectal cancer tissues were processed using these enzymes, and cell viability, dissociation efficiency, and isolation quality were assessed via Trypan Blue exclusion assay and 7-AAD staining with flow cytometry. Cancer stem cells marked by LGR5 and CD133 were quantified via flow cytometry, while organoid generation and growth were monitored over 11 days using confocal microscopy. TrypLE and T/E demonstrated superior preservation of cell viability but limited dissociation efficiency, yielding lower cell count per milligram of tissue. In contrast, Collagenase and Hyaluronidase demonstrated superior tissue dissociation, yielding higher total cell counts and the highest proportions of LGR5positive and CD133positive stem cell populations. Collagenase produced the highest organoid counts, while Hyaluronidase supported the largest organoid expansion, with both enzymes generating larger organoid surface areas and a greater number of organoids compared to TrypLE and T/E. These results highlight Collagenase and Hyaluronidase as optimal choices for PDO generation, providing a framework for optimizing dissociation protocols. This study underscores the critical influence of enzymatic dissociation methods on the establishment and reliability of colorectal cancer patient-derived organoids, providing a foundation for optimizing PDO protocols and advancing their translational application in precision oncology.
Collapse
Affiliation(s)
- Gizem Calibasi-Kocal
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Türkiye.
| | - Tolga Sever
- Department of Oncology, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| | - Aras Emre Canda
- Private Clinic for Colorectal Cancer Surgery and Peritoneal Carcinomatosis, Izmir, Turkey
- Acibadem Izmir Kent Hospital Affiliated Surgeon, Izmir, Turkey
| | | | - Halil Ates
- Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Yasemin Basbinar
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Türkiye
| | - Ender Ellidokuz
- Department of Internal Diseases, Gastroenterology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
21
|
Kuhn MR, Wolcott EA, Langer EM. Developments in gastrointestinal organoid cultures to recapitulate tissue environments. Front Bioeng Biotechnol 2025; 13:1521044. [PMID: 40313639 PMCID: PMC12043594 DOI: 10.3389/fbioe.2025.1521044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/21/2025] [Indexed: 05/03/2025] Open
Abstract
Culture platforms that closely mimic the spatial architecture, cellular diversity, and extracellular matrix composition of native tissues can serve as invaluable tools for a range of scientific discovery and biomedical applications. Organoids have emerged as a promising alternative to both traditional 2D cell culture and animal models, offering a physiologically relevant 3D culture system for studying human cell biology. Organoids provide a manipulable platform to investigate organ development and function as well as to model patient-specific phenotypes. This mini review examines various methods used for culturing organoids to model normal and disease conditions in gastrointestinal tissues. We focus on how the matrix composition and media formulations can impact cell signaling, altering the baseline cellular phenotypes as well as response to perturbations. We discuss future directions for optimizing organoid culture conditions to improve basic and translational potential.
Collapse
Affiliation(s)
- Madeline R. Kuhn
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Emma A. Wolcott
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Ellen M. Langer
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
22
|
Quinn CH, Julson JR, Erwin MH, Markert HR, Bownes LV, Stewart JE, Shirley S, Yoon KJ, Aye JM, Markert JM, Beierle EA. Investigation of an oncolytic herpes simplex virus as a potential therapeutic agent for gastroenteropancreatic neuroendocrine neoplasms. Sci Rep 2025; 15:13356. [PMID: 40247049 PMCID: PMC12006505 DOI: 10.1038/s41598-025-98588-7] [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: 10/06/2024] [Accepted: 04/14/2025] [Indexed: 04/19/2025] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) present unique challenges due to their heterogeneity and limited treatment options. Oncolytic virotherapy has emerged as a promising therapeutic for other NETs and thus, we sought to investigate the potential of an engineered oncolytic herpes simplex virus (oHSV), M002, for GEP-NETS. We employed an established long-term passage GEP-NET cell line and a unique, human pediatric patient-derived xenograft GEP-NET line. We found the virus to effectively infect, replicate within, and kill both cell lines in vitro. Similar effects were noted in vivo, with M002 decreasing tumor growth and improving overall survival in mice bearing tumors from both the established cell line and human GEP-NET PDX. Overall, these studies provide an evaluation of an oncolytic HSV in GEP-NETs, highlighting its therapeutic potential and considerations for clinical translation.
Collapse
Affiliation(s)
- Colin H Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA
| | - Janet R Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA
| | - Michael H Erwin
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA
| | - Hooper R Markert
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA
| | - Larua V Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA
| | - Jerry E Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA
| | - Sorina Shirley
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA
| | - Karina J Yoon
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jamie M Aye
- Division of Hematology Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, 1600 7th Ave South, Lowder, Suite 300, Birmingham, AL, 35233, USA.
| |
Collapse
|
23
|
Tao ZH, Han JX, Xu J, Zhao E, Wang M, Wang Z, Lin XL, Xiao XY, Hong J, Chen H, Chen YX, Chen HM, Fang JY. Screening of patient-derived organoids identifies mitophagy as a cell-intrinsic vulnerability in colorectal cancer during statin treatment. Cell Rep Med 2025; 6:102039. [PMID: 40154491 PMCID: PMC12047522 DOI: 10.1016/j.xcrm.2025.102039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/26/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Statins, commonly used to lower cholesterol, are associated with improved prognosis in colorectal cancer (CRC), though their effectiveness varies. This study investigates the anti-cancer effects of atorvastatin in CRC using patient-derived organoids (PDOs) and PDO-derived xenograft (PDOX) models. Our findings reveal that atorvastatin induces mitochondrial dysfunction, leading to apoptosis in cancer cells. In response, cancer cells induce mitophagy to clear damaged mitochondria, enhancing survival and reducing statin efficacy. Analysis of a clinical cohort confirms mitophagy's role in diminishing statin effectiveness. Importantly, inhibiting mitophagy significantly enhances the anti-cancer effects of atorvastatin in CRC PDOs, xenograft models, and azoxymethane (AOM)-dextran sulfate sodium (DSS) mouse models. These findings identify mitophagy as a critical pro-survival mechanism in CRC during statin treatment, providing insights into the variable responses observed in epidemiological studies. Targeting this vulnerability through combination therapy can elicit potent therapeutic responses.
Collapse
Affiliation(s)
- Zhi-Hang Tao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ji-Xuan Han
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Enhao Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Lin Lin
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiu-Ying Xiao
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Hong
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Min Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
24
|
Shin AE, Sugiura K, Kariuki SW, Cohen DA, Flashner SP, Klein-Szanto AJ, Nishiwaki N, De D, Vasan N, Gabre JT, Lengner CJ, Sims PA, Rustgi AK. LIN28B-mediated PI3K/AKT pathway activation promotes metastasis in colorectal cancer models. J Clin Invest 2025; 135:e186035. [PMID: 39808497 PMCID: PMC11996871 DOI: 10.1172/jci186035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer death because of metastatic spread. LIN28B is overexpressed in 30% of CRCs and promotes metastasis, yet its mechanisms remain unclear. In this study, we genetically modified CRC cell lines to overexpress LIN28B, resulting in enhanced PI3K/AKT pathway activation and liver metastasis in mice. We developed genetically modified mouse models with constitutively active Pik3ca that form intestinal tumors progressing to liver metastases with an intact immune system, addressing the limitations of previous Pik3ca-mutant models, including long tumor latency, mixed histology, and lack of distant metastases. The PI3Kα-specific inhibitor alpelisib reduced migration and invasion in vitro and metastasis in vivo. We present a comprehensive analysis of vertical inhibition of the PI3K/AKT pathway in CRC using the FDA-approved drugs alpelisib and capivasertib (an AKT inhibitor) in combination with LY2584702 (a ribosomal protein S6 kinase inhibitor) in CRC cell lines and mouse- and patient-derived organoids. Tissue microarrays from patients with CRC verified that LIN28B and PI3K/AKT pathway activation correlate with CRC progression. These findings highlight the critical role of the LIN28B-mediated PI3K/AKT pathway in CRC metastasis, the therapeutic potential of targeted inhibition, and the promise of patient-derived organoids in precision medicine in metastatic CRC.
Collapse
Affiliation(s)
- Alice E. Shin
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Kensuke Sugiura
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | | | - David A. Cohen
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons; Columbia University Irving Medical Center, New York, New York, USA
| | | | | | | | - Dechokyab De
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Neil Vasan
- Division of Hematology and Oncology, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Joel T. Gabre
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Christopher J. Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter A. Sims
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, and
| |
Collapse
|
25
|
Liu C, Shi C, Wang S, Qi R, Gu W, Yu F, Zhang G, Qiu F. Bridging the gap: how patient-derived lung cancer organoids are transforming personalized medicine. Front Cell Dev Biol 2025; 13:1554268. [PMID: 40302940 PMCID: PMC12037501 DOI: 10.3389/fcell.2025.1554268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Lung cancer is a major malignancy that poses a significant threat to human health, with its complex pathogenesis and molecular characteristics presenting substantial challenges for treatment. Traditional two-dimensional cell cultures and animal models are limited in their ability to accurately replicate the characteristics of different lung cancer patients, thereby hindering research on disease mechanisms and treatment strategies. The development of organoid technology has enabled the growth of patient-derived tumor cells in three-dimensional cultures, which can stably preserve the tumor's tissue morphology, genomic features, and drug response. There have been significant advancements in the field of patient-derived lung cancer organoids (PDLCOs), challenges remain in the reproducibility and standardization of PDLCOs models due to variations in specimen sources, subsequent processing techniques, culture medium formulations, and Matrigel batches. This review summarizes the cultivation and validation processes of PDLCOs and explores their clinical applications in personalized treatment, drug screening after resistance, PDLCOs biobanks construction, and drug development. Additionally, the integration of PDLCOs with cutting-edge technologies in various fields, such as tumor assembloid techniques, artificial intelligence, organoid-on-a-chip, 3D bioprinting, gene editing, and single-cell RNA sequencing, has greatly expanded their clinical potential. This review, incorporating the latest research developments in PDLCOs, provides an overview of their cultivation, clinical applications, and interdisciplinary integration, while also addressing the prospects and challenges of PDLCOs in precision medicine for lung cancer.
Collapse
Affiliation(s)
- Chaoxing Liu
- Department of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Nanchang Key Laboratory of Tumor Gene Diagnosis and Innovative Treatment Research, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chao Shi
- Department of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Nanchang Key Laboratory of Tumor Gene Diagnosis and Innovative Treatment Research, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Siya Wang
- Nanchang Key Laboratory of Tumor Gene Diagnosis and Innovative Treatment Research, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Rong Qi
- Department of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Nanchang Key Laboratory of Tumor Gene Diagnosis and Innovative Treatment Research, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Weiguo Gu
- Department of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Nanchang Key Laboratory of Tumor Gene Diagnosis and Innovative Treatment Research, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Feng Yu
- Department of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Guohua Zhang
- Nanchang Key Laboratory of Tumor Gene Diagnosis and Innovative Treatment Research, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Feng Qiu
- Department of Oncology, Gaoxin Branch of the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
26
|
Chia S, Wen Seow JJ, Peres da Silva R, Suphavilai C, Shirgaonkar N, Murata-Hori M, Zhang X, Yong EY, Pan J, Thangavelu MT, Periyasamy G, Yap A, Anand P, Muliaditan D, Chan YS, Siyu W, Yong CW, Hong N, Ran G, Sim NL, Guo YA, Yi Teh AX, Wei Ling CC, Wei Tan EK, Pei Cherylin FW, Chang M, Han S, Seow-En I, Chen Hui LR, Hsia Gan AH, Yap CK, Ng HH, Skanderup AJ, Chinswangwatanakul V, Riansuwan W, Trakarnsanga A, Pithukpakorn M, Tanjak P, Chaiboonchoe A, Park D, Kim DK, Iyer NG, Tsantoulis P, Tejpar S, Kim JE, Kim TI, Sampattavanich S, Tan IB, Nagarajan N, DasGupta R. CAN-Scan: A multi-omic phenotype-driven precision oncology platform identifies prognostic biomarkers of therapy response for colorectal cancer. Cell Rep Med 2025; 6:102053. [PMID: 40187357 PMCID: PMC12047494 DOI: 10.1016/j.xcrm.2025.102053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/10/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Application of machine learning (ML) on cancer-specific pharmacogenomic datasets shows immense promise for identifying predictive response biomarkers to enable personalized treatment. We introduce CAN-Scan, a precision oncology platform, which applies ML on next-generation pharmacogenomic datasets generated from a freeze-viable biobank of patient-derived primary cell lines (PDCs). These PDCs are screened against 84 Food and Drug Administration (FDA)-approved drugs at clinically relevant doses (Cmax), focusing on colorectal cancer (CRC) as a model system. CAN-Scan uncovers prognostic biomarkers and alternative treatment strategies, particularly for patients unresponsive to first-line chemotherapy. Specifically, it identifies gene expression signatures linked to resistance against 5-fluorouracil (5-FU)-based drugs and a focal copy-number gain on chromosome 7q, harboring critical resistance-associated genes. CAN-Scan-derived response signatures accurately predict clinical outcomes across four independent, ethnically diverse CRC cohorts. Notably, drug-specific ML models reveal regorafenib and vemurafenib as alternative treatments for BRAF-expressing, 5-FU-insensitive CRC. Altogether, this approach demonstrates significant potential in improving biomarker discovery and guiding personalized treatments.
Collapse
Affiliation(s)
- Shumei Chia
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
| | - Justine Jia Wen Seow
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Rafael Peres da Silva
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Chayaporn Suphavilai
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Niranjan Shirgaonkar
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Maki Murata-Hori
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Xiaoqian Zhang
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Elena Yaqing Yong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Jiajia Pan
- National Cancer Centre, Singapore, Singapore
| | - Matan Thangavelu Thangavelu
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Experimental Drug Development Centre (EDDC), A∗STAR, Singapore, Singapore
| | - Giridharan Periyasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Experimental Drug Development Centre (EDDC), A∗STAR, Singapore, Singapore
| | - Aixin Yap
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Padmaja Anand
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Daniel Muliaditan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Yun Shen Chan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Wang Siyu
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Chua Wei Yong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Nguyen Hong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Gao Ran
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Ngak Leng Sim
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Yu Amanda Guo
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | | | | | - Emile Kwong Wei Tan
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | - Fu Wan Pei Cherylin
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | - Meihuan Chang
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | - Shuting Han
- National Cancer Centre, Singapore, Singapore
| | - Isaac Seow-En
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | | | - Anna Hwee Hsia Gan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Choon Kong Yap
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Huck Hui Ng
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anders Jacobsen Skanderup
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Vitoon Chinswangwatanakul
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Woramin Riansuwan
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Atthaphorn Trakarnsanga
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Manop Pithukpakorn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol, Bangkok, Thailand
| | - Pariyada Tanjak
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Amphun Chaiboonchoe
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Daye Park
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Keon Kim
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | | | - Petros Tsantoulis
- Hôpitaux Universitaires de Genève, University of Geneva, Geneva, Switzerland
| | - Sabine Tejpar
- Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jung Eun Kim
- R&D center PODO Therapeutics Co. 338 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13493, Republic of Korea
| | - Tae Il Kim
- R&D center PODO Therapeutics Co. 338 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13493, Republic of Korea; Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Iain Beehuat Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; National Cancer Centre, Singapore, Singapore; Duke-National University of Singapore Medical School, Singapore, Singapore.
| | - Niranjan Nagarajan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Ramanuj DasGupta
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; CRUK Scotland Institute, School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
27
|
Kim SY, van de Wetering M, Clevers H, Sanders K. The future of tumor organoids in precision therapy. Trends Cancer 2025:S2405-8033(25)00073-1. [PMID: 40185656 DOI: 10.1016/j.trecan.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Tumoroids are cultures of patient-derived tumor cells, which are grown in 3D in the presence of an extracellular matrix extract and specific growth factors. Tumoroids can be generated from adult as well as pediatric cancers, including epithelial cancers, sarcomas, and brain cancers. Tumoroids retain multi-omic characteristics of their corresponding tumor and recapitulate interpatient and intratumor heterogeneity. Retrospective and prospective studies have demonstrated that tumoroids predict patient responses to anticancer therapies, making them a promising tool for precision oncology. However, several challenges remain before tumoroids can be fully integrated into clinical decision-making, including success rates of tumoroid establishment and turnaround times. This review discusses the current advances, challenges, and future directions of tumoroid-based models in cancer research and precision therapy.
Collapse
Affiliation(s)
- Seok-Young Kim
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, The Netherlands; Current address: Roche Pharmaceutical Research and Early Development (pRED) of F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Karin Sanders
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
28
|
Penrose HM, Sinha S, Tindle C, Zablan K, Le HN, Neill J, Ghosh P, Boland BS. A Living Organoid Biobank of Crohn's Disease Patients Reveals Distinct Clinical Correlates of Molecular Subtypes of Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.01.25325058. [PMID: 40236416 PMCID: PMC11998810 DOI: 10.1101/2025.04.01.25325058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Current clinical decision-making lacks reliable preclinical models to predict patient outcomes. Here, we establish patient-derived organoids (PDOs) as predictive tools in Crohn's disease (CD), a complex, heterogeneous disorder. Using a living biobank of adult stem cell-derived colonic-PDOs, we identified two molecular CD subtypes-Immune-Deficient Infectious CD ( IDICD ) and Stress and Senescence-Induced Fibrostenotic CD ( S2FCD )-each with distinct genomic, transcriptomic and functional profiles, along with paired therapeutics. By prospectively linking colonic PDO-derived phenotypes to real-world patient outcomes, we uncovered that while S2FCD associates with severe colonic disease, IDICD associates with severe ileal disease, prior ileocecal surgery, and future disease progression. This approach transforms PDOs from static descriptive models into dynamic tools that capture the past, present, and future of disease behavior and reveals their utility as patient-specific predictive platforms, extending their use beyond oncology to complex inflammatory diseases. Findings also suggest that colonic immune dysfunction may drive ileal-CD, independent of colonic involvement. GRAPHICAL ABSTRACT In Brief In this work, Penrose et al. demonstrate the potential of patient-derived organoids (PDOs) as predictive tools in Crohn's disease (CD) that capture the past, present, and future of disease behavior, thereby advancing PDO-informed precision medicine beyond oncology into complex inflammatory diseases. HIGHLIGHTS A living PDO biobank identified two molecular CD subtypes with distinct functional phenotypes.PDO subtyping tracked severity of ileal disease, prior surgery and future disease progression.Colonic immune dysfunction may drive ileal-CD, independent of colonic involvement.Colonic CD-PDOs are dynamic platforms for outcome-deterministic therapeutic testing.
Collapse
|
29
|
Xiu J, Xue R, Duan X, Yao F, Liu X, Meng F, Xiong C, Huang J. Mechanical characterization of nonlinear elasticity of growing intestinal organoids with a microinjection method. Acta Biomater 2025; 196:271-280. [PMID: 40032216 DOI: 10.1016/j.actbio.2025.02.054] [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: 10/15/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
Mechanical properties of intestinal organoids are crucial for intestinal development, homeostatic renewal, and pathogenesis. However, characterizing these properties remains challenging. Here, we developed a microinjection-based method to quantify the growth time-dependent nonlinear elasticity of intestinal organoids. With aid of the neo-Hookean hyperelastic constitutive model, we discovered that the global elastic modulus of intestinal organoids increased linearly during the early stages of culture, followed by a sharp rise, indicating a time-dependent nonlinear hardening behaviour during growth. The global modulus of intestinal organoids was found to correlate with the cell phenotype ratio, revealing a significant relationship between mechanical properties and biological phenotypes. Furthermore, we developed a biomechanical model on the basis of the unsteady Bernoulli equation to quantitatively explore the global mechanical responses of intestinal organoids, which showed good agreement with the experimental data. The work not only elucidated the mechanical response and modulus characteristics of small intestinal organoids from a biomechanical perspective, but also presented a new microinjection-based methodology for quantifying the mechanical properties of organoids, offering significant potential for various organoid-related applications. STATEMENT OF SIGNIFICANCE: Mechanical properties of intestinal organoids are essential for intestinal development, homeostatic renewal, and pathogenesis. However, how to quantitatively characterize their global mechanical properties remains challenging. Here, we developed a new microinjection-based experimental platform to quantify spatiotemporal dynamics of mechanical responses and global elasticity of intestinal organoids. Unlike traditional nanoindentation methods, the proposed characterization technique can quantitatively measure the global mechanical properties of organoids, which is crucial for detecting the inherent relationship between the global mechanical properties and the biological phenotypes of organoids. Likewise, it established a methodological foundation for revealing the mechanobiological characteristics associated with the growth and development of various organoids. This can enhance our understanding of mechanobiological mechanisms of organoids and is beneficial for various organoid-related applications.
Collapse
Affiliation(s)
- Jidong Xiu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Rui Xue
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Xiaocen Duan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Fangyun Yao
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Fanlu Meng
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China.
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
30
|
Xu X, Zhang Y, Huang G, Perekatt A, Wang Y, Chen L. Advances and applications of gut organoids: modeling intestinal diseases and therapeutic development. LIFE MEDICINE 2025; 4:lnaf012. [PMID: 40276096 PMCID: PMC12018802 DOI: 10.1093/lifemedi/lnaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Gut organoids are 3D cellular structures derived from adult or pluripotent stem cells, capable of closely replicating the physiological properties of the gut. These organoids serve as powerful tools for studying gut development and modeling the pathogenesis of intestinal diseases. This review provides an in-depth exploration of technological advancements and applications of gut organoids, with a focus on their construction methods. Additionally, the potential applications of gut organoids in disease modeling, microenvironmental simulation, and personalized medicine are summarized. This review aims to offer perspectives and directions for understanding the mechanisms of intestinal health and disease as well as for developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoting Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Yuping Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Guoxin Huang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou 515041, China
| | - Ansu Perekatt
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Yan Wang
- Center for Translation Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
- Institute of Microphysiological Systems, Southeast University, Nanjing 211189, China
| |
Collapse
|
31
|
Yang J, Li W, Zhang Z, Xu Z, Zhu W, Wang J, Wang W. Development and Applications of Organoids in Gynecological Diseases. Stem Cell Rev Rep 2025; 21:629-644. [PMID: 39666266 PMCID: PMC11965162 DOI: 10.1007/s12015-024-10833-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Organoids are rapidly self-organizing 3D in vitro cultures derived from pluripotent stem cells (PSCs) or adult stem cells (ASCs) that possess disease-like characteristics with high success rates. Due to their ability to retain tissue structure, biological phenotypes, and genetic information, they have been utilized as a novel in vitro model for disease research. In recent years, scientists have established self-organizing 3D organoids for human endometrium, fallopian tubes, ovaries, and cervix by culturing stem cells with cytokines in 3D scaffolds. The integration of organoids with animal models, organ-on-a-chip systems, and 3D printing technologies offers a novel preclinical model for exploring disease mechanisms and developing treatments. This review elaborate on the recent research progress of stem cells-formed organoids in the field of gynecology from the aspects of constructing gynecological disease organoids, drug screening and new drug development, simulation modeling, allogeneic transplantation, regenerative medicine and personalized treatment."
Collapse
Affiliation(s)
- Jian Yang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenwen Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zihan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhonglei Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenjing Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Anhui Women and Children's Medical Center, Hefei, Anhui, China
| | - Wenyan Wang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
32
|
Kato Y, Seishima R, Hattori K, Kato H, Ishida H, Shigeta K, Okabayashi K, Sugihara E, Takimoto T, Nakamura K, Nishihara H, Saya H, Kitagawa Y. Significance of homologous recombinant deficiency as a biomarker for drug sensitivity in colorectal cancer. Br J Cancer 2025; 132:533-542. [PMID: 39934338 PMCID: PMC11920058 DOI: 10.1038/s41416-025-02950-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/20/2024] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a substantial global health concern due to its limited treatment options, especially for oxaliplatin (L-OHP) regimen resistance. This study used organoid-based screening methodologies to evaluate drug responses in CRC while validating the approach with patient-derived CRC organoids and investigating potential biomarkers. METHODS Patient-derived organoids were created from CRC surgical specimens, and drug screening were performed. Selected organoids with high and low L-OHP sensitivity underwent next-generation sequencing (NGS), and in vivo experiments using xenotransplantation were used to validate in vitro results. Moreover, the clinical application of homologous recombination deficiency (HRD) as a biomarker was investigated. RESULTS Organoid drug screening revealed differences in L-OHP sensitivity among 34 patient-derived CRC organoids, and NGS deemed HRD as a potential biomarker. In vivo experiments validated the correlation between HRD status and L-OHP sensitivity, and clinical data suggested the potential of HRD as a biomarker for recurrence-free survival in patients treated with L-OHP. Additionally, HRD exhibited potential as a biomarker for other platinum agents and poly (ADP-ribose) polymerase inhibitors in CRC. CONCLUSIONS The study underscores HRD as a potential biomarker for predicting L-OHP sensitivity, expanding its application to other drugs in CRC. Organoid screening is reliable, providing insights into the intricate association between genetic features and treatment responses.
Collapse
Affiliation(s)
- Yujin Kato
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Ryo Seishima
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Department of Surgery, Fujita Health University, Toyoake, 470-1192, Japan.
| | - Kaoru Hattori
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hirochika Kato
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroki Ishida
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kohei Shigeta
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Eiji Sugihara
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, 470-1192, Japan
- Research Promotion Headquarters, Open Facility Center, Fujita Health University, Toyoake, 470-1192, Japan
| | - Tetsuya Takimoto
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, 470-1192, Japan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, 470-1192, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| |
Collapse
|
33
|
Yao N, Jing N, Lin J, Niu W, Yan W, Yuan H, Xiong Z, Hou Q, Qiao X, Liu Q, Cao J, Li N. Patient-derived tumor organoids for cancer immunotherapy: culture techniques and clinical application. Invest New Drugs 2025; 43:394-404. [PMID: 40232355 PMCID: PMC12048417 DOI: 10.1007/s10637-025-01523-w] [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: 11/15/2024] [Accepted: 03/03/2025] [Indexed: 04/16/2025]
Abstract
Cancer immunotherapy has revolutionized tumor treatment. However, robust and effective testing platforms remain lacking, especially for the selection of the optimized therapy at the patient-specific level. Unlike conventional treatment evaluations, testing platforms for cancer immunotherapy must incorporate not only tumor cells but also the tumor microenvironment (TME), including immune components. Recently, emergence of patient-derived tumor organoids (PDTOs), an in vitro preclinical model, has provided a novel approach for studying tumor evolution and assessing treatment responses, and shows great potential when coculturing with immune cells to study the mechanisms of immunotherapy efficacy and resistance. However, traditional organoid technology is limited in capturing the full impact of the TME on tumor behaviors due to the absence of stromal components. To circumvent these restrictions, complex organoid cocultures with immune cells, cancer-associated fibroblasts and vasculatures are developed. In this review, we summarized recent advances in PDTO culture techniques for modeling the TME and explored the application of complex tumor organoids in cancer immunotherapy.
Collapse
Affiliation(s)
- Ningning Yao
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China
| | - Na Jing
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China
| | - Jianzhong Lin
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Wenxia Niu
- Department of Oncology, The Second Clinical Medical College of Shanxi Medical University, Shanxi Medical University, Taiyuan, 030001, China
| | - Wenxing Yan
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China
| | - Hongqin Yuan
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China
| | - Zeyi Xiong
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China
| | - Qing Hou
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China
| | - Xiaxi Qiao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Quanming Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianzhong Cao
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China.
| | - Ning Li
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No.3, Zhigong New Street, Xinghualing District, Taiyuan, 030013, China.
| |
Collapse
|
34
|
Dong Y, Li J, Dai Y, Zhang X, Wang T, Zhao B, Liu W, Chen L, Yang S, Du P, Jiao Z. Redox-responsive metal-organic framework nanocapsules enhance tumor chemo-immunotherapy by modulating tumor metabolic reprogramming. Mater Today Bio 2025; 31:101487. [PMID: 39896279 PMCID: PMC11786678 DOI: 10.1016/j.mtbio.2025.101487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/04/2025] [Accepted: 01/11/2025] [Indexed: 02/04/2025] Open
Abstract
Immunotherapy, particularly immune checkpoint blockade (ICB), has opened the era of modern oncology, offering significant promise for modern oncology. However, the efficacy of immunotherapy is frequently curtailed by the immunosuppressive tumor microenvironment (ITM), a milieu shaped by tumor metabolic reprogramming. Herein, a novel tumor microenvironment-responsive nanocapsules (DNMCs) were developed that simultaneously modulate tumor metabolism and the ITM to enhance the effectiveness of chemo-immunotherapy. DNMCs consist of an acidic and redox-sensitive metal-organic framework (MOF) encapsulating Doxorubicin (DOX) and the indoleamine-2,3-dioxygenase1 (IDO1) inhibitor NLG919. In the tumor microenvironment, DNMCs degrade, rapidly releasing DOX and NLG919. DOX induces immunogenic cell death (ICD), while NLG919 regulates amino acid metabolism by modulating IDO1 activity, thereby reversing the immunosuppressive of ITM. Consequently, DNMCs elicit effective anti-tumor immune responses, characterized by an increased density of tumor-infiltrating CD8+ cytotoxic T cells as well as depletion of immunosuppressive regulatory T cells (Tregs), thus effectively suppressing pancreatic cancer growth in KPC mice through combined chemo-immunotherapy. Overall, DNMCs exhibit significant tumor growth inhibition in pancreatic cancer patient-derived organoids (PDOs) and mouse models. This study presents a promising approach to enhancing chemo-immunotherapy by targeting tumor metabolic reprogramming and augmenting immune response against malignant tumors.
Collapse
Affiliation(s)
- Yuman Dong
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Jieru Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Yiwei Dai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Xinyu Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Tao Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Bin Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Wenbo Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Li Chen
- Department of Orthopaedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Shaopei Yang
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Pengcheng Du
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Zuoyi Jiao
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| |
Collapse
|
35
|
Qin T, Wang J, Wang J, Du Q, Wang L, Liu H, Liu W, Li X, Jiang Y, Xu Q, Yu J, Liu H, Wang T, Li M, Huang D. Nuclear to Cytoplasmic Transport Is a Druggable Dependency in HDAC7-driven Small Cell Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413445. [PMID: 39887933 PMCID: PMC11984897 DOI: 10.1002/advs.202413445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Immunotherapy has gained approval for use in small cell lung cancer (SCLC), yet only a subset of patients (10-20%) experience meaningful benefits, underscoring the urgent need for more effective therapeutic approaches. This work discovers a distinct HDAC7-high SCLC phenotype characterized by enhanced proliferative potential, which recurs across various subtypes and serves as a predictor of poorer survival outcomes. By analyzing public datasets, this work finds a strong correlation between c-Myc and HDAC7. RNA sequencing and cellular experiments show that XPO1 is a key regulator in the HDAC7/c-Myc axis. HDAC7 promotes β-catenin deacetylation, phosphorylation modulation, nuclear translocation, and formation of the β-catenin/TCF/LEF1 complex, which binds to c-Myc and XPO1 promoters. Activation of the HDAC7/β-catenin pathway upregulates c-Myc and XPO1 expression, while c-Myc also boosts XPO1 expression. Given the difficulty in targeting c-Myc directly, this work tests selinexor and vorinostat in SCLC xenograft models, with selinexor showing superior results. High HDAC7 expression is linked to increased SCLC proliferation, poorer prognosis, and enhanced sensitivity to selinexor in SCLC cell lines and organoid models. Collectively, this work uncovers a novel HDAC7/c-Myc/XPO1 signaling axis that promotes SCLC progression, suggesting that HDAC7 may warrant further investigation as a potential biomarker for assessing selinexor sensitivity in SCLC patients.
Collapse
Affiliation(s)
- Tingting Qin
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Jingya Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Jian Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Qingwu Du
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Liuchun Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Hailin Liu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Wenting Liu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Xueyang Li
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Yantao Jiang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Qi Xu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Junjie Yu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Huiyan Liu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Ting Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Mengjie Li
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Dingzhi Huang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060
- Tianjin's Clinical Research Center for CancerDepartment of Thoracic OncologyTianjin Lung Cancer CenterTianjin Cancer Institute & HospitalTianjin Medical UniversityTianjin300060P. R. China
| |
Collapse
|
36
|
Scholtes MP, Akbarzadeh M, Galaras A, Nakauma-Gonzáles JA, Bazrafshan A, Solanki V, Torenvliet B, Beikmohammadi L, Lozovanu V, Romal S, Moulos P, Vavouraki N, Kan TW, Algoe M, van Royen ME, Sacchetti A, van den Bosch TPP, Eussen B, de Klein A, van Leenders GJLH, Boormans JL, Hatzis P, Palstra RJ, Zuiverloon TCM, Mahmoudi T. Integrative analysis of patient-derived tumoroids and ex vivo organoid modelling of ARID1A loss in bladder cancer reveals therapeutic molecular targets. Cancer Lett 2025; 614:217506. [PMID: 39892702 DOI: 10.1016/j.canlet.2025.217506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/06/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
Somatic mutations in ARID1A (AT-rich interactive domain-containing protein 1A) are present in approximately 25 % of bladder cancers (BC) and are associated with poor prognosis. With a view to discover effective treatment options for ARID1A-deficient BC patients, we set out to identify targetable effectors dysregulated consequent to ARID1A deficiency. Integrative analyses of ARID1A depletion in normal organoids and data mining in publicly available datasets revealed upregulation of DNA repair and cell cycle-associated genes consequent to loss of ARID1A and identified CHEK1 (Checkpoint kinase 1) and chromosomal passenger complex member BIRC5 (Baculoviral IAP Repeat Containing 5) as therapeutically drug-able candidate molecular effectors. Ex vivo treatment of patient-derived BC tumoroids with clinically advanced small molecule inhibitors targeting CHEK1 or BIRC5 was associated with increased DNA damage signalling and apoptosis, and selectively induced cell death in tumoroids lacking ARID1A protein expression. Thus, integrating public datasets with patient-derived organoid modelling and ex-vivo drug testing can uncover key molecular effectors and mechanisms of oncogenic transformation, potentially leading to novel therapeutic strategies. Our data point to ARID1A protein expression as a suitable candidate biomarker for the selection of BC patients responsive to therapies targeting BIRC5 and CHEK1.
Collapse
Affiliation(s)
- Mathijs P Scholtes
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maryam Akbarzadeh
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Biochemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Alexandros Galaras
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Center "Alexander Fleming", the Netherlands; Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - J Alberto Nakauma-Gonzáles
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ameneh Bazrafshan
- Department of Biochemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Vandana Solanki
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Bram Torenvliet
- Department of Biochemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Leila Beikmohammadi
- Department of Biochemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Valeria Lozovanu
- Department of Biochemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Shahla Romal
- Department of Biochemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Panagiotis Moulos
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Center "Alexander Fleming", the Netherlands; Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Nikoleta Vavouraki
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Center "Alexander Fleming", the Netherlands
| | - Tsung Wai Kan
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mahesh Algoe
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrea Sacchetti
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Thierry P P van den Bosch
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Bert Eussen
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annelies de Klein
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Geert J L H van Leenders
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Joost L Boormans
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Pantelis Hatzis
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Center "Alexander Fleming", the Netherlands
| | - Robert-Jan Palstra
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Tahlita C M Zuiverloon
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Tokameh Mahmoudi
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
37
|
Yang Q, Li M, Xiao Z, Feng Y, Lei L, Li S. A New Perspective on Precision Medicine: The Power of Digital Organoids. Biomater Res 2025; 29:0171. [PMID: 40129676 PMCID: PMC11931648 DOI: 10.34133/bmr.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/26/2025] Open
Abstract
Precision medicine is a personalized medical model based on the individual's genome, phenotype, and lifestyle that provides tailored treatment plans for patients. In this context, tumor organoids, a 3-dimensional preclinical model based on patient-derived tumor cell self-organization, combined with digital analysis methods, such as high-throughput sequencing and image processing technology, can be used to analyze the genome, transcriptome, and cellular heterogeneity of tumors, so as to accurately track and assess the growth process, genetic characteristics, and drug responsiveness of tumor organoids, thereby facilitating the implementation of precision medicine. This interdisciplinary approach is expected to promote the innovation of cancer diagnosis and enhance personalized treatment. In this review, the characteristics and culture methods of tumor organoids are summarized, and the application of multi-omics, such as bioinformatics and artificial intelligence, and the digital methods of organoids in precision medicine research are discussed. Finally, this review explores the main causes and potential solutions for the bottleneck in the clinical translation of digital tumor organoids, proposes the prospects of multidisciplinary cooperation and clinical transformation to narrow the gap between laboratory and clinical settings, and provides references for research and development in this field.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Yekai Feng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital,
Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
38
|
Peng Z, Lv X, Sun H, Zhao L, Huang S. 3D tumor cultures for drug resistance and screening development in clinical applications. Mol Cancer 2025; 24:93. [PMID: 40119343 PMCID: PMC11927140 DOI: 10.1186/s12943-025-02281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
Tumor drug resistance presents a growing challenge in medical practice, particularly during anti-cancer therapies, where the emergence of drug-resistant cancer cells significantly complicates clinical treatment. In recent years, three-dimensional (3D) tumor culture technology, which more effectively simulates the in vivo physiological environment, has gained increasing attention in tumor drug resistance research and clinical applications. By mimicking the in vivo cellular microenvironment, 3D tumor culture technology not only recapitulates cell-cell interactions but also more faithfully reproduces the biological effects of therapeutic agents. Consequently, 3D tumor culture technology is emerging as a crucial tool in biomedical and clinical research. We summarize the benefits of 3D culture models and organoid technology, explore their application in the realm of drug resistance, drug screening, and personalized therapy, and discuss their potential application prospects and challenges in clinical transformation, with the aim of providing insights for optimizing cancer treatment strategies and advancing precision therapy.
Collapse
Affiliation(s)
- Zheng Peng
- Department of Clinical Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, Guangxi, China
| | - Xiaolan Lv
- Department of Clinical Laboratory, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Hao Sun
- Faculty of Science, Autonomous University of Madrid, Spainish National Research Council -Consejo Superior de Investigaciones Científicas,(UAM-CSIC), Madrid, 28049, Spain
| | - Lina Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Shigao Huang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
39
|
Tang J, Zheng Q, Wang Q, Zhao Y, Ananthanarayanan P, Reina C, Šabanović B, Jiang K, Yang MH, Meny CC, Wang H, Agerbaek MØ, Clausen TM, Gustavsson T, Wen C, Borghi F, Mellano A, Fenocchio E, Gregorc V, Sapino A, Theander TG, Fu D, Aicher A, Salanti A, Shen B, Heeschen C. Protocol for the creation and utilization of 3D pancreatic cancer models from circulating tumor cells. STAR Protoc 2025; 6:103635. [PMID: 39946239 PMCID: PMC11870243 DOI: 10.1016/j.xpro.2025.103635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/13/2024] [Accepted: 01/22/2025] [Indexed: 03/03/2025] Open
Abstract
We introduce a protocol for generating 3D organoids from circulating tumor cells (CTCs), enabling longitudinal functional and molecular analyses in pancreatic cancer patients, including those with unresectable disease, which constitutes the majority of cases. We outline the process for isolating and characterizing CTCs from the blood of pancreatic cancer patients and provide detailed instructions for initiating, passaging, and phenotyping CTC-derived organoids. Additionally, we describe techniques for utilizing these organoids in drug screening with a focus on stemness-related pathways. For complete details on the use and execution of this protocol, please refer to Tang et al.1.
Collapse
Affiliation(s)
- Jiajia Tang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Quan Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qi Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yaru Zhao
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Preeta Ananthanarayanan
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Chiara Reina
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Berina Šabanović
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Ke Jiang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ming-Hsin Yang
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Clara Csilla Meny
- 2(nd) Institute for Pathology and Experimental Oncology Research, Semmelweis University, 1085 Budapest, Hungary
| | - Huimin Wang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mette Ø Agerbaek
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen N, Denmark; VarCT Diagnostics, Ole Maaloes Vej 3, 2200 Copenhagen, Denmark
| | - Thomas Mandel Clausen
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tobias Gustavsson
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen N, Denmark; VAR2Pharmaceuticals, Ole Maaloes Vej 3, 2200 Copenhagen, Denmark
| | - Chenlei Wen
- Research Institute of Pancreatic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Pancreatic Disease Centre, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Felice Borghi
- Department of Surgical Oncology, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Alfredo Mellano
- Department of Surgical Oncology, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Elisabetta Fenocchio
- Department of Medical Oncology, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Vanesa Gregorc
- Department of Medical Oncology, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Anna Sapino
- Department of Pathology, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy
| | - Thor G Theander
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Da Fu
- Research Institute of Pancreatic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Pancreatic Disease Centre, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alexandra Aicher
- Precision Immunotherapy, Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404328, Taiwan
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Baiyong Shen
- Research Institute of Pancreatic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Pancreatic Disease Centre, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Christopher Heeschen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute - FPO - IRCCS, 10060 Candiolo (TO), Italy.
| |
Collapse
|
40
|
Dondi C, Tsikritsis D, Vorng JL, Greenidge G, Kepiro IE, Belsey NA, McMahon G, Gilmore IS, Ryadnov MG, Shaw M. Multiparametric physicochemical analysis of a type 1 collagen 3D cell culture model using light and electron microscopy and mass spectrometry imaging. Sci Rep 2025; 15:9578. [PMID: 40113888 PMCID: PMC11926111 DOI: 10.1038/s41598-025-93700-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Three-dimensional cell culture systems underpin cell-based technologies ranging from tissue scaffolds for regenerative medicine to tumor models and organoids for drug screening. However, to realise the full potential of these technologies requires analytical methods able to capture the diverse information needed to characterize constituent cells, scaffold components and the extracellular milieu. Here we describe a multimodal imaging workflow which combines fluorescence, vibrational and second harmonic generation microscopy with secondary ion mass spectrometry imaging and transmission electron microscopy to analyse the morphological, chemical and ultrastructural properties of cell-seeded scaffolds. Using cell nuclei as landmarks we register fluorescence with label-free optical microscopy images and high mass resolution with high spatial resolution secondary ion mass spectrometry images, with an accuracy comparable to the intrinsic spatial resolution of the techniques. We apply these methods to investigate relationships between cell distribution, cytoskeletal morphology, scaffold fiber organisation and biomolecular composition in type I collagen scaffolds seeded with human dermal fibroblasts.
Collapse
Affiliation(s)
- Camilla Dondi
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | | | - Jean-Luc Vorng
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Gina Greenidge
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Ibolya E Kepiro
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Natalie A Belsey
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
- School of Chemistry and Chemical Engineering, University of Surrey, Guildford, GU2 7XH, UK
| | - Greg McMahon
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Ian S Gilmore
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Maxim G Ryadnov
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Michael Shaw
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK.
- UCL Hawkes Institute and Department of Computer Science, University College London, London, UK.
| |
Collapse
|
41
|
Taglieri M, Di Gregorio L, Matis S, Uras CRM, Ardy M, Casati S, Marchese M, Poggi A, Raffaghello L, Benelli R. Colorectal Organoids: Models, Imaging, Omics, Therapy, Immunology, and Ethics. Cells 2025; 14:457. [PMID: 40136707 PMCID: PMC11941511 DOI: 10.3390/cells14060457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Colorectal epithelium was the first long-term 3D organoid culture established in vitro. Identification of the key components essential for the long-term survival of the stem cell niche allowed an indefinite propagation of these cultures and the modulation of their differentiation into various lineages of mature intestinal epithelial cells. While these methods were eventually adapted to establish organoids from different organs, colorectal organoids remain a pioneering model for the development of new applications in health and disease. Several basic and applicative aspects of organoid culture, modeling, monitoring and testing are analyzed in this review. We also tackle the ethical problems of biobanking and distribution of these precious research tools, frequently confined in the laboratory of origin or condemned to destruction at the end of the project.
Collapse
Affiliation(s)
- Martina Taglieri
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Linda Di Gregorio
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Serena Matis
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Chiara Rosa Maria Uras
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Massimo Ardy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Sara Casati
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” CNR, 80131 Naples, Italy;
- Common Service ELSI, BBMRI.it (UNIMIB National Node Headquarter), 20126 Milan, Italy
| | - Monica Marchese
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Alessandro Poggi
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Lizzia Raffaghello
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Roberto Benelli
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| |
Collapse
|
42
|
Gao T, He X, Wang J, Liu J, Hu X, Bai C, Yin S, Shi Y, Wang Y, Tan Z, Cao F, Li S, Shi YJ, Xue R, Li J, He Y, Li J, Lu H, Zhang H, Zhang L, Fang Z, Wang X, Liu M, Fu W, Tang L, Ye B, Fan Z, Xi JJ. Self-assembled patient-derived tumor-like cell clusters for personalized drug testing in diverse sarcomas. Cell Rep Med 2025; 6:101990. [PMID: 40054460 PMCID: PMC11970405 DOI: 10.1016/j.xcrm.2025.101990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/30/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Several patient-derived tumor models have emerged recently. However, soft tissue sarcomas (STSs) present a challenge in developing preclinical drug-testing models due to their non-epithelial and complex nature. Here, we report a model termed patient-derived tumor-like cell clusters (PTCs) derived from STS patients. PTCs result from the self-assembly and proliferation of mesenchymal stem cells (MSCs), epithelial cells, and immune cells, faithfully recapitulating the morphology and function of the original tumors. Through standardized culture and drug-response assessment protocols, PTCs facilitate personalized drug testing, evaluating hundreds of therapies within two weeks. Notably, PTCs exhibit 100% accuracy in distinguishing between complete or partial response and disease progression. We demonstrate the utility of PTCs in guiding chemotherapy selection for a patient with relapse and metastases following conventional therapy, who exhibited a positive response after non-conventional therapy identified through PTC. These findings underscore the potential of PTCs for prospective use in clinical decision-making regarding therapy selection.
Collapse
Affiliation(s)
- Tian Gao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Junyi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiongbing Hu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Chujie Bai
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shenyi Yin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Yunfei Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanmin Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhichao Tan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shu Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yan-Jie Shi
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ruifeng Xue
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Juan Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Yang He
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jiaxin Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Huinan Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China
| | - Hanshuo Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; GeneX Health Co., Ltd., Beijing 100195, China
| | - Lu Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhiwei Fang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xinyu Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mengmeng Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Wenjun Fu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lei Tang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Buqing Ye
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhengfu Fan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Jianzhong Jeff Xi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China; Peking University Yangtze Center of Future Health Technology, Wuxi 214111, China.
| |
Collapse
|
43
|
Su H, Chen L, Wu J, Cheng Z, Li J, Ren Y, Xu J, Dang Y, Zheng M, Cao Y, Gao J, Dai C, Hu X, Xie H, Chen J, Luo T, Zhu J, Wu C, Sha W, Chen C, Liu H. Proteogenomic characterization reveals tumorigenesis and progression of lung cancer manifested as subsolid nodules. Nat Commun 2025; 16:2414. [PMID: 40069142 PMCID: PMC11897189 DOI: 10.1038/s41467-025-57364-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Lung adenocarcinoma (LUAD) radiologically displayed as subsolid nodules (SSNs) is prevalent. Nevertheless, the precise clinical management of SSNs necessitates a profound understanding of their tumorigenesis and progression. Here, we analyze 66 LUAD displayed as SSNs covering 3 histological stages including adenocarcinoma in situ (AIS), minimally invasive adenocarcinoma (MIA) and invasive adenocarcinoma (IAC) by incorporating genomics, proteomics, phosphoproteomics and glycoproteomics. Intriguingly, cholesterol metabolism is aberrantly regulated in the preneoplastic AIS stage. Importantly, target ablation of proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the initiation of LUAD. Furthermore, sustained endoplasmic reticulum stress is demonstrated to be a hallmark and a reliable biomarker of AIS progression to IAC. Consistently, target promotion of ER stress profoundly retards LUAD progression. Our study provides comprehensive proteogenomic landscape of SSNs, sheds lights on the tumorigenesis and progression of SSNs and suggests preventive and therapeutic strategies for LUAD.
Collapse
Affiliation(s)
- Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Li Chen
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhongyi Cheng
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jing Li
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Junfang Xu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yifang Dang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Mengge Zheng
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Yajuan Cao
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jiani Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Tao Luo
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Jun Zhu
- Jingjie PTM BioLab (Hangzhou). Co. Inc, Hangzhou, 310000, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Wei Sha
- Department of tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Haipeng Liu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| |
Collapse
|
44
|
Langenberg KPS, van Hooff SR, Koopmans B, Strijker JGM, Kholosy WM, Ober K, Zwijnenburg DA, van der Hoek JJF, Keller KM, Vernooij L, Schild LG, Looze EJ, Ebus ME, Essing AHW, Vree PD, Tas ML, Matser YAH, Wienke J, Volckmann R, Tops BBJ, Kester LA, Badloe S, Hehir-Kwa JY, Kemmeren P, Goemans BF, Zwaan CM, Oehme I, Jäger N, Witt O, van Eijkelenburg NKA, Dierselhuis MP, Tytgat GAM, Wijnen MHW, van Noesel MM, de Krijger RR, Eising S, Koster J, Dolman EM, Molenaar JJ. Exploring high-throughput drug sensitivity testing in neuroblastoma cell lines and patient-derived tumor organoids in the era of precision medicine. Eur J Cancer 2025; 218:115275. [PMID: 39954414 PMCID: PMC11884408 DOI: 10.1016/j.ejca.2025.115275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Despite druggable events to be present in 80 % of neuroblastomapatients within the Princess Máxima Center precision medicine program 'iTHER', clinical uptake of treatment recommendations has been low, and the clinical impact for individual patients remains hard to predict. This stresses the need for a method integrating genomics and transcriptomics with functional approaches into therapeutic decision making. METHODS We aimed to launch an online repository integrating genomics and transcriptomics with high-throughput drug screening (HTS) of nineteen commonly used neuroblastoma cell lines and fifteen neuroblastoma patient-derived organoids (NBL-PDOs). Cell lines, NBL-PDOs and their parental tumors were characterized utilizing (lc)WGS, WES and RNAseq. Cells were exposed to ∼200 compounds. Results were transferred to the R2 visualization platform. RESULTS A powerful reference set of cell lines is available, reflecting distinct known pharmacologic vulnerabilities. HTS identified additional therapeutic vulnerabilities, such as a striking correlation between a positive mesenchymal signature and sensitivity to BCL2-inhibitor venetoclax. Finally, we explored personalized drug sensitivities within iTHER, demonstrating HTS can support genomic and transcriptomic results, thereby strengthening the rationale for clinical uptake. CONCLUSION We established a dynamic publicly available dataset with detailed genomic, transcriptomic, and pharmacological annotation of classical neuroblastoma cell lines as well as novel sharable NBL-PDOs, representing the heterogeneous landscape of neuroblastoma. We anticipate that in vitro drug screening will be complementary to genomic-guided precision medicine by supporting clinical decision making, thereby improving prognosis for all neuroblastoma patients in the future.
Collapse
Affiliation(s)
- Karin P S Langenberg
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Sander R van Hooff
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Bianca Koopmans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Josephine G M Strijker
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Waleed M Kholosy
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Kimberley Ober
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Danny A Zwijnenburg
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Jessica J F van der Hoek
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Kaylee M Keller
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Lindy Vernooij
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Linda G Schild
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Eleonora J Looze
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Marli E Ebus
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Anke H W Essing
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Paula de Vree
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Michelle L Tas
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Yvette A H Matser
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Judith Wienke
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Richard Volckmann
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Bastiaan B J Tops
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Lennart A Kester
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Shashi Badloe
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jayne Y Hehir-Kwa
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Patrick Kemmeren
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Bianca F Goemans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 430, Heidelberg 69120, Germany.
| | - Nathalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands.
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 430, Heidelberg 69120, Germany; Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital,National Center for Tumor Diseases (NCT) Network, Heidelberg, Germany.
| | | | - Miranda P Dierselhuis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Marc H W Wijnen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Division Imaging & Cancer, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Selma Eising
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jan Koster
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Emmy M Dolman
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| |
Collapse
|
45
|
Ahmed A, Cox E, Lane L, Rominiyi O, Danson S, Bryant HE, Wells G, King D. Ex Vivo Drug Screening: An Emerging Paradigm in the Treatment of Childhood Cancer. J Pediatr Hematol Oncol 2025:00043426-990000000-00553. [PMID: 40085807 DOI: 10.1097/mph.0000000000003017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 03/16/2025]
Abstract
Developing and providing the right therapy for the right patient (or personalized targeted treatments) is key to reducing side-effects and improving survival in childhood cancers. Most efforts aiming to personalize childhood cancer treatment use genomic analysis of malignancies to identify potentially targetable genetic events. But it is becoming clear that not all patients will have an actionable change, and in those that do there is no additional way to determine if treatments will be effective. Ex vivo drug screening is a laboratory technique used to test the effects of various drugs or compounds, on biological tissues or cells that have been removed from an organism. This information is then used to predict which cancer treatments will be most effective based on the therapeutic response in the tissue or cells removed from that individual. Its utility in personalizing treatments in childhood cancer is increasingly recognized. In this review we describe the different methods for ex vivo drug screening and the advantages and disadvantages of each technique. We also present recent evidence that ex vivo screening may have utility in a variety of childhood malignancies including an overview of current clinical trials appraising its use. Finally, we discuss the research questions and hurdles that must be overcome before ex vivo screening can be widely used in pediatric oncology.
Collapse
Affiliation(s)
- Anees Ahmed
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, The University of Sheffield
| | - Ellen Cox
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, The University of Sheffield
| | - Louis Lane
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, The University of Sheffield
| | - Ola Rominiyi
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, The University of Sheffield
- Department of Neurosurgery, Royal Hallamshire Hospital
| | - Sarah Danson
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, The University of Sheffield
- Department of Oncology, Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust
| | - Helen E Bryant
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, The University of Sheffield
| | - Greg Wells
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, The University of Sheffield
| | - David King
- Department of Paediatric Oncology, Sheffield Children's Hospital, Sheffield, UK
| |
Collapse
|
46
|
Mercadante M, Scheben A, Estrada J, Savas-Carstens J, Sullivan W, Housel N, Volpari T, Hebner J, Sapar M, Rusielewicz T, Monsma FJ, Semrau S, Wang Y, Martin LA. A patient-derived ovarian cancer organoid platform to study susceptibility to natural killer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641285. [PMID: 40093054 PMCID: PMC11908259 DOI: 10.1101/2025.03.06.641285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Intratumoral heterogeneity drives therapy resistance and relapses in advanced stage cancers, such as ovarian cancer. Here, we present a live cell imaging assay using patient-derived ovarian cancer organoids for real time capture and quantification of natural killer cell-mediated apoptotic events in >500 organoids simultaneously. Our assay revealed significant inter- and intratumor response heterogeneity and identified a rare resistant organoid population, opening avenues to test immunomodulatory strategies that overcome resistance.
Collapse
Affiliation(s)
| | - Armin Scheben
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Jacob Estrada
- The New York Stem Cell Foundation Research Institute, New York, NY
| | | | - William Sullivan
- The New York Stem Cell Foundation Research Institute, New York, NY
| | | | - Tatiana Volpari
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Jax Hebner
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Maria Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Tom Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY
| | | | - Stefan Semrau
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Yinan Wang
- The New York Stem Cell Foundation Research Institute, New York, NY
| | - Laura A Martin
- The New York Stem Cell Foundation Research Institute, New York, NY
| |
Collapse
|
47
|
Liu D, Chen Z, Deng W, Lan J, Zhu Y, Wang H, Xu X, Zhang Y, Wu X, Yang K, Cai J. An Organoid Model for the Therapeutic Effect of Hyperthermic Intraperitoneal Chemotherapy for Colorectal Cancer. Ann Surg Oncol 2025; 32:1925-1940. [PMID: 39589577 PMCID: PMC11811434 DOI: 10.1245/s10434-024-16469-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Consensus regarding the hyperthermic intraperitoneal chemotherapy (HIPEC) for colorectal cancer (CRC) regimen remains elusive. In this study, patient-derived tumor organoids from CRC were utilized as a preclinical model for in vitro drug testing of HIPEC regimens commonly used in clinical practice. This approach was used to facilitate the clinical formulation of HIPEC. METHOD Tumor tissues and corresponding clinical data were obtained from patients diagnosed with CRC at the Sixth Affiliated Hospital of Sun Yat-Sen University. Qualified samples were cultured and passaged. We aimed to assess the sensitivity of in vitro hyperthermic perfusion using five different regimens, i.e. mitomycin C, mitomycin C combined with cisplatin, mitomycin C combined with 5-fluorouracil, oxaliplatin, and oxaliplatin combined with 5-fluorouracil. RESULTS Tumor organoids obtained from 46 patients with CRC were cultured, and in vitro hyperthermic perfusion experiments were conducted on 42 organoids using five different regimens. The average inhibition rate of mitomycin C was 85.2% (95% confidence interval [CI] 80.4-89.9%), mitomycin C combined with cisplatin was 85.5% (95% CI 80.2-90.7%), mitomycin C combined with 5-fluorouracil was 65.6% (95% CI 59.6-71.6%), oxaliplatin was 37.9% (95% CI 31.5-44.3%), and oxaliplatin combined with 5-fluorouracil was 40.7% (95% CI 33.9-47.5%). CONCLUSION In vitro hyperthermic perfusion demonstrates that the inhibition rate of mitomycin C, both alone and in combination with cisplatin, surpasses that of the combination of mitomycin C with 5-fluorouracil and oxaliplatin. In clinical practice, the combination of mitomycin C and cisplatin can be regarded as the optimal choice for HIPEC in CRC.
Collapse
Affiliation(s)
- Duo Liu
- Department of Colorectal Surgery, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
| | - Zexin Chen
- Guangdong Research Center of Organoid Engineering and Technology, Accurate International Biotechnology Co. Ltd., Guangzhou, China
| | - Weihao Deng
- Department of Pathology, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianqiang Lan
- Guangdong Research Center of Organoid Engineering and Technology, Accurate International Biotechnology Co. Ltd., Guangzhou, China
| | - Yu Zhu
- Guangdong Research Center of Organoid Engineering and Technology, Accurate International Biotechnology Co. Ltd., Guangzhou, China
| | - Huaiming Wang
- Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xing Xu
- Department of Breast and Thyroid Surgery, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
| | - Yuanxin Zhang
- Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiangwei Wu
- Qiantang Biotechnology Co. Ltd., Suzhou, China
| | - Keli Yang
- Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Jian Cai
- Department of Colorectal Surgery, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China.
| |
Collapse
|
48
|
Nojima Y, Yao R, Suzuki T. Single-cell RNA sequencing and machine learning provide candidate drugs against drug-tolerant persister cells in colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167693. [PMID: 39870146 DOI: 10.1016/j.bbadis.2025.167693] [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: 11/25/2024] [Revised: 12/24/2024] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Drug resistance often stems from drug-tolerant persister (DTP) cells in cancer. These cells arise from various lineages and exhibit complex dynamics. However, effectively targeting DTP cells remains challenging. We used single-cell RNA sequencing (scRNA-Seq) data and machine learning (ML) models to identify DTP cells in patient-derived organoids (PDOs) and computationally screened candidate drugs targeting these cells in familial adenomatous polyposis (FAP), associated with a high risk of colorectal cancer. Three PDOs (benign and malignant tumor organoids and a normal organoid) were evaluated using scRNA-Seq. ML models constructed based on public scRNA-Seq data classified DTP versus non-DTP cells. Candidate drugs for DTP cells in a malignant tumor organoid were identified from public drug sensitivity data. From FAP scRNA-Seq data, a specific TC1 cell cluster in tumor organoids was identified. The ML model identified up to 36 % of TC1 cells as DTP cells, a higher proportion than those for other clusters. A viability assay using a malignant tumor organoid demonstrated that YM-155 and THZ2 exert synergistic effects with trametinib. The constructed ML model is effective for DTP cell identification based on scRNA-Seq data for FAP and provides candidate treatments. This approach may improve DTP cell targeting in the treatment of colorectal and other cancers.
Collapse
Affiliation(s)
- Yosui Nojima
- Center for Mathematical Modeling and Data Science, Osaka University, 1-3 Machikaneyama, Toyonaka, Osaka 560-8531, Japan.
| | - Ryoji Yao
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| | - Takashi Suzuki
- Center for Mathematical Modeling and Data Science, Osaka University, 1-3 Machikaneyama, Toyonaka, Osaka 560-8531, Japan.
| |
Collapse
|
49
|
Richter KM, Wrage M, Krekeler C, De Oliveira T, Conradi LC, Menck K, Bleckmann A. Model systems to study tumor-microbiome interactions in early-onset colorectal cancer. EMBO Mol Med 2025; 17:395-413. [PMID: 39948421 PMCID: PMC11903813 DOI: 10.1038/s44321-025-00198-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/13/2025] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) is a major health problem, with an alarming increase of early-onset CRC (EO-CRC) cases among individuals under 50 years of age. This trend shows the urgent need for understanding the underlying mechanisms leading to EO-CRC development and progression. There is significant evidence that the gut microbiome acts as a key player in CRC by triggering molecular changes in the colon epithelium, leading to tumorigenesis. However, a comprehensive collection and comparison of methods to study such tumor-microbiome interactions in the context of EO-CRC is sparse. This review provides an overview of the available in vivo, ex vivo as well as in vitro approaches to model EO-CRC and assess the effect of gut microbes on tumor development and growth. By comparing the advantages and limitations of each model system, it highlights that, while no single model is perfect, each is suitable for studying specific aspects of microbiome-induced tumorigenesis. Taken together, multifaceted approaches can simulate the human body's complexity, aiding in the development of effective treatment and prevention strategies for EO-CRC.
Collapse
Affiliation(s)
- Katharina M Richter
- Department of Medicine A, University of Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Marius Wrage
- Department of Medicine A, University of Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Carolin Krekeler
- Department of Medicine A, University of Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, 37075, Goettingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, 37075, Goettingen, Germany
| | - Kerstin Menck
- Department of Medicine A, University of Muenster, 48149, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany
| | - Annalen Bleckmann
- Department of Medicine A, University of Muenster, 48149, Muenster, Germany.
- West German Cancer Center, University Hospital Muenster, 48149, Muenster, Germany.
| |
Collapse
|
50
|
Moll M, Baumjohann D. Boosting human immunology: harnessing the potential of immune organoids. EMBO Mol Med 2025; 17:385-394. [PMID: 39870882 PMCID: PMC11903751 DOI: 10.1038/s44321-025-00193-8] [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: 06/28/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/29/2025] Open
Abstract
Studying the human immune system in vivo is challenging and often not possible. Therefore, most human immunology studies have been predominantly confined to peripheral blood analyses, which by themselves have inherent limitations, as many immune reactions take place within tissues. For example, potent antibody responses that contribute to fighting infections and provide protection following vaccination require cellular interactions between B cells and T cells in specialized micro-anatomical structures called germinal centers, which are found in secondary lymphoid organs such as spleen, lymph nodes, and tonsils. Thus, there is a clear demand for novel enhanced experimental systems that faithfully recapitulate the intricate dynamics of the human immune system as much as possible. In this review, we discuss recent advances in versatile human tonsil/adenoid tissue-based ex vivo immune organoid cultures as well as related cancer and autoimmunity-focused experimental setups. These systems have been implemented as translational immunology platforms for in-depth analyses of human B and T cell-mediated immune responses, thereby facilitating mechanistic studies as well as drug and vaccine testing in a human-first approach.
Collapse
Affiliation(s)
- Maximilian Moll
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Dirk Baumjohann
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|