1
|
Cong R, Lu C, Li X, Xu Z, Wang Y, Sun S. Tumor organoids in cancer medicine: from model systems to natural compound screening. PHARMACEUTICAL BIOLOGY 2025; 63:89-109. [PMID: 39893515 PMCID: PMC11789228 DOI: 10.1080/13880209.2025.2458149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
CONTEXT The advent of tissue engineering and biomedical techniques has significantly advanced the development of three-dimensional (3D) cell culture systems, particularly tumor organoids. These self-assembled 3D cell clusters closely replicate the histopathological, genetic, and phenotypic characteristics of primary tissues, making them invaluable tools in cancer research and drug screening. OBJECTIVE This review addresses the challenges in developing in vitro models that accurately reflect tumor heterogeneity and explores the application of tumor organoids in cancer research, with a specific focus on the screening of natural products for antitumor therapies. METHODS This review synthesizes information from major databases, including Chemical Abstracts, Medicinal and Aromatic Plants Abstracts, ScienceDirect, Google Scholar, Scopus, PubMed and Springer Link. Publications were selected without date restrictions, using terms such as 'organoid', 'natural product', 'pharmacological', 'extract', 'nanomaterial' and 'traditional uses'. Articles related to agriculture, ecology, synthetic work or published in languages other than English were excluded. RESULTS AND CONCLUSIONS The review identifies key challenges related to the efficiency and variability of organoid generation and discusses ongoing efforts to enhance their predictive capabilities in drug screening and personalized medicine. Recent studies utilizing patient-derived organoid models for natural compound screening are highlighted, demonstrating the potential of these models in developing new classes of anticancer agents. The integration of natural products with patient-derived organoid models presents a promising approach for discovering novel anticancer compounds and elucidating their mechanisms of action.
Collapse
Affiliation(s)
- Rong Cong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can Lu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaqin Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Shusen Sun
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA
| |
Collapse
|
2
|
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] [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
|
3
|
Sun J, Zhao W, Zhang L, Wu S, Xue S, Cao H, Xu B, Li X, Hu N, Jiang T, Xu Y, Wang Z, Zhang C, Ren J. Centromere protein U mediates the ubiquitination and degradation of RPS3 to facilitate temozolomide resistance in glioblastoma. Drug Resist Updat 2025; 80:101214. [PMID: 40023134 DOI: 10.1016/j.drup.2025.101214] [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: 12/08/2024] [Revised: 02/10/2025] [Accepted: 02/15/2025] [Indexed: 03/04/2025]
Abstract
AIMS Temozolomide (TMZ) is the first-line chemotherapeutic agent for glioblastoma (GBM) therapy; however, resistance to TMZ remains a major obstacle in GBM treatment. The aim of this study is to elucidate the mechanisms underlying TMZ resistance and explore how to enhance the sensitivity of GBM to TMZ. METHODS GBM organoids were generated from patient samples, and organoid-based TMZ sensitivity testing was performed. Transcriptome sequencing was conducted on GBM organoids, which identified Centromere protein U (CENPU) as a novel key gene mediating TMZ resistance. Histopathological assessments were carried out using immunohistochemistry (IHC) and Hematoxylin and Eosin (HE) staining. Single-cell sequencing data were utilized to determine the functional states of CENPU in GBM cells. Intracranial and subcutaneous glioma mouse models were constructed to evaluate the effect of CENPU on TMZ sensitivity. The underlying mechanisms were further investigated using immunofluorescence, lentivirus transduction, co-immunoprecipitation, mass spectrometry, alkaline comet assay et al. RESULTS: CENPU was found to be highly expressed in TMZ-resistant GBM organoids and enhanced the TMZ resistance of GBM cells by promoting DNA damage repair. Its abnormal expression correlates with poor clinical outcomes in glioma patients. In vivo studies demonstrated that downregulation of CENPU enhances the sensitivity of GBM to TMZ. Correspondingly, rescue of CENPU expression reversed this effect on TMZ sensitivity in GBM cells. Mechanistically, CENPU cooperates with TRIM5α to promote the ubiquitination and degradation of RPS3 by inducing its polyubiquitination at the K214 residue. This process subsequently activates the ERK1/2 pathway and promotes the expression of E2F1 and RAD51. Consequently, the degradation of RPS3 and upregulation of RAD51 in GBM cells enhance DNA damage repair, thereby contributing to TMZ resistance. CONCLUSION Our study identified CENPU as a novel key gene mediating TMZ resistance and elucidated its molecular mechanisms, providing a new target to overcome TMZ resistance in GBM.
Collapse
Affiliation(s)
- Jinmin Sun
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China; Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Wenyu Zhao
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Lei Zhang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Sicheng Wu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Senrui Xue
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Haowei Cao
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Biao Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xinmiao Li
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Nan Hu
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Zhifei Wang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Chao Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong, Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Jing Ren
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
4
|
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
|
5
|
Wang Y, Sun X, Lu B, Zhang D, Yin Y, Liu S, Chen L, Zhang Z. Current applications, future Perspectives and challenges of Organoid technology in oral cancer research. Eur J Pharmacol 2025; 993:177368. [PMID: 39947346 DOI: 10.1016/j.ejphar.2025.177368] [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: 11/14/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Oral cancer poses significant health risks with an increasing incidence annually. Despite advancements in treatment methods, their efficacy is frequently constrained by cancer heterogeneity and drug resistance, leading to minimal improvement in the 5-year survival rate. Therefore, there is a critical need for new treatment methods leaded by representative preclinical research models. Compared to other models, organoids can more precisely simulate the tissue structure, genetic characteristics, and tumor microenvironment (TME) of in vivo tumors, exhibiting high tumor specificity. This makes organoid technology a valuable tool in investigating tumor development, mechanisms of metastasis, drug screening, prediction of clinical responses, and personalized patient treatment. Moreover, integrating organoid technology with other biotechnologies could expand its applications in tissue regeneration. Although organoid technology is increasingly utilized in oral cancer research, a systematic review in this field is absent. This paper is to bridge the gap by reviewing the development and current status of organoid research, highlighting its applications, future prospects, and challenges in oral cancer. It aims to provide novel insights into the role of organoids in precision treatment and regenerative medicine for oral cancer.
Collapse
Affiliation(s)
- Yunyi Wang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiang Sun
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bingxu Lu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Danya Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yaping Yin
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Shuguang Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Lei Chen
- Department of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Zhaoqiang Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Holowatyj AN, Overman MJ, Votanopoulos KI, Lowy AM, Wagner P, Washington MK, Eng C, Foo WC, Goldberg RM, Hosseini M, Idrees K, Johnson DB, Shergill A, Ward E, Zachos NC, Shelton D. Defining a 'cells to society' research framework for appendiceal tumours. Nat Rev Cancer 2025; 25:293-315. [PMID: 39979656 DOI: 10.1038/s41568-024-00788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/22/2025]
Abstract
Tumours of the appendix - a vestigial digestive organ attached to the colon - are rare. Although we estimate that around 3,000 new appendiceal cancer cases are diagnosed annually in the USA, the challenges of accurately diagnosing and identifying this tumour type suggest that this number may underestimate true population incidence. In the current absence of disease-specific screening and diagnostic imaging modalities, or well-established risk factors, the incidental discovery of appendix tumours is often prompted by acute presentations mimicking appendicitis or when the tumour has already spread into the abdominal cavity - wherein the potential misclassification of appendiceal tumours as malignancies of the colon and ovaries also increases. Notwithstanding these diagnostic difficulties, our understanding of appendix carcinogenesis has advanced in recent years. However, there persist considerable challenges to accelerating the pace of research discoveries towards the path to improved treatments and cures for patients with this group of orphan malignancies. The premise of this Expert Recommendation article is to discuss the current state of the field, to delineate unique challenges for the study of appendiceal tumours, and to propose key priority research areas that will deliver a more complete picture of appendix carcinogenesis and metastasis. The Appendix Cancer Pseudomyxoma Peritonei (ACPMP) Research Foundation Scientific Think Tank delivered a consensus of core research priorities for appendiceal tumours that are poised to be ground-breaking and transformative for scientific discovery and innovation. On the basis of these six research areas, here, we define the first 'cells to society' research framework for appendix tumours.
Collapse
Affiliation(s)
- Andreana N Holowatyj
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Patrick Wagner
- Division of Surgical Oncology, Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Mary K Washington
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cathy Eng
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Wai Chin Foo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Mojgan Hosseini
- Department of Pathology, University of California, San Diego, San Diego, CA, USA
| | - Kamran Idrees
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ardaman Shergill
- Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Erin Ward
- Section of Surgical Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicholas C Zachos
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Deborah Shelton
- Appendix Cancer Pseudomyxoma Peritonei (ACPMP) Research Foundation, Springfield, PA, USA
| |
Collapse
|
7
|
Chaudhary N, La Ferlita A, Choudhary BS, Jog E, Kazi M, Yahya S, Dalwai A, Ostwal V, Singh S, Redkar S, Khapare N, Kailaje V, B A, Gera P, Bal M, Verma N, Thorat R, Saklani A, Sehgal L, Dalal SN. Patient-Derived Organoids and Xenografts Uncover Therapeutic Vulnerabilities in Colorectal Signet Ring Cell Carcinomas. Clin Cancer Res 2025; 31:1359-1373. [PMID: 39879477 DOI: 10.1158/1078-0432.ccr-24-2329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/28/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
PURPOSE Identifying therapeutic targets for signet ring cell carcinoma (SRCC) of the colon and rectum is a clinical challenge because of the lack of patient-derived organoids (PDO) or patient-derived xenografts (PDX). To address this unmet need, we present a robust method for establishing PDO and PDX models. We demonstrate that these models identify novel therapeutic strategies targeting therapy resistance and peritoneal metastasis. EXPERIMENTAL DESIGN We derived nine PDO and PDX models from patients with colorectal SRCC. Detailed histopathologic characterization confirmed the fidelity of these models to the original tumors. Drug sensitivity assays were conducted in vitro and in vivo to assess the therapeutic efficacy and impact on peritoneal metastasis. An RNA sequencing analysis was performed to identify critical pathways contributing to therapy resistance and metastatic progression. RESULTS We successfully developed and characterized PDO and PDX models from nine patients with SRCC. The SRCC PDO and PDX models exhibited histopathologic features consistent with those of the original tumors, including high mucin content and eccentric nuclei. They demonstrated increased sensitivity to FOLFIRI combined with paclitaxel or vincristine, reducing peritoneal metastasis. RNA sequencing analysis revealed the upregulation of autophagy genes in SRCC. Treatment with chloroquine alone resulted in decreased tumor growth and peritoneal metastasis. CONCLUSIONS Our study establishes PDO and PDX models as robust platforms for studying SRCC and identifying potential therapeutic strategies. Combining FOLFIRI with paclitaxel/vincristine or chloroquine alone inhibits tumor growth and prevents peritoneal metastasis, showing promise for clinical translation. These findings suggest that combining FOLFIRI with intraperitoneal paclitaxel warrants further investigation in phase I clinical trials for patients with SRCC.
Collapse
Affiliation(s)
- Nazia Chaudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Alessandro La Ferlita
- Division of Hematology, College of Medicine, The Ohio State University, Columbus, Ohio
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio
| | - Bhagya Shree Choudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Eeshrita Jog
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Mufaddal Kazi
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
- Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Showket Yahya
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Afiya Dalwai
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Vikas Ostwal
- Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Satishkumar Singh
- Division of Hematology, College of Medicine, The Ohio State University, Columbus, Ohio
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio
| | - Siddhi Redkar
- Electron Microscopy Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Nileema Khapare
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Vaishali Kailaje
- Digital Imaging Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Akshaya B
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Poonam Gera
- Department of Biorepository, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Munita Bal
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Nandini Verma
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
- TNBC Precision Medicine Research Group, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Avanish Saklani
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
- Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
- Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, India
| | - Lalit Sehgal
- Division of Hematology, College of Medicine, The Ohio State University, Columbus, Ohio
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| |
Collapse
|
8
|
Chen P, Zhou JB, Chu XP, Feng YY, Zeng QB, Lei JH, Wong KP, Chan TI, Lam CW, Zhu WL, Chu WK, Hu F, Luo GH, Chan KI, Deng CX. Establishing a cryopreserved biobank of living tumor tissues for drug sensitivity testing. Bioact Mater 2025; 46:582-596. [PMID: 40061435 PMCID: PMC11889390 DOI: 10.1016/j.bioactmat.2024.09.008] [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: 01/17/2024] [Revised: 07/05/2024] [Accepted: 09/04/2024] [Indexed: 03/17/2025] Open
Abstract
The cryopreservation of cancer tissues to generate frozen libraries is a common practice used worldwide for storing patient samples for later applications. However, frozen samples stored by existing methods cannot be used for initiating living cell cultures, such as patient-derived tumor organoids (PDOs), which offer great potential for personalized treatment. To overcome this challenge, we developed a novel procedure for culturing PDOs using frozen live tumor tissues. We show that tumor specimens stored using this technique maintain their viability and can be successfully used to generate organoids even after long-term freezing, with an impressive success rate of 95.2 %. Importantly, we found that the structural features, tumor marker expression, and drug responses of organoids derived from frozen tissues are similar to those derived from fresh tissues. Moreover, organoids derived from frozen tissues can be routinely passaged and frozen, making them ideal for high-throughput drug screening at any time. Notably, cryopreserved tumor tissues can also be utilized in air-liquid interface (ALI) culture. This method allows for preserving the original tumor microenvironment, making it an invaluable resource for conducting tests on antitumor drug responses, including immune checkpoint inhibitors (ICIs). This innovation has the potential to enable the identification of potentially effective drugs for patients and facilitate the development of novel therapeutic drugs. Thus, we have established protocols for the long-term cryopreservation of cancer tissues to maintain their viability and microenvironment, which are useful for personalized therapy.
Collapse
Affiliation(s)
- Ping Chen
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing-Bo Zhou
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Xiang-Peng Chu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Yang-Yang Feng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Qi-Bing Zeng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Josh-Haipeng Lei
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Ka-Pou Wong
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | | | | | - Wen-Li Zhu
- Kiang Wu Hospital, Macau SAR 999078, China
| | | | - Feng Hu
- Kiang Wu Hospital, Macau SAR 999078, China
| | | | | | - Chu-Xia Deng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| |
Collapse
|
9
|
Lee J, Kim Y, Lee C, Jeon SS, Seo H, Lee J, Choi J, Kang M, Kim E, Shin K. Generation of prostate cancer assembloids modeling the patient-specific tumor microenvironment. PLoS Genet 2025; 21:e1011652. [PMID: 40163511 DOI: 10.1371/journal.pgen.1011652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Prostate cancer (PC) is the most frequently diagnosed malignancy among men and contributes significantly to cancer-related mortality. While recent advances in in vitro PC modeling systems have been made, there remains a lack of robust preclinical models that faithfully recapitulate the genetic and phenotypic characteristics across various PC subtypes-from localized PC (LPC) to castration-resistant PC (CRPC)-along with associated stromal cells. Here, we established human PC assembloids from LPC and CRPC tissue by reconstituting tumor organoids with corresponding cancer-associated fibroblasts (CAFs), thereby incorporating aspects of the tumor microenvironment (TME). Established PC organoids exhibited high concordance in genomic landscape with parental tumors, and the tumor assembloids showed a higher degree of phenotypic similarity to parental tumors compared to tumor organoids without CAFs. PC assembloids displayed increased proliferation and reduced sensitivity to anti-cancer treatments, indicating that PC assembloids are potent tools for understanding PC biology, investigating interaction between tumor and CAFs, and identifying personalized therapeutic targets.
Collapse
Affiliation(s)
- Juhee Lee
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Yunhee Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cheol Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seong Soo Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hae Seo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jongwon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Eunjee Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Kunyoo Shin
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Jia H, Chen X, Zhang L, Chen M. Cancer associated fibroblasts in cancer development and therapy. J Hematol Oncol 2025; 18:36. [PMID: 40156055 PMCID: PMC11954198 DOI: 10.1186/s13045-025-01688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key players in cancer development and therapy, and they exhibit multifaceted roles in the tumor microenvironment (TME). From their diverse cellular origins, CAFs undergo phenotypic and functional transformation upon interacting with tumor cells and their presence can adversely influence treatment outcomes and the severity of the cancer. Emerging evidence from single-cell RNA sequencing (scRNA-seq) studies have highlighted the heterogeneity and plasticity of CAFs, with subtypes identifiable through distinct gene expression profiles and functional properties. CAFs influence cancer development through multiple mechanisms, including regulation of extracellular matrix (ECM) remodeling, direct promotion of tumor growth through provision of metabolic support, promoting epithelial-mesenchymal transition (EMT) to enhance cancer invasiveness and growth, as well as stimulating cancer stem cell properties within the tumor. Moreover, CAFs can induce an immunosuppressive TME and contribute to therapeutic resistance. In this review, we summarize the fundamental knowledge and recent advances regarding CAFs, focusing on their sophisticated roles in cancer development and potential as therapeutic targets. We discuss various strategies to target CAFs, including ECM modulation, direct elimination, interruption of CAF-TME crosstalk, and CAF normalization, as approaches to developing more effective treatments. An improved understanding of the complex interplay between CAFs and TME is crucial for developing new and effective targeted therapies for cancer.
Collapse
Affiliation(s)
- Hongyuan Jia
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Xingmin Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Linling Zhang
- Department of Respiratory and Critical Care, Chengdu Third People's Hospital, Chengdu, China
| | - Meihua Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
11
|
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 2025:102039. [PMID: 40154491 DOI: 10.1016/j.xcrm.2025.102039] [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: 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
|
12
|
Yang D, Zhang X, Hu Z, Sun Q, Fu H, Yao J, Zheng B, Zhang X, Wang W. Organoid-based single cell sequencing revealed the lineage evolution during docetaxel treatment in gastric cancer. Cancer Lett 2025; 619:217617. [PMID: 40118243 DOI: 10.1016/j.canlet.2025.217617] [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: 01/31/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/23/2025]
Abstract
Docetaxel resistance in gastric cancer poses a major therapeutic challenge. In this study, we established docetaxel-sensitive and -resistant gastric cancer organoids and performed single-cell RNA sequencing to identify cellular and molecular alterations. We observed significant shifts in cell populations, with increased secretory, immune-chemotactic, and transitional gastric cancer cells in the resistant group. Key resistance-related genes, including FOS, IFI27, and PTTG1IP, were upregulated in resistant organoids and gastric cancer patients. A pseudo-time trajectory analysis revealed that resistant cells predominantly occupied terminal differentiation stages. Knocking down FOS, IFI27, and PTTG1IP enhanced docetaxel sensitivity in both cell lines and organoids, regulating ROS production, autophagy, and apoptosis. In vivo, silencing these genes reduced tumor growth in response to docetaxel. These findings suggest that targeting FOS, IFI27, and PTTG1IP could overcome resistance and improve treatment outcomes for gastric cancer patients.
Collapse
Affiliation(s)
- Dejun Yang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China.
| | - Xin Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital (Changhai Hospital) of Naval Medical University, Yangpu District, No. 168 Changhai Road, Shanghai, 200433, China
| | - Zunqi Hu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Qiang Sun
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Hongbing Fu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Jun Yao
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Binbin Zheng
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Xin Zhang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China.
| | - Weijun Wang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital (Changzheng Hospital) of Naval Medical University, Huangpu District, No. 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
13
|
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] [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
|
14
|
Chen J, Liu L, Yang Y, Luo J, Liu S. Patient-derived organoid models of malignant phyllodes tumours for drug sensitivity testing and identification of targeted therapeutic strategies. J Drug Target 2025:1-11. [PMID: 40059613 DOI: 10.1080/1061186x.2025.2473010] [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: 10/18/2024] [Revised: 01/16/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Malignant phyllodes tumours (MPT) of the breast are rare fibroepithelial neoplasms. It exhibits rapid growth, large size, and a high local recurrence rate. METHODS In this study, we established novel patient-derived organoid (PDO) models from two primary MPT samples and conducted comprehensive genetic profiling and drug screening. RESULTS The PDO models faithfully recapped the histopathological and molecular features of the primary tumours, including stromal overgrowth, leaf-like projections, and the expression of key diagnostic markers. Drug testing revealed significant heterogeneity in response profiles to chemotherapeutic reagents between the two MPT-derived organoids, implying the importance of personalised drug testing. Next-generation sequencing analysis identified recurrent mutations in TP53, RB1, EGFR, ATM, and RECQL4, which correlated with the drug sensitivity profiles observed in the organoid models. Targeted therapeutic drugs, such as Abemaciclib (targeting the RB1 pathway) with an IC50 value of 1.744 µM, and Alflutinib Mesylate (targeting the EGFR pathway) with an IC50 value of 0.9150 µM, exhibited significant cytotoxic effects in the MPT2 organoid models. CONCLUSIONS This study highlights the novel application of PDOs for studying the molecular landscape of MPTs and identifying effective therapeutic targets, offering a promising platform for guiding personalised treatment strategies for this rare and challenging cancer.
Collapse
Affiliation(s)
- Jie Chen
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liangquan Liu
- Department of Breast Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunxu Yang
- Chengdu OrganoidMed Medical Laboratory, West China Health Valley, Chengdu, China
| | - Jing Luo
- Department of Breast Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengchun Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Telang NT. Natural Bioactive Agents: Testable Stem Cell-Targeting Alternatives for Therapy-Resistant Breast Cancer. Int J Mol Sci 2025; 26:2529. [PMID: 40141171 PMCID: PMC11942498 DOI: 10.3390/ijms26062529] [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: 01/20/2025] [Revised: 02/17/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Long-term treatment options for conventional chemo-endocrine therapy and molecular-pathway-based targeted therapy are associated with acquired therapy resistance and the emergence of drug-resistant cancer-initiating stem cell populations, leading to the progression of metastatic disease. These treatment options are based on the expression status of estrogen receptor-α (ER-α), progesterone receptor (PR) hormone receptors, and/or of human epidermal growth factor receptor-2 (HER-2). The breast cancer subtypes Luminal A, Luminal B, and HER-2-enriched express hormone/growth factor receptors and exhibit a favorable response to hormone receptor modulators and growth factor receptor antagonists. The triple-negative breast cancer subtype lacks the expression of hormone/growth factor receptors and responds only to cytotoxic conventional chemotherapy. The clinical limitations, due to the modest therapeutic responses of chemo-resistant cancer-initiating stem cells, emphasize the need for the identification of stem cells targeting testable alternatives for therapy-resistant breast cancer. Developed drug-resistant stem cell models exhibit upregulated expression of select cellular biomarker tumor spheroid (TS) formations and cluster of differentiation44 (CD44), DNA-binding protein (NANOG), and octamer-binding protein-4 (OCT-4) molecular biomarkers that represent novel experimentally modifiable quantitative endpoints. Naturally occurring dietary phytochemicals and nutritional herbs containing polyphenols, flavones, terpenes, saponins, lignans, and tannins have documented human consumption, lack systemic toxicity, lack phenotypic drug resistance, and exhibit preclinical efficacy. Constituent bioactive agents may provide testable stem cell-targeting alternatives. The present report provides an overview of (i) clinically relevant cellular models and drug-resistant cancer stem cell models for breast cancer subtypes, (ii) evidence for preclinical efficacy and mechanistic leads for natural phytochemicals and nutritional herbs, and (iii) the potential for the stem cell-targeting efficacy of natural bioactive agents as testable drug candidates for therapy-resistant breast cancer.
Collapse
Affiliation(s)
- Nitin T Telang
- Cancer Prevention Research Program, Palindrome Liaisons Consultants, Montvale, NJ 07645-1559, USA
| |
Collapse
|
16
|
Issing C, Menche C, Richter MR, Mosa MH, von der Grün J, Fleischmann M, Thoenissen P, Winkelmann R, Darvishi T, Loth AG, Ghanaati S, Rödel F, Wild PJ, Brandts CH, Stöver T, Farin HF. Head and neck tumor organoid biobank for modelling individual responses to radiation therapy according to the TP53/HPV status. J Exp Clin Cancer Res 2025; 44:85. [PMID: 40045309 PMCID: PMC11881459 DOI: 10.1186/s13046-025-03345-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/22/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Head and neck cancers (HNC) represent an extremely heterogeneous group of diseases with a poorly predictable therapy outcome. Patient-derived tumor organoids (PDTO) offer enormous potential for individualized therapy testing and a better mechanistic understanding of the main HNC drivers. METHODS Here, we have established a comprehensive molecularly and functionally characterized head and neck organoid biobank (HNOB) recapitulating the clinically relevant subtypes of TP53 mutant and human papillomavirus type 16 (HPV 16) infection-driven HNC. Organoids were exposed to radiotherapy, and responses were correlated with clinical data. Genetically engineered normal and tumor organoids were used for testing the direct functional consequences of TP53-loss and HPV infection. RESULTS The HNOB consisting of 18 organoid models, including 15 tumor models, was generated. We identified subtype-associated transcriptomic signatures and pathological features, including sensitivity to TP53 stabilization by the MDM2 inhibitor Nutlin-3. Furthermore, we describe an in vitro radio response assay revealing phenotypic heterogeneity linked to the individual patient's treatment outcome, including relapse probability. Using genetically engineered organoids, the possibility of co-existence of both cancer drivers was confirmed. TP53 loss, as well as HPV, increased growth in normal and tumor organoids. TP53 loss-of-function alone was insufficient to promote radiation resistance, whereas HPV 16 oncogenes E6/E7 mediated radiosensitivity via induction of cell cycle arrest. CONCLUSION Our results highlight the translational value of the head and neck organoid models not only for patient stratification but also for mechanistic validation of therapy responsiveness of specific cancer drivers.
Collapse
Affiliation(s)
- Christian Issing
- Department of Otorhinolaryngology, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany.
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany.
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany.
- University Cancer Center (UCT) Frankfurt, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany.
- Mildred-Scheel Early Career Center Frankfurt, Frankfurt/Main, Germany.
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Constantin Menche
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mara Romero Richter
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Mohammed H Mosa
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Jens von der Grün
- Mildred-Scheel Early Career Center Frankfurt, Frankfurt/Main, Germany
- Department of Radio-oncology, University Hospital Zürich, Zürich, Switzerland
| | - Maximilian Fleischmann
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Philipp Thoenissen
- Clinic of Oral, Cranio-Maxillofacial and Plastic Facial Surgery, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Ria Winkelmann
- Dr. Senckenberg Institute for Pathology and Human Genetics, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Tahmineh Darvishi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Andreas G Loth
- Department of Otorhinolaryngology, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Shahram Ghanaati
- Clinic of Oral, Cranio-Maxillofacial and Plastic Facial Surgery, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Franz Rödel
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Peter J Wild
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- University Cancer Center (UCT) Frankfurt, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
- Dr. Senckenberg Institute for Pathology and Human Genetics, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Christian H Brandts
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- University Cancer Center (UCT) Frankfurt, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Timo Stöver
- Department of Otorhinolaryngology, Goethe University Frankfurt, University Hospital, Frankfurt/Main, Germany
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany.
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany.
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
17
|
Portik D, Lacombe D, Faivre-Finn C, Achard V, Andratschke N, Correia D, Spalek M, Guckenberger M, Ost P, Ehret F. The 2024 State of Science report from the European Organisation for Research and Treatment of Cancer's Radiation Oncology Scientific Council. Eur J Cancer 2025; 220:115334. [PMID: 40127505 DOI: 10.1016/j.ejca.2025.115334] [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: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Radiotherapy (RT) is a central pillar of a multimodal cancer treatment approach. The ongoing advances in the fields of RT, imaging technologies, cancer biology, and others yield the potential to refine the use of RT. The European Organisation for Research and Treatment of Cancer (EORTC) hosted a dedicated workshop to identify and prioritize key research questions and to define future RT-based treatment strategies to improve the survival and quality of life of cancer patients. METHODS An initial call for relevant RT research topics led to the formation of workgroups to develop these into new clinical research proposals and projects. The EORTC Radiation Oncology Scientific Council (ROSC) State of Science workshop was held in Brussels, Belgium, in February 2024, bringing together EORTC members and international stakeholders to connect and work on the proposals. RESULTS Four topics of interest were identified: I) De-escalation of RT, minimizing toxicity while maintaining patients' quality of life, II) Technology-driven RT utilizing advances in treatment techniques, such as spatially fractionated RT to improve outcomes in patients with bulky disease and localized high tumor burden, III) Biology-driven RT, integrating the rapid advances in cancer biology and functional imaging to guide and personalize RT, and IV) New indications adding value and expanding the use of RT. CONCLUSION The EORTC ROSC State of Science workshop prioritized clinical questions to be addressed in prospective clinical research projects to advance RT care and improve patient outcomes.
Collapse
Affiliation(s)
- Daniel Portik
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium; Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, the Netherlands.
| | - Denis Lacombe
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium
| | - Corinne Faivre-Finn
- Department of Clinical Oncology, The Christie Hospital NHS Foundation Trust, University of Manchester, Manchester, United Kingdom
| | - Vérane Achard
- Department of Radiotherapy, Institut Bergonié, Bordeaux, France and University of Geneva, Geneva, Switzerland
| | - Nicolaus Andratschke
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Dora Correia
- Department of Radiation Oncology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Mateusz Spalek
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Piet Ost
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Iridium Network, Radiation Oncology, Wilrijk, Belgium
| | - Felix Ehret
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
18
|
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
|
19
|
Giron-Michel J, Padelli M, Oberlin E, Guenou H, Duclos-Vallée JC. State-of-the-Art Liver Cancer Organoids: Modeling Cancer Stem Cell Heterogeneity for Personalized Treatment. BioDrugs 2025; 39:237-260. [PMID: 39826071 PMCID: PMC11906529 DOI: 10.1007/s40259-024-00702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/20/2025]
Abstract
Liver cancer poses a global health challenge with limited therapeutic options. Notably, the limited success of current therapies in patients with primary liver cancers (PLCs) may be attributed to the high heterogeneity of both hepatocellular carcinoma (HCCs) and intrahepatic cholangiocarcinoma (iCCAs). This heterogeneity evolves over time as tumor-initiating stem cells, or cancer stem cells (CSCs), undergo (epi)genetic alterations or encounter microenvironmental changes within the tumor microenvironment. These modifications enable CSCs to exhibit plasticity, differentiating into various resistant tumor cell types. Addressing this challenge requires urgent efforts to develop personalized treatments guided by biomarkers, with a specific focus on targeting CSCs. The lack of effective precision treatments for PLCs is partly due to the scarcity of ex vivo preclinical models that accurately capture the complexity of CSC-related tumors and can predict therapeutic responses. Fortunately, recent advancements in the establishment of patient-derived liver cancer cell lines and organoids have opened new avenues for precision medicine research. Notably, patient-derived organoid (PDO) cultures have demonstrated self-assembly and self-renewal capabilities, retaining essential characteristics of their respective in vivo tissues, including both inter- and intratumoral heterogeneities. The emergence of PDOs derived from PLCs serves as patient avatars, enabling preclinical investigations for patient stratification, screening of anticancer drugs, efficacy testing, and thereby advancing the field of precision medicine. This review offers a comprehensive summary of the advancements in constructing PLC-derived PDO models. Emphasis is placed on the role of CSCs, which not only contribute significantly to the establishment of PDO cultures but also faithfully capture tumor heterogeneity and the ensuing development of therapy resistance. The exploration of PDOs' benefits in personalized medicine research is undertaken, including a discussion of their limitations, particularly in terms of culture conditions, reproducibility, and scalability.
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France.
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France.
| | - Maël Padelli
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- Department of Biochemistry and Oncogenetics, Paul Brousse Hospital, AP-HP, Villejuif, France
| | - Estelle Oberlin
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Hind Guenou
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Jean-Charles Duclos-Vallée
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- INSERM UMR-S 1193, Paul Brousse Hospital, Villejuif, France
- Hepato-Biliary Department, Paul Brousse Hospital, APHP, Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hepatinov, Villejuif, France
| |
Collapse
|
20
|
Kim J, Kim R, Lee W, Kim GH, Jeon S, Lee YJ, Lee JS, Kim KH, Won J, Lee W, Park K, Kim HJ, Im S, Lee KJ, Park C, Kim J, Lee JY. Assembly of glioblastoma tumoroids and cerebral organoids: a 3D in vitro model for tumor cell invasion. Mol Oncol 2025; 19:698-715. [PMID: 39473365 PMCID: PMC11887666 DOI: 10.1002/1878-0261.13740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/02/2024] [Accepted: 09/13/2024] [Indexed: 03/08/2025] Open
Abstract
Glioblastoma (GBM) has a fatal prognosis because of its aggressive and invasive characteristics. Understanding the mechanism of invasion necessitates an elucidation of the relationship between tumor cells and the tumor microenvironment. However, there has been a scarcity of suitable models to investigate this. In this study, we established a glioblastoma-cerebral organoid assembloid (GCOA) model by co-culturing patient-derived GBM tumoroids and human cerebral organoids. Tumor cells from the tumoroids infiltrated the cerebral organoids, mimicking the invasive nature of the parental tumors. Using time-lapse imaging, various invasion patterns of cancer cells within cerebral organoids resembling a normal tissue milieu were monitored. Both single- and collective-cell invasion was captured in real-time. We also confirmed the formation of an intercellular tumor network and tumor-normal-cell interactions. Furthermore, the transcriptomic characterization of GCOAs revealed distinct features of invasive tumor cells. Overall, this study established the GCOA as a three-dimensional (3D) in vitro assembloid model to investigate invasion mechanisms and interactions between tumor cells and their microenvironment.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
| | - Rokhyun Kim
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
| | - Wonseok Lee
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
- Department of Transitional MedicineSeoul National University College of MedicineSeoulKorea
- Department of Neurosurgery, Seoul National University HospitalSeoul National University College of MedicineSeoulKorea
| | - Gyu Hyun Kim
- Laboratory of Synaptic Circuit Plasticity, Neural Circuits Research GroupKorea Brain Research InstituteDaeguKorea
| | - Seeun Jeon
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
| | - Yun Jin Lee
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
| | - Jong Seok Lee
- Division of Pediatric NeurosurgerySeoul National University Children's HospitalSeoulKorea
| | - Kyung Hyun Kim
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
- Division of Pediatric NeurosurgerySeoul National University Children's HospitalSeoulKorea
| | - Jae‐Kyung Won
- Department of Pathology, Seoul National University HospitalSeoul National University College of MedicineSeoulKorea
| | - Woochan Lee
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
| | - Kyunghyuk Park
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
| | - Hyun Je Kim
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
- Cancer Research Institute, Medical Research CenterSeoul National University College of MedicineSeoulKorea
| | - Sun‐Wha Im
- Department of Biochemistry and Molecular BiologyKangwon National University School of MedicineChuncheonKorea
| | - Kea Joo Lee
- Laboratory of Synaptic Circuit Plasticity, Neural Circuits Research GroupKorea Brain Research InstituteDaeguKorea
| | - Chul‐Kee Park
- Department of Neurosurgery, Seoul National University HospitalSeoul National University College of MedicineSeoulKorea
| | - Jong‐Il Kim
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
- Cancer Research Institute, Medical Research CenterSeoul National University College of MedicineSeoulKorea
- Department of Biochemistry and Molecular BiologySeoul National University College of MedicineSeoulKorea
| | - Ji Yeoun Lee
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
- Division of Pediatric NeurosurgerySeoul National University Children's HospitalSeoulKorea
- Neuroscience Research Institute, Medical Research CenterSeoul National University College of MedicineSeoulKorea
| |
Collapse
|
21
|
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
|
22
|
Ji L, Chen J, He L, Zhang F, Deng Z, Lin J, Qi Z, Luo X, Giuliano AE, Cui X, Lin SL, Cui Y. Reversal of endocrine resistance via N6AMT1-NEDD4L pathway-mediated p110α degradation. Oncogene 2025; 44:530-544. [PMID: 39623076 PMCID: PMC11832415 DOI: 10.1038/s41388-024-03238-3] [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: 07/14/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 02/19/2025]
Abstract
Approximately 70% of breast cancer (BC) cases are luminal-type (estrogen receptor-positive, ER+), suitable for endocrine therapy with tamoxifen as the most commonly used drug. However, about 30% of these patients develop tamoxifen resistance due to various mechanisms, primarily involving PI3K pathway activation through mutations or unknown pathways. Here, we discover, via bioinformatics analysis and clinical samples, that N6 adenine-specific DNA methyltransferase 1 (N6AMT1) is highly expressed in luminal breast cancer but downregulated in tamoxifen-resistant (TamR) BC cells. ChIP-qPCR and luciferase reporter assays showed that FOXA1 binds to the N6AMT1 promoter and enhances its transcription. In TamR models, FOXA1 and N6AMT1 are downregulated, increasing p110α protein levels (but not mRNA), phospho-AKT levels, and tamoxifen resistance. In vivo, N6AMT1 overexpression enhanced tamoxifen sensitivity, while knockdown reduced it; this sensitivity could be restored with the p110α inhibitor A66. Clinically, decreased N6AMT1 expression correlates with poor prognosis in luminal BC patients. In TamR BC organoids, combining tamoxifen with A66 further reduced growth compared to either treatment alone. Mechanistically, increased p110α levels result from inhibited degradation by E3 ubiquitin ligase NEDD4L. These findings suggest N6AMT1 as a potential luminal breast cancer biomarker and highlight the N6AMT1-p110α pathway as a therapeutic target to sensitize cells to tamoxifen.
Collapse
Affiliation(s)
- Likeng Ji
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiongyu Chen
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lifang He
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Fan Zhang
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zihao Deng
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiediao Lin
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhaochang Qi
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xi Luo
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Armando E Giuliano
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stanley Li Lin
- Department of Cell Biology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yukun Cui
- Shantou Key Laboratory of Precision Diagnosis and Treatment in Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
23
|
Li T, Bo RQ, Yan J, Johnson NL, Liao MT, Li Y, Chen Y, Lin J, Li J, Chu FH, Ding X. Global landscape of hepatic organoid research: A bibliometric and visual study. World J Hepatol 2025; 17:95624. [PMID: 40027550 PMCID: PMC11866153 DOI: 10.4254/wjh.v17.i2.95624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/11/2024] [Accepted: 11/12/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Hepatic organoid-based modelling, through the elucidation of a range of in vivo biological processes and the recreation of the intricate liver microenvironment, is yielding groundbreaking insights into the pathophysiology and personalized medicine approaches for liver diseases. AIM This study was designed to analyse the global scientific output of hepatic organoid research and assess current achievements and future trends through bibliometric analysis. METHODS Articles were retrieved from the Web of Science Core Collection, and CiteSpace 6.3.R1 was employed to analyse the literature, including outputs, journals, and countries, among others. RESULTS Between 2010 and 2024, a total of 991 articles pertaining to hepatic organoid research were published. The journal Hepatology published the greatest number of papers, and journals with an impact factor greater than 10 constituted 60% of the top 10 journals. The United States and Utrecht University were identified as the most prolific country and institution, respectively. Clevers H emerged as the most prolific author, whereas Huch M had the highest number of cocitations, suggesting that both are ideal candidates for academic collaboration. Research on hepatic organoids has exhibited a progressive shift in focus, evolving from initial investigations into model building, differentiation research in stem cells, bile ducts, and progenitor cells, to a broader spectrum encompassing lipid metabolism, single-cell RNA sequencing, and therapeutic applications. The phrases exhibiting citation bursts from 2022 to 2024 include "drug resistance", "disease model", and "patient-derived tumor organoids". CONCLUSION Research on hepatic organoids has increased over the past decade and is expected to continue to grow. Key research areas include applications for liver diseases and drug development. Future trends likely to gain focus include patient-derived tumour organoids, disease modelling, and personalized medicine.
Collapse
Affiliation(s)
- Tao Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Rong-Qiang Bo
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jun Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Nadia L Johnson
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Meng-Ting Liao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yuan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yan Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jie Lin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jian Li
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fu-Hao Chu
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
24
|
Dornhof J, Kieninger J, Rupitsch SJ, Weltin A. Microsensor systems for cell metabolism - from 2D culture to organ-on-chip (2019-2024). LAB ON A CHIP 2025; 25:1149-1168. [PMID: 39775787 DOI: 10.1039/d4lc00437j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Cell cultures, organs-on-chip and microphysiological systems become increasingly relevant as in vitro models, e.g., in drug development, disease modelling, toxicology or cancer research. It has been underlined repeatedly that culture conditions and metabolic cues have a strong or even essential influence on the reproducibility and validity of such experiments but are often not appropriately measured or controlled. Here we review microsensor systems for cell metabolism for the continuous measurement of culture conditions in microfluidic and lab-on-chip platforms. We identify building blocks, features and essential advantages to underline the relevance of these systems and to derive appropriate requirements for development and practical use. We discuss different formats and geometries of cell culture, microfluidics and the resulting consequences for sensor placement, as the prerequisite for understanding the various approaches and classification of the systems. The major chemical and biosensors based on electrochemical and optical principles are discussed for general understanding and to contextualize current developments. We then review selected recent sensor systems with real-world implementations of sensing in cell cultures and organs-on-chip, employing a helpful characterization. That includes formats and cell models, microfluidic systems and sensor types applied in static and dynamic monitoring of 2D and 3D cell cultures, as well as single spheroids. We discuss notable advances, particularly with respect to sensor performance and the demonstration of long-term continuous measurements. We outline current approaches to system fabrication technologies, material choice, and interfacing, and comment on recent trends. Finally, we conclude with critical remarks on the current state of sensors in cell culture monitoring and identify avenues for future improvements for both developers and users of such systems, which will lead to better and more predictive in vitro models.
Collapse
Affiliation(s)
- Johannes Dornhof
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Jochen Kieninger
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Stefan J Rupitsch
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| | - Andreas Weltin
- Laboratory for Electrical Instrumentation and Embedded Systems, IMTEK - Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110 Freiburg, Germany.
| |
Collapse
|
25
|
Hamada A, Kita Y, Sakatani T, Nakamura K, Takada H, Ikeuchi R, Koike S, Masuda N, Murakami K, Sano T, Goto T, Saito R, Teramoto Y, Fujimoto M, Hatano N, Kamada M, Ogawa O, Kobayashi T. PTEN loss drives p53 LOH and immune evasion in a novel urothelial organoid model harboring p53 missense mutations. Oncogene 2025:10.1038/s41388-025-03311-5. [PMID: 39987272 DOI: 10.1038/s41388-025-03311-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/15/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
Despite missense mutation accounts for over 60% of p53 alterations while homozygous deletion (HOM) for only 5% or less in advanced bladder cancer cases, most of the previously reported mouse models are deficient of p53. Accordingly, few studies have addressed the mechanisms of missense mutation occurrence and its functional advantage over HOM in bladder cancer development. Organoids derived from Krt5-expressing mouse urothelium (K5-mUrorganoid) demonstrated the crucial role of Pten loss in driving loss of wild-type allele of Trp53 (Trp53R172H/LOH), which conferred tumorigenic ability to K5-mUrorganoid in athymic mice. These tumors recapitulated the histological and genetic characteristics of the human basal-squamous subtype bladder cancer. Both Trp53R172H/Δ; PtenΔ/Δ and Trp53Δ/Δ; PtenΔ/Δ K5-mUrorganoids formed tumors in athymic mice, whereas only Trp53R172H/Δ; PtenΔ/Δ K5-mUrorganoid formed tumors even when directly inoculated in immunocompetent syngeneic mice. The absence of wild-type Trp53 was associated with upregulation of proliferative signaling, and the presence of a mutant Trp53 allele was associated with immune-excluded microenvironment. This study highlights the functional significance of p53 mutant LOH in bladder carcinogenesis conferring several hallmarks of cancer such as sustaining proliferative signaling and avoiding immune destruction, thus provides a novel immunocompetent mouse model of urothelial carcinoma harboring p53 mutations as a novel tool for cancer immunology research.
Collapse
Affiliation(s)
- Akihiro Hamada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Kita
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toru Sakatani
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Nakamura
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Takada
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Ikeuchi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuhei Koike
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norihiko Masuda
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Kaoru Murakami
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Sano
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology and Andrology, Kansai Medical University, Osaka, Japan
| | - Takayuki Goto
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryoichi Saito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Teramoto
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masakazu Fujimoto
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Narumi Hatano
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mayumi Kamada
- Department of Biomedical Data Intelligence, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology, Otsu Red Cross Hospital, Shiga, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
26
|
Nagashima Y, Yamamoto H, Elbadawy M, Ishihara Y, Tsurukami I, Abugomaa A, Kaneda M, Yamawaki H, Usui T, Sasaki K. Establishment of an experimental model of canine apocrine gland anal sac adenocarcinoma organoid culture using a three-dimensional culture method. Sci Rep 2025; 15:6108. [PMID: 39972078 PMCID: PMC11840023 DOI: 10.1038/s41598-025-90623-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 02/14/2025] [Indexed: 02/21/2025] Open
Abstract
Canine apocrine gland anal sac adenocarcinoma (AGASACA) is a rare, malignant tumor in dogs. To date, few cell lines are available and used to establish the current treatment protocols. Organoids are three-dimensional cell cultures derived mainly from stem cells and can reproduce tissue's epithelial structure, function, and genetics, and thus, of great promise in precision medicine. In the current investigation, 10 AGASACA organoid lines were developed from surgically removed tissues of AGASACA-affected dogs and analyzed for comparison with the original tissues. AGASACA organoids were successfully generated from all cases and were positive for CK7, HER2, p53, p63, VEGF, and Ki67, and negative for CK20, consistent with previous reports in dogs and humans. Electron microscopic imaging of AGASACA organoids showed organelles, including numerous granules and fat droplets that characterize apocrine gland cells. AGASACA organoids were tumorigenic in vivo in immunodeficient mice. In addition, treatment of the AGASACA organoids with carboplatin, mitoxantrone, toceranib, and lapatinib revealed different sensitivity profiles among lineages, with carboplatin and lapatinib, in particular, being divided into sensitive and resistant ones. In contrast, mitoxantrone and toceranib showed generally high efficacy in all organoids. In conclusion, our established AGASACA organoids have the potential to be an experimental tool for the development of novel therapies for canine and human apocrine gland adenocarcinoma.
Collapse
Affiliation(s)
- Yuko Nagashima
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan
| | - Haru Yamamoto
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan
- AIRDEC Mini CO., LTD, 1-2-36 Kajino-Cho, Koganei, Tokyo, 184-0002, Japan
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan.
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, 13736, Elqaliobiya, Egypt.
- Department of Pathology, College of Veterinary Medicine, Precision One Health Initiative, University of Georgia, Athens, GA, 30602, USA.
| | - Yusuke Ishihara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan
| | - Issei Tsurukami
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan
- Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, 35-1, Higashi 23 Ban-Cho, Towada, Aomori, 034-8628, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan.
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu, Tokyo, 183-8509, Japan
| |
Collapse
|
27
|
Huang L, Liao C, Xiong Z, Chen Z, Zhang S. Hsa-miR-526b-5p Regulates the Sensitivity of Colorectal Cancer to 5-Fluorouracil by Targeting TP53 in Organoid Models. Biochem Genet 2025:10.1007/s10528-025-11045-y. [PMID: 39953363 DOI: 10.1007/s10528-025-11045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
This study aimed to explore the mechanisms through which microRNAs (miRNAs) regulate 5-fluorouracil (5-FU) sensitivity in colorectal cancer (CRC) using organoid models. Fresh tissue samples from CRC tumors were collected, and CRC organoids were isolated and cultured. The consistency between CRC organoids and their derived tissues was validated. CRC organoids were treated with 5-FU, and ATP activity was measured. High-throughput sequencing of CRC organoids, combined with Gene Expression Omnibus (GEO) data analysis, was performed to examine miRNA expression following 5-FU treatment. Next, we investigated the cellular function of miR-526b-5p in CRC organoids and cells. Dual-luciferase reporter assays validated the binding of miR-526b-5p to the 3' UTR of TP53 mRNA. We successfully established CRC organoids that exhibited characteristics consistent with their source tissues. 5-FU treatment suppressed the proliferation and ATP activity of CRC organoids. High-throughput sequencing of CRC organoids, combined with GEO data analysis and quantitative reverse transcription polymerase chain reaction (qRT-PCR) validation, revealed that hsa-miR-526b-5p levels were elevated following 5-FU treatment in CRC organoids and cells. Furthermore, hsa-miR-526b-5p was upregulated in CRC tissues compared to adjacent normal tissues, correlating with poor survival in CRC patients. Overexpression of hsa-miR-526b-5p mitigated the inhibitory effects of 5-FU on CRC organoid proliferation, migration, invasion, and ferroptosis. In contrast, silencing of hsa-miR-526b-5p impaired cell function and ferroptosis. Additionally, overexpression of hsa-miR-526b-5p decreased TP53 mRNA and protein levels while increasing the expression of SLC7A11 mRNA and protein. Silencing of hsa-miR-526b-5p resulted in the opposite effect. hsa-miR-526b-5p directly targeted and inhibited TP53 expression. Overexpression of TP53 diminished the promotive effect of hsa-miR-526b-5p on ferroptosis-related proteins GPX4 and SLC7A11, whereas inhibition of TP53 reversed the impact of hsa-miR-526b-5p silencing. Our study demonstrates that hsa-miR-526b-5p targets TP53 to regulate 5-FU sensitivity in CRC through the ferroptosis pathway based on CRC organoid models.
Collapse
Affiliation(s)
- Lizhe Huang
- Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Cun Liao
- Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Zuming Xiong
- Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Zhongyang Chen
- Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China
| | - Sen Zhang
- Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, China.
| |
Collapse
|
28
|
Mallya D, Gadre MA, Varadharajan S, Vasanthan KS. 3D bioprinting for the construction of drug testing models-development strategies and regulatory concerns. Front Bioeng Biotechnol 2025; 13:1457872. [PMID: 40028291 PMCID: PMC11868281 DOI: 10.3389/fbioe.2025.1457872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
A drug to be successfully launched in the market requires a significant amount of capital, resources and time, where the unsuccessful results in the last stages lead to catastrophic failure for discovering drugs. This is the very reason which calls for the invention of innovative models that can closely mimic the human in vivo model for producing reliable results. Throughout the innovation line, there has been improvement in the rationale in silico designing but yet there is requirement for in vitro-in vivo correlations. During the evolving of the drug testing models, the 3D models produced by different methods have been proven to produce better results than the traditional 2D models. However, the in vitro fabrications of live tissues are still bottleneck in realizing their complete potential. There is an urgent need for the development of single, standard and simplified in vitro 3D tissue models that can be reliable for investigating the biological and pathological aspects of drug discovery, which is yet to be achieved. The existing pre-clinical models have considerable drawbacks despite being the gold standard in pre-clinical research. The major drawback being the interspecies differences and low reliability on the generated results. This gap could be overcome by the fabrication of bioengineered human disease models for drug screening. The advancement in the fabrication of 3D models will provide a valuable tool in screening drugs at different stages as they are one step closer to bio-mimic human tissues. In this review, we have discussed on the evolution of preclinical studies, and different models, including mini tissues, spheroids, organoids, bioengineered three dimensional models and organs on chips. Furthermore, we provide details of different disease models fabricated across various organs and their applications. In addition to this, the review also focuses on the limitations and the current prospects of the role of three dimensionally bioprinted models in drug screening and development.
Collapse
Affiliation(s)
- Divya Mallya
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mrunmayi Ashish Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - S. Varadharajan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kirthanashri S. Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
29
|
Tao W, Sun Q, Xu B, Wang R. Towards the Prediction of Responses to Cancer Immunotherapy: A Multi-Omics Review. Life (Basel) 2025; 15:283. [PMID: 40003691 PMCID: PMC11856636 DOI: 10.3390/life15020283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor treatment has undergone revolutionary changes with the development of immunotherapy, especially immune checkpoint inhibitors. Because not all patients respond positively to immune therapeutic agents, and severe immune-related adverse events (irAEs) are frequently observed, the development of the biomarkers evaluating the response of a patient is key for the application of immunotherapy in a wider range. Recently, various multi-omics features measured by high-throughput technologies, such as tumor mutation burden (TMB), gene expression profiles, and DNA methylation profiles, have been proved to be sensitive and accurate predictors of the response to immunotherapy. A large number of predictive models based on these features, utilizing traditional machine learning or deep learning frameworks, have also been proposed. In this review, we aim to cover recent advances in predicting tumor immunotherapy response using multi-omics features. These include new measurements, research cohorts, data sources, and predictive models. Key findings emphasize the importance of TMB, neoantigens, MSI, and mutational signatures in predicting ICI responses. The integration of bulk and single-cell RNA sequencing has enhanced our understanding of the tumor immune microenvironment and enabled the identification of predictive biomarkers like PD-L1 and IFN-γ signatures. Public datasets and machine learning models have also improved predictive tools. However, challenges remain, such as the need for large and diverse clinical datasets, standardization of multi-omics data, and model interpretability. Future research will require collaboration among researchers, clinicians, and data scientists to address these issues and enhance cancer immunotherapy precision.
Collapse
Affiliation(s)
- Weichu Tao
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
| | - Qian Sun
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
| | - Bingxiang Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (W.T.); (Q.S.)
| |
Collapse
|
30
|
Zou RQ, Dai YS, Liu F, Yang SQ, Hu HJ, Li FY. Hepatobiliary organoid research: the progress and applications. Front Pharmacol 2025; 16:1473863. [PMID: 40008122 PMCID: PMC11850396 DOI: 10.3389/fphar.2025.1473863] [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: 07/31/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Organoid culture has emerged as a forefront technology in the life sciences field. As "in vitro micro-organs", organoids can faithfully recapitulate the organogenesis process, and conserve the key structure, physiological function and pathological state of the original tissue or organ. Consequently, it is widely used in basic and clinical studies, becoming important preclinical models for studying diseases and developing therapies. Here, we introduced the definition and advantages of organoids and described the development and advances in hepatobiliary organoids research. We focus on applying hepatobiliary organoids in benign and malignant diseases of the liver and biliary tract, drug research, and regenerative medicine to provide valuable reference information for the application of hepatobiliary organoids. Despite advances in research and treatment, hepatobiliary diseases including carcinoma, viral hepatitis, fatty liver and bile duct defects have still been conundrums of the hepatobiliary field. It is necessary and crucial to study disease mechanisms, establish efficient and accurate research models and find effective treatment strategies. The organoid culture technology shed new light on solving these issues. However, the technology is not yet mature, and many hurdles still exist that need to be overcome. The combination with new technologies such as CRISPR-HOT, organ-on-a-chip may inject new vitality into future development.
Collapse
Affiliation(s)
- Rui-Qi Zou
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Shi Dai
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fei Liu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Si-Qi Yang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai-Jie Hu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fu-Yu Li
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
31
|
de Moraes EC, Siqueira Furtuoso Rodrigues MM, de Menezes RC, Vinícius-Araújo M, Valadares MC, Bakuzis AF. Human 3D Lung Cancer Tissue Photothermal Therapy Using Zn- and Co-Doped Magnetite Nanoparticles. ACS Biomater Sci Eng 2025; 11:1084-1095. [PMID: 39853243 DOI: 10.1021/acsbiomaterials.4c01901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Iron oxide-based nanoparticles are promising materials for cancer thermal therapy and immunotherapy. However, several proofs of concept reported data with murine tumor models that might have limitations for clinical translation. Magnetite is nowadays the most popular nanomaterial, but doping with distinct ions can enhance thermal therapy, namely, magnetic nanoparticle hyperthermia (MNH) and photothermal therapy (PTT). In this study, we used a 3D alveolar reconstructed A549 lung cancer tissue model and investigated the thermal properties, toxicity, and impact of the thermal dose on tissue viability and inflammatory response using magnetite codoped with 40% Zn and 2% Co divalent ions. The ZnCo-doped magnetite nanoparticles are not toxic up to an NP concentration of 30 mg/mL. PTT showed a better heat generation response than MNH under the evaluated conditions, while NP showed a high external photothermal conversion efficiency of ∼1.3 g·L-1·cm-1 at 808 nm. PTT study is carried out at different temperatures, 43 and 47 °C, for 15 min. Tissue viability decreased with increasing thermal dose, while intracelullar ROS levels increased, mitochondrial activity decreased, and active caspase-3 increased, suggesting cell death via apoptosis. Nanoparticles and PTT did not influence the cytokine TNF, IL-10, IL-1B, and IL-12p70. In contrast, IL-6 and IL-8 were triggered by NP and PTT. Increased expression of IL-6 and IL-8 with higher thermal doses is correlated with tissue injury results, suggesting the potential role in activating and attracting immune cells to the site of thermal-mediated tissue injury.
Collapse
Affiliation(s)
| | | | - Rafaela Campos de Menezes
- ToxIn-Laboratory of Education and Research in In Vitro Toxicology, Federal University of Goiás, Goiânia 74690-631, Brazil
| | | | - Marize Campos Valadares
- ToxIn-Laboratory of Education and Research in In Vitro Toxicology, Federal University of Goiás, Goiânia 74690-631, Brazil
| | - Andris Figueiroa Bakuzis
- Institute of Physics, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil
- CNanoMed, Federal University of Goiás, Goiânia, Goiás 74690-631, Brazil
| |
Collapse
|
32
|
Steindl A, Valiente M. Potential of ex vivo organotypic slice cultures in neuro-oncology. Neuro Oncol 2025; 27:338-351. [PMID: 39504579 PMCID: PMC11812025 DOI: 10.1093/neuonc/noae195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Over recent decades, in vitro and in vivo models have significantly advanced brain cancer research; however, each presents distinct challenges for accurately mimicking in situ conditions. In response, organotypic slice cultures have emerged as a promising model recapitulating precisely specific in vivo phenotypes through an ex vivo approach. Ex vivo organotypic brain slice models can integrate biological relevance and patient-specific variability early in drug discovery, thereby aiming for more precise treatment stratification. However, the challenges of obtaining representative fresh brain tissue, ensuring reproducibility, and maintaining essential central nervous system (CNS)-specific conditions reflecting the in situ situation over time have limited the direct application of ex vivo organotypic slice cultures in robust clinical trials. In this review, we explore the benefits and possible limitations of ex vivo organotypic brain slice cultures in neuro-oncological research. Additionally, we share insights from clinical experts in neuro-oncology on how to overcome these current limitations and improve the practical application of organotypic brain slice cultures beyond academic research.
Collapse
Affiliation(s)
- Ariane Steindl
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
33
|
Logun M, Wang X, Sun Y, Bagley SJ, Li N, Desai A, Zhang DY, Nasrallah MP, Pai ELL, Oner BS, Plesa G, Siegel D, Binder ZA, Ming GL, Song H, O'Rourke DM. Patient-derived glioblastoma organoids as real-time avatars for assessing responses to clinical CAR-T cell therapy. Cell Stem Cell 2025; 32:181-190.e4. [PMID: 39657679 PMCID: PMC11808387 DOI: 10.1016/j.stem.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Patient-derived tumor organoids have been leveraged for disease modeling and preclinical studies but rarely applied in real time to aid with interpretation of patient treatment responses in clinics. We recently demonstrated early efficacy signals in a first-in-human, phase 1 study of dual-targeting chimeric antigen receptor (CAR)-T cells (EGFR-IL13Rα2 CAR-T cells) in patients with recurrent glioblastoma. Here, we analyzed six sets of patient-derived glioblastoma organoids (GBOs) treated concurrently with the same autologous CAR-T cell products as patients in our phase 1 study. We found that CAR-T cell treatment led to target antigen reduction and cytolysis of tumor cells in GBOs, the degree of which correlated with CAR-T cell engraftment detected in patients' cerebrospinal fluid (CSF). Furthermore, cytokine release patterns in GBOs mirrored those in patient CSF samples over time. Our findings highlight a unique trial design and GBOs as a valuable platform for real-time assessment of CAR-T cell bioactivity and insights into immunotherapy efficacy.
Collapse
Affiliation(s)
- Meghan Logun
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Xin Wang
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yusha Sun
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephen J Bagley
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nannan Li
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arati Desai
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Y Zhang
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - MacLean P Nasrallah
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Emily Ling-Lin Pai
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bike Su Oner
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Donald Siegel
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zev A Binder
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Hongjun Song
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Donald M O'Rourke
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Sun Y, Liu J, Zhu L, Huang F, Dong Y, Liu S, Chen S, Ji W, Lu J, Liu L, Li S. Treatment Response to Oncolytic Virus in Patient-Derived Breast Cancer and Hypopharyngeal Cancer Organoids: Evaluation via a Microfluidics Organ-on-a-Chip System. Bioengineering (Basel) 2025; 12:146. [PMID: 40001666 PMCID: PMC11851931 DOI: 10.3390/bioengineering12020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
In this study, we present an oncolytic virus (OV) evaluation system established using microfluidic organ-on-a-chip (OOC) systems and patient-derived organoids (PDOs), which was used in the development of a novel oncolytic virus, AD4-GHPE. An OV offers advantages such as good targeting ability and minimal side effects, and it has achieved significant breakthroughs when combined with immunotherapy in recent clinical trials. The development of OVs has become an emerging research focus. PDOs can preserve the heterogeneity of in situ tumor tissues, whereas microfluidic OOC systems can automate and standardize various experimental procedures. These systems have been applied in cutting-edge drug screening and cell therapy experiments; however, their use in functionally complex oncolytic viruses remains to be explored. In this study, we constructed a novel recombinant oncolytic adenovirus, AD4-GHPE, and evaluated OOC systems and PDOs through various functional validations in hypopharyngeal and breast cancer organoids. The results confirmed that AD4-GHPE exhibits three antitumor mechanisms, namely, tumor-specific cytotoxicity, a reduction in programmed death ligand 1 (PD-L1) expression in tumor cells to increase CD8+ T-cell activity, and granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion. The evaluation system combining OOC systems and PDOs was efficient and reliable, providing personalized OV treatment recommendations for patients and offering industrialized and standardized research ideas for the development of OVs.
Collapse
Affiliation(s)
- Yu Sun
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jiaqi Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Li Zhu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Yanbo Dong
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shuang Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Siyi Chen
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wei Ji
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Jingjing Lu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| | - Liangfa Liu
- Department of Otolaryngology and Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.S.); (Y.D.)
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100071, China; (J.L.)
| |
Collapse
|
35
|
Zheng M, Qu J, Xiang D, Xing L. Organoids in lung cancer brain metastasis: Foundational research, clinical translation, and prospective outlooks. Biochim Biophys Acta Rev Cancer 2025; 1880:189235. [PMID: 39647672 DOI: 10.1016/j.bbcan.2024.189235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 12/01/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Brain metastasis stands as a leading contributor to mortality in lung cancer patients, yet the intricate mechanism underlying this phenomenon remains elusive. This underscores the need for robust preclinical models and effective treatment strategies. Emerging as viable in vitro models that closely replicate actual tumors, three-dimensional culture systems, particularly organoids derived from non-malignant cells or cancer organoids, have emerged as promising avenues. This review delves into the forefronts of fundamental research and clinical applications focused on lung cancer brain metastasis-derived organoids, highlighting current challenges and delineating prospects. These studies offer tremendous potential for clinical application despite being in nascent status.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jialin Qu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Dongxi Xiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Department of Biliary-Pancreatic Surgery, the Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| |
Collapse
|
36
|
Kalla J, Pfneissl J, Mair T, Tran L, Egger G. A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. Cell Oncol (Dordr) 2025; 48:1-26. [PMID: 38806997 PMCID: PMC11850459 DOI: 10.1007/s13402-024-00960-8] [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] [Accepted: 05/11/2024] [Indexed: 05/30/2024] Open
Abstract
Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organoids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced culturing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are currently used for precision medicine.
Collapse
Affiliation(s)
- Jessica Kalla
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
37
|
Chauhdari T, Zaidi SA, Su J, Ding Y. Organoids meet microfluidics: recent advancements, challenges, and future of organoids-on-chip. IN VITRO MODELS 2025; 4:71-88. [PMID: 40160209 PMCID: PMC11950471 DOI: 10.1007/s44164-025-00086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
Organoids are three-dimensional, miniaturized tissue-like structures derived from either stem cells or primary cells, emerging as powerful in vitro models for studying developmental biology, disease pathology, and drug discovery. These organoids more accurately mimic cell-cell interactions and complexities of human tissues compared to traditional cell cultures. However, challenges such as limited nutrient supply and biomechanical cue replication hinder their maturation and viability. Microfluidic technologies, with their ability to control fluid flow and mimic the mechanical environment of tissues, have been integrated with organoids to create organoid-on-chip models that address these limitations. These models not only improve the physiological relevance of organoids but also enable more precise investigation of disease mechanisms and therapeutic responses. By combining microfluidics and organoids, several advanced organoids-on-chip models have been developed to investigate mechanical and biochemical cues involved in disease progression. This review discusses various methods to develop organoids-on-chip and the recently established organoids-on-chip models with their advanced functions. Finally, we highlighted potential strategies to enhance the functionality of organoid models, aiming to overcome current limitations and bridge the gap between current cell culture models and clinical applications, advancing personalized medicine, and improving therapeutic testing.
Collapse
Affiliation(s)
- Talha Chauhdari
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| | - Syeda Armana Zaidi
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| | - Jilei Su
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| | - Yongsheng Ding
- College of Life Sciences, University of Chinese Academy of Sciences, No. 1 Yanqihu East Rd, Huairou District, 101408 Beijing PR China
| |
Collapse
|
38
|
Zhang W, Wu C, Huang H, Bleu P, Zambare W, Alvarez J, Wang L, Paty PB, Romesser PB, Smith JJ, Chen XS. Enhancing chemotherapy response prediction via matched colorectal tumor-organoid gene expression analysis and network-based biomarker selection. Transl Oncol 2025; 52:102238. [PMID: 39754813 PMCID: PMC11754497 DOI: 10.1016/j.tranon.2024.102238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) presents significant challenges in chemotherapy response prediction due to its molecular heterogeneity. Current methods often fail to account for the complexity and variability inherent in individual tumors. METHODS We developed a novel approach using matched CRC tumor and organoid gene expression data. We applied Consensus Weighted Gene Co-expression Network Analysis (WGCNA) across three datasets: CRC tumors, matched organoids, and an independent organoid dataset with IC50 drug response values, to identify key gene modules and hub genes linked to chemotherapy response, particularly 5-fluorouracil (5-FU). FINDINGS Our integrative analysis identified significant gene modules and hub genes associated with CRC chemotherapy response. The predictive model built from these findings demonstrated superior accuracy over traditional methods when tested on independent datasets. The matched tumor-organoid data approach proved effective in capturing relevant biomarkers, enhancing prediction reliability. INTERPRETATION This study provides a robust framework for improving CRC chemotherapy response predictions by leveraging matched tumor and organoid gene expression data. Our approach addresses the limitations of previous methods, offering a promising strategy for personalized treatment planning in CRC. Future research should aim to validate these findings and explore the integration of more comprehensive drug response data. FUNDING This research was supported by US National Cancer Institute grant R37CA248289, and Sylvester Comprehensive Cancer Center. which receives funding from the National Cancer Institute award P30CA240139. This work was supported by National Institutes of Health (NIH) under the following grants: T32CA009501-31A1 and R37CA248289. This work was also supported by the MSK P30CA008748 grant.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chao Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hanchen Huang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Paulina Bleu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wini Zambare
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Janet Alvarez
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lily Wang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Philip B Paty
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - X Steven Chen
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
39
|
Zhao Z, Wu X, Zhang T, Zhou M, Liu S, Yang R, Li JP. Evaluation of Multispecific Drugs Based on Patient-Derived Immunocompetent Tumor Organoids. Chembiochem 2025; 26:e202400731. [PMID: 39800663 DOI: 10.1002/cbic.202400731] [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: 09/05/2024] [Revised: 12/12/2024] [Indexed: 01/24/2025]
Abstract
The evolution of antitumor drug development has transitioned from single-agent chemotherapy to targeted therapy, immunotherapy, and more recently, multispecific drugs. These innovative drugs target multiple cellular or molecular pathways simultaneously, offering a more comprehensive anticancer approach and addressing some of the limitations inherent in traditional monotherapies. However, preclinical assessment of multispecific drugs remains challenging, as conventional tumor models often lack the necessary complexity to accurately reflect the interactions between various cell types and targets. Patient-derived immunocompetent tumor organoids (PDITOs), which incorporate both tumor cells and immune cells, present a promising platform for the evaluation of these drugs. Beyond their use in drug evaluation, PDITOs can also be utilized in personalized drug screening and predicting patient-specific treatment outcomes, thus advancing both multispecific drug development and precision medicine. This perspective discusses the current landscape of multispecific drug development and the methodologies for constructing PDITOs. It also addresses the associated challenges and introduces the concept of employing these organoids to optimize the evaluation and rational design of multispecific drug therapies.
Collapse
Affiliation(s)
- Zihan Zhao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Tianyang Zhang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
- University of Glasgow, University Avenue, Glasgow, G12 8QQ, UK
| | - Meng Zhou
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Siyang Liu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, 210008, China
| | - Jie P Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
40
|
Carrasco-Mantis A, Reina-Romo E, Sanz-Herrera JA. A multiphysics hybrid continuum - agent-based model of in vitro vascularized organoids. Comput Biol Med 2025; 185:109559. [PMID: 39709871 DOI: 10.1016/j.compbiomed.2024.109559] [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: 10/11/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Organoids are 3D in vitro models that fulfill a hierarchical function, representing a small version of living tissues and, therefore, a good approximation of cellular mechanisms. However, one of the main disadvantages of these models is the appearance of a necrotic core due to poor vascularization. The aim of this work is the development of a numerical framework that incorporates the mechanical stimulation as a key factor in organoid vascularization. Parameters, such as fluid velocity and nutrient consumption, are analyzed along the organoid evolution. METHODS The mathematical model created for this purpose combines continuum and discrete approaches. In the continuum part, the fluid flow and the diffusion of oxygen and nutrients are modeled using a finite element method approach. Meanwhile, the growth of the organoid, blood vessel evolution, as well as their interaction with the surrounding environment, are modeled using agent-based methods. RESULTS Continuum model outcomes include the distribution of shear stress, pressure and fluid velocity around the organoid surface, in addition to the concentration of oxygen and nutrients in its interior. The agent models account for cell proliferation, differentiation, organoid growth and blood vessel morphology, for the different case studies considered. CONCLUSIONS Two main conclusions are achieved in this work: (i) the results of the study quantitatively predict in vitro data, with an enhanced blood vessel invasion under high fluid flow and (ii) the diffusion and consumption model parameters of the organoid cells determine the thickness of the proliferative, quiescent, hypoxic and necrotic layers.
Collapse
Affiliation(s)
| | - Esther Reina-Romo
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla, Spain
| | | |
Collapse
|
41
|
Wu D, Huang Q, Xu Y, Cao R, Yang M, Xie J, Zhang D. Clinical efficacy and future application of indigo naturalis in the treatment of ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118782. [PMID: 39236777 DOI: 10.1016/j.jep.2024.118782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/03/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by non-specific inflammation. Managing UC presents significant challenges due to its chronic nature and high recurrence rates. Indigo naturalis has emerged as a potential therapeutic agent in clinical UC treatment, demonstrating advantages in alleviating refractory UC and maintaining remission periods compared to other therapeutic approaches. AIM OF REVIEW This review aims to elucidate the potential mechanisms underlying the therapeutic effects of indigo naturalis in UC treatment, assess its clinical efficacy, advantages, and limitations, and provide insights into methods and strategies for utilizing indigo naturalis in UC management. MATERIALS AND METHODS Comprehensive data on indigo naturalis were collected from reputable online databases including PubMed, GreenMedical, Web of Science, Google Scholar, China National Knowledge Infrastructure Database, and National Intellectual Property Administration. RESULTS Clinical studies have demonstrated that indigo naturalis, either alone or in combination with other drugs, yields favorable outcomes in UC treatment. Its mechanisms of action involve modulation of the AHR receptor, anti-inflammatory properties, regulation of intestinal flora, restoration of the intestinal barrier, and modulation of immunity. Despite its efficacy in managing refractory UC and prolonging remission periods, indigo naturalis treatment is associated with adverse reactions, quality variations, and inadequate pharmacokinetic investigations. CONCLUSION The therapeutic effects of indigo naturalis in UC treatment are closely linked to its ability to regulate the AHR receptor, exert anti-inflammatory effects, mcodulate intestinal flora, restore the intestinal barrier, and regulate immunity. Addressing the current shortcomings, including adverse reactions, quality control issues, and insufficient pharmacokinetic data, is crucial for optimizing the clinical utility of indigo naturalis in UC management. By refining patient-centered treatment strategies, indigo naturalis holds promise for broader application in UC treatment, thereby alleviating the suffering of UC patients.
Collapse
Affiliation(s)
- Dianzhen Wu
- Sichuan Medical Products Administration, Chengdu, 610017, China
| | - Qi Huang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yingbi Xu
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruiyi Cao
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ming Yang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Jin Xie
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Dingkun Zhang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Tianfu TCM Innovation Harbour, Chengdu University of Traditional Chinese Medicine, Chengdu, 611930, China.
| |
Collapse
|
42
|
Song D, Wang X, Zhao Z, Yang R, Zhang S, Guo Z. Targeting Ribosome Biogenesis for Cancer Therapy with Oral Platinum Complexes. JACS AU 2025; 5:73-81. [PMID: 39886599 PMCID: PMC11775699 DOI: 10.1021/jacsau.4c00652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 02/01/2025]
Abstract
Cancer cells often upregulate ribosome biogenesis to meet increased protein synthesis demands for rapid proliferation; therefore, targeting ribosome biogenesis has emerged as a promising cancer therapeutic strategy. Herein, we introduce two Pt complexes, ataluren monosubstituted platinum(IV) (SPA, formula: c,c,t,-[Pt(NH3)2Cl2(OH)(C15H8FN2O3)], where C15H8FN2O3 = ataluren) and ataluren bisubstituted platinum(IV) complex (DPA, formula: c,c,t,-[Pt(NH3)2Cl2(C15H8FN2O3)2], where C15H8FN2O3 = ataluren), which effectively suppress ribosome biogenesis by inhibiting 47s pre-RNA expression. Furthermore, SPA and DPA induce nucleolar stress by dispersing nucleolar protein NPM1, ultimately inhibiting protein generation in tumor cells. More importantly, DPA exhibits superior cytotoxicity to various cancer cells and in vivo antitumor efficacy compared to cisplatin, with lower systemic toxicity. Notably, in clinically relevant models, including orthotopic hepatic tumor-bearing mice and patient-derived bladder cancer organoids, DPA outperforms cisplatin significantly, with the added benefit of oral administration, enhancing clinical feasibility. To our knowledge, DPA emerges as the pioneering Pt(IV) agent targeting the ribosome, providing new insights for designing next-generation metal-based therapeutics.
Collapse
Affiliation(s)
- Dongfan Song
- School
of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation
Center (ChemBIC), State Key Laboratory of
Coordination Chemistry, Najing University, Nanjing 210023, PR China
| | - Xiaoyu Wang
- School
of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation
Center (ChemBIC), State Key Laboratory of
Coordination Chemistry, Najing University, Nanjing 210023, PR China
| | - Zihan Zhao
- Department
of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical
School, Nanjing University, Nanjing 210093, PR China
| | - Rong Yang
- Department
of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical
School, Nanjing University, Nanjing 210093, PR China
| | - Shuren Zhang
- School
of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation
Center (ChemBIC), State Key Laboratory of
Coordination Chemistry, Najing University, Nanjing 210023, PR China
| | - Zijian Guo
- School
of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation
Center (ChemBIC), State Key Laboratory of
Coordination Chemistry, Najing University, Nanjing 210023, PR China
| |
Collapse
|
43
|
Testa M, Gaggianesi M, D’Accardo C, Porcelli G, Turdo A, Di Marco C, Patella B, Di Franco S, Modica C, Di Bella S, Lopresti F, Stassi G, La Carrubba V, Todaro M. A Novel Tumor on Chip Mimicking the Breast Cancer Microenvironment for Dynamic Drug Screening. Int J Mol Sci 2025; 26:1028. [PMID: 39940796 PMCID: PMC11816644 DOI: 10.3390/ijms26031028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
In light of the emerging breakthroughs in cancer biology, drug discovery, and personalized medicine, Tumor-on-Chip (ToC) platforms have become pivotal tools in current biomedical research. This study introduced a novel rapid prototyping approach for the fabrication of a ToC device using laser-patterned poly(methyl methacrylate) (PMMA) layers integrated with a polylactic acid (PLA) electrospun scaffold, enabling dynamic drug delivery and the assessment of therapeutic efficacy in cancer cells. Traditional drug screening methods, such as conventional cell cultures, mimic certain aspects of cancer progression but fail to capture critical features of the tumor microenvironment (TME). While animal models offer a closer approximation of tumor complexity, they are limited in their ability to predict human drug responses. Here, we evaluated the ability of our ToC device to recapitulate the interactions between cancer and TME cells and its efficacy in evaluating the drug response of breast cancer cells. The functional design of the proposed ToC system offered substantial potential for a wide range of applications in cancer research, significantly accelerating the preclinical assessment of new therapeutic agents.
Collapse
Affiliation(s)
- Maria Testa
- Department of Biomedicina, Neuroscienze e Diagnostica avanzata (Bind), University of Palermo, 90127 Palermo, Italy;
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (C.D.M.); (B.P.); (V.L.C.)
| | - Miriam Gaggianesi
- Department of Precision Medicine in Medical, Surgical, and Critical Areas (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy; (M.G.); (C.D.); (G.P.); (S.D.F.); (C.M.); (S.D.B.)
| | - Caterina D’Accardo
- Department of Precision Medicine in Medical, Surgical, and Critical Areas (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy; (M.G.); (C.D.); (G.P.); (S.D.F.); (C.M.); (S.D.B.)
| | - Gaetana Porcelli
- Department of Precision Medicine in Medical, Surgical, and Critical Areas (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy; (M.G.); (C.D.); (G.P.); (S.D.F.); (C.M.); (S.D.B.)
| | - Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.T.); (M.T.)
| | - Chiara Di Marco
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (C.D.M.); (B.P.); (V.L.C.)
| | - Bernardo Patella
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (C.D.M.); (B.P.); (V.L.C.)
| | - Simone Di Franco
- Department of Precision Medicine in Medical, Surgical, and Critical Areas (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy; (M.G.); (C.D.); (G.P.); (S.D.F.); (C.M.); (S.D.B.)
| | - Chiara Modica
- Department of Precision Medicine in Medical, Surgical, and Critical Areas (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy; (M.G.); (C.D.); (G.P.); (S.D.F.); (C.M.); (S.D.B.)
| | - Sebastiano Di Bella
- Department of Precision Medicine in Medical, Surgical, and Critical Areas (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy; (M.G.); (C.D.); (G.P.); (S.D.F.); (C.M.); (S.D.B.)
| | - Francesco Lopresti
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (C.D.M.); (B.P.); (V.L.C.)
| | - Giorgio Stassi
- Department of Precision Medicine in Medical, Surgical, and Critical Areas (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy; (M.G.); (C.D.); (G.P.); (S.D.F.); (C.M.); (S.D.B.)
| | - Vincenzo La Carrubba
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (C.D.M.); (B.P.); (V.L.C.)
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.T.); (M.T.)
| |
Collapse
|
44
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 DOI: 10.3390/cells14030170] [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: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
| | - Daria N Kindt
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
| | - Aleksandra V Sagaidak
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
| | - Daria S Novikova
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
| | - Vyacheslav G Tribulovich
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia
| |
Collapse
|
45
|
Kes MMG, Morales-Rodriguez F, Zaal EA, de Souza T, Proost N, van de Ven M, van den Heuvel-Eibrink MM, Jansen JWA, Berkers CR, Drost J. Metabolic profiling of patient-derived organoids reveals nucleotide synthesis as a metabolic vulnerability in malignant rhabdoid tumors. Cell Rep Med 2025; 6:101878. [PMID: 39708810 DOI: 10.1016/j.xcrm.2024.101878] [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: 11/22/2023] [Revised: 08/27/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
Malignant rhabdoid tumor (MRT) is one of the most aggressive childhood cancers for which no effective treatment options are available. Reprogramming of cellular metabolism is an important hallmark of cancer, with various metabolism-based drugs being approved as a cancer treatment. In this study, we use patient-derived tumor organoids (tumoroids) to map the metabolic landscape of several pediatric cancers. Combining gene expression analyses and metabolite profiling using mass spectrometry, we find nucleotide biosynthesis to be a particular vulnerability of MRT. Treatment of MRT tumoroids with de novo nucleotide synthesis inhibitors methotrexate (MTX) and BAY-2402234 lowers nucleotide levels in MRT tumoroids and induces apoptosis. Lastly, we demonstrate in vivo efficacy of MTX in MRT patient-derived xenograft (PDX) mouse models. Our study reveals nucleotide biosynthesis as an MRT-specific metabolic vulnerability, which can ultimately lead to better treatment options for children suffering from this lethal pediatric malignancy.
Collapse
Affiliation(s)
- Marjolein M G Kes
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Francisco Morales-Rodriguez
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Esther A Zaal
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Terezinha de Souza
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Natalie Proost
- Preclinical Intervention Unit of the Mouse Clinic for Cancer and Ageing (MCCA), Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marieke van de Ven
- Preclinical Intervention Unit of the Mouse Clinic for Cancer and Ageing (MCCA), Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Division of Child Health, Wilhelmina Children's Hospital, Utrecht University, Utrecht, the Netherlands
| | - Jeroen W A Jansen
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Celia R Berkers
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
46
|
Nia HT, Munn LL, Jain RK. Probing the physical hallmarks of cancer. Nat Methods 2025:10.1038/s41592-024-02564-4. [PMID: 39815103 DOI: 10.1038/s41592-024-02564-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/11/2024] [Indexed: 01/18/2025]
Abstract
The physical microenvironment plays a crucial role in tumor development, progression, metastasis and treatment. Recently, we proposed four physical hallmarks of cancer, with distinct origins and consequences, to characterize abnormalities in the physical tumor microenvironment: (1) elevated compressive-tensile solid stresses, (2) elevated interstitial fluid pressure and the resulting interstitial fluid flow, (3) altered material properties (for example, increased tissue stiffness) and (4) altered physical micro-architecture. As this emerging field of physical oncology is being advanced by tumor biologists, cell and developmental biologists, engineers, physicists and oncologists, there is a critical need for model systems and measurement tools to mechanistically probe these physical hallmarks. Here, after briefly defining these physical hallmarks, we discuss the tools and model systems available for probing each hallmark in vitro, ex vivo, in vivo and in clinical settings. We finally review the unmet needs for mechanistic probing of the physical hallmarks of tumors and discuss the challenges and unanswered questions associated with each hallmark.
Collapse
Affiliation(s)
- Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Lance L Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Skarne N, D'Souza RCJ, Palethorpe HM, Bradbrook KA, Gomez GA, Day BW. Personalising glioblastoma medicine: explant organoid applications, challenges and future perspectives. Acta Neuropathol Commun 2025; 13:6. [PMID: 39799339 PMCID: PMC11724554 DOI: 10.1186/s40478-025-01928-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: 09/17/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive adult brain cancer, characterised by poor prognosis and a dismal five-year survival rate. Despite significant knowledge gains in tumour biology, meaningful advances in patient survival remain elusive. The field of neuro-oncology faces many disease obstacles, one being the paucity of faithful models to advance preclinical research and guide personalised medicine approaches. Recent technological developments have permitted the maintenance, expansion and cryopreservation of GBM explant organoid (GBO) tissue. GBOs represent a translational leap forward and are currently the state-of-the-art in 3D in vitro culture system, retaining brain cancer heterogeneity, and transiently maintaining the immune infiltrate and tumour microenvironment (TME). Here, we provide a review of existing brain cancer organoid technologies, in vivo xenograft approaches, evaluate in-detail the key advantages and limitations of this rapidly emerging technology, and consider solutions to overcome these difficulties. GBOs currently hold significant promise, with the potential to emerge as the key translational tool to synergise and enhance next-generation omics efforts and guide personalised medicine approaches for brain cancer patients into the future.
Collapse
Affiliation(s)
- Niclas Skarne
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia.
| | - Rochelle C J D'Souza
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia
| | - Helen M Palethorpe
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5000, Australia
| | - Kylah A Bradbrook
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia
| | - Guillermo A Gomez
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5000, Australia
| | - Bryan W Day
- Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, 4072, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, 4059, Australia.
| |
Collapse
|
48
|
Lederer AK, Görrissen N, Nguyen TT, Kreutz C, Rasel H, Bartsch F, Lang H, Endres K. Exploring the effects of gut microbiota on cholangiocarcinoma progression by patient-derived organoids. J Transl Med 2025; 23:34. [PMID: 39789543 PMCID: PMC11716211 DOI: 10.1186/s12967-024-06012-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: 08/21/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Recent research indicates a role of gut microbiota in development and progression of life-threatening diseases such as cancer. Carcinomas of the biliary ducts, the so-called cholangiocarcinomas, are known for their aggressive tumor biology, implying poor prognosis of affected patients. An impact of the gut microbiota on cholangiocarcinoma development and progression is plausible due to the enterohepatic circulation and is therefore the subject of scientific debate, however evidence is still lacking. This review aimed to discuss the suitability of complex cell culture models to investigate the role of gut microbiota in cholangiocarcinoma progression. MAIN BODY Clinical research in this area is challenging due to poor comparability of patients and feasibility reasons, which is why translational models are needed to understand the basis of tumor progression in cholangiocarcinoma. A promising approach to investigate the influence of gut microbiota could be an organoid model. Organoids are 3D cell models cultivated in a modifiable and controlled condition, which can be grown from tumor tissue. 3D cell models are able to imitate physiological and pathological processes in the human body and thus contribute to a better understanding of health and disease. CONCLUSION The use of complex cell cultures such as organoids and organoid co-cultures might be powerful and valuable tools to study not only the growth behavior and growth of cholangiocarcinoma cells, but also the interaction with the tumor microenvironment and with components of the gut microbiota.
Collapse
Affiliation(s)
- Ann-Kathrin Lederer
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany.
- Center for Complementary Medicine, Department of Medicine II, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, 79106, Freiburg, Germany.
| | - Nele Görrissen
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Tinh Thi Nguyen
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, 55131, Mainz, Germany
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics (IMBI), Faculty of Medicine and Medical Center, 79106, Freiburg, Germany
| | - Hannah Rasel
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Fabian Bartsch
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Hauke Lang
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, 55131, Mainz, Germany
- Faculty of Computer Sciences and Microsystems Technology, University of Applied Sciences Kaiserslautern, 66482, Zweibrücken, Germany
| |
Collapse
|
49
|
Sekeroglu ZA, Sekeroglu V. A Review on Patient-derived 3D Micro Cancer Approach for Drug Screen in Personalized Cancer Medicine. Curr Cancer Drug Targets 2025; 25:118-130. [PMID: 38445692 DOI: 10.2174/0115680096285910240206044830] [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: 11/26/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 03/07/2024]
Abstract
Precision medicine in oncology aims to identify an individualized treatment plan based on genomic alterations in a patient's tumor. It helps to select the most beneficial therapy for an individual patient. As it is now known that no patient's cancer is the same, and therefore, different patients may respond differently to conventional treatments, precision medicine, which replaces the one-size-fits-all approach, supports the development of tailored treatments for specific cancers of different patients. Patient-specific organoid or spheroid models as 3D cell culture models are very promising for predicting resistance to anti-cancer drugs and for identifying the most effective cancer therapy for high-throughput drug screening combined with genomic analysis in personalized medicine. Because tumor spheroids incorporate many features of solid tumors and reflect resistance to drugs and radiation, as in human cancers, they are widely used in drug screening studies. Testing patient-derived 3D cancer spheroids with some anticancer drugs based on information from molecular profiling can reveal the sensitivity of tumor cells to drugs and provide the right compounds to be effective against resistant cells. Given that many patients do not respond to standard treatments, patient-specific treatments will be more effective, less toxic. They will affect survival better compared to the standard approach used for all patients.
Collapse
Affiliation(s)
- Zulal Atlı Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| | - Vedat Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| |
Collapse
|
50
|
Cassani M, Fernandes S, Pagliari S, Cavalieri F, Caruso F, Forte G. Unraveling the Role of the Tumor Extracellular Matrix to Inform Nanoparticle Design for Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409898. [PMID: 39629891 PMCID: PMC11727388 DOI: 10.1002/advs.202409898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Indexed: 01/14/2025]
Abstract
The extracellular matrix (ECM)-and its mechanobiology-regulates key cellular functions that drive tumor growth and development. Accordingly, mechanotherapy is emerging as an effective approach to treat fibrotic diseases such as cancer. Through restoring the ECM to healthy-like conditions, this treatment aims to improve tissue perfusion, facilitating the delivery of chemotherapies. In particular, the manipulation of ECM is gaining interest as a valuable strategy for developing innovative treatments based on nanoparticles (NPs). However, further progress is required; for instance, it is known that the presence of a dense ECM, which hampers the penetration of NPs, primarily impacts the efficacy of nanomedicines. Furthermore, most 2D in vitro studies fail to recapitulate the physiological deposition of matrix components. To address these issues, a comprehensive understanding of the interactions between the ECM and NPs is needed. This review focuses on the main features of the ECM and its complex interplay with NPs. Recent advances in mechanotherapy are discussed and insights are offered into how its combination with nanomedicine can help improve nanomaterials design and advance their clinical translation.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Stefania Pagliari
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|