1
|
Wu Y, Zhang F, Du F, Huang J, Wei S. Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review). Mol Med Rep 2025; 31:140. [PMID: 40183402 PMCID: PMC11976518 DOI: 10.3892/mmr.2025.13505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Malignant tumors notably decrease life expectancy. Despite advances in cancer diagnosis and treatment, the mechanisms underlying tumorigenesis, progression and drug resistance have not been fully elucidated. An emerging method to study tumors is tumor organoids, which are a three‑dimensional miniature structure. These retain the patient‑specific tumor heterogeneity while demonstrating the histological, genetic and molecular features of original tumors. Compared with conventional cancer cell lines and animal models, patient‑derived tumor organoids are more advanced at physiological and clinical levels. Their synergistic combination with other technologies, such as organ‑on‑a‑chip, 3D‑bioprinting, tissue‑engineered cell scaffolds and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR‑associated protein 9, may overcome limitations of the conventional 3D organoid culture and result in the development of more appropriate model systems that preserve the complex tumor stroma, inter‑organ and intra‑organ communications. The present review summarizes the evolution of tumor organoids and their combination with advanced technologies, as well as the application of tumor organoids in basic and clinical research.
Collapse
Affiliation(s)
- Ying Wu
- Department of Obstetrics and Gynecology, The 920th Hospital of Joint Logistics Support Force, Kunming, Yunnan 650032, P.R. China
| | - Fan Zhang
- Department of Comprehensive Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| | - Furong Du
- Department of Medicine, Kingbio Medical Co., Ltd., Chongqing 401123, P.R. China
| | - Juan Huang
- Department of Breast Surgery and Multidisciplinary Breast Cancer Center, Clinical Research Center of Breast Cancer in Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shuqing Wei
- Department of Comprehensive Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| |
Collapse
|
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
|
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] [MESH Headings] [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
|
4
|
Berk Ş. Comprehensive bibliometric analysis and perspectives on therapies targeting colon cancer stem cells over a 40-year period. Regen Ther 2025; 29:19-34. [PMID: 40124468 PMCID: PMC11930536 DOI: 10.1016/j.reth.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
The presence of cancer stem cells (CSCs) is one of the primary causes of recurring therapy resistance because they have two main capacities: self-renewal and avoiding apoptotic pathways. Despite their relevance, no full bibliometric analysis has yet been done in this topic. The goal of this work is to use bibliometric analysis to map the fundamental and emergent areas in therapeutics targeting colon cancer stem cells. To perform bibliometric analysis on colon cancer stem cells (CCSCs) literature, spanning roughly the last 40 years, in order to establish a firm base for future projections by emphasizing the findings of the most notable research. All information pertinent to CCSCs was accessed from Web of Science Core Collection database. In order to identify and analyze the research hotspots and trends related to this topic, Biblioshiny (RStudio) and VOSviewer were utilized to ascertain the countries/regions, institutions, journals, authors, references, and keywords involved. The targeted time span covered 1735 research-, and review articles. The most frequent keywords were "colorectal cancer," "cancer stem cells," and "colon cancer," while the most trending keywords in the last few years were "protein stability," "spheroid formation," "ubiquitination," "exosomes," "patient-derived organoids," and "gut microbiota." Over the past 40 years, there has been a significant advancement in researchers' understanding of colon cancer stem cells. In addition, the cluster map of co-cited literature showed that colon cancer stem cell research has emerged as a research hotspot. It was also anticipated that the main focus of the future efforts appears to involve clinical applications of cell-targeted colon cancer therapy. These results provide researchers with a comprehensive understanding of this field and provide insightful ideas for further research.
Collapse
Affiliation(s)
- Şeyda Berk
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, 58140, Turkey
| |
Collapse
|
5
|
Lu X, Hu C, Duan L, Chen K, Hao H, He Y. Establishment of matched bladder cancer PDX and PDX-derived organoid model and evaluation of anti-tumor efficacy of abemaciclib. Clin Transl Oncol 2025; 27:2207-2219. [PMID: 39436622 DOI: 10.1007/s12094-024-03666-3] [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: 04/16/2024] [Accepted: 08/07/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Bladder cancer is one of the most common malignancies of the urinary system and there's a significant unmet need for new effective therapeutics for bladder cancer. The limited number of available models to study malignant bladder tumors is one of the obstructions in developing bladder cancer therapeutics. Patient-derived xenograft (PDX) and organoid (PDO) models are more representatives of human cancer than cell lines and cell line-derived xenograft (CDX) and are likely to be more promising and efficient in predicting drug response and finding new therapeutics. METHODS Three pairs of patient-derived xenograft (PDX) models of bladder cancer and their corresponding PDX-derived organoids (PDXOs) were successfully established. These models were utilized to assess the efficacy of abemaciclib. The sensitivity of the drug was determined through the Cell Counting Kit-8 (CCK8) assay in PDXO cultures, corroborated by the EdU incorporation assay. Additionally, the in vivo tumor growth was monitored in the matched PDX models. RESULTS In vitro PDXO cultures and in vivo PDX tumor models consistently demonstrated that abemaciclib had varying degrees of inhibitory effects across different bladder cancer (BC) patients. Notably, our study further revealed that treatment with abemaciclib significantly modified the expression patterns of CyclinD1/CDK4. This was achieved by not only diminishing their expression levels but also by shifting their expression from a membrane-associated localization to the nucleus. CONCLUSION Our research provided compelling evidence attesting to the reliability and potential of PDX and PDXO models in the realm of precision medicine. These models are instrumental in identifying patients who are likely to respond favorably to a specific drug treatment.
Collapse
Affiliation(s)
- Xiongbing Lu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Chao Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Lingxing Duan
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Ke Chen
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Hua Hao
- Department of Pathology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| | - Yuanqiao He
- Center of Laboratory Animal Science, Nanchang University, Nanchang, 330031, China.
- Jiangxi Province Key Laboratory of Laboratory Animal, Nanchang, 330031, China.
- Nanchang Royo Biotech Co, Ltd., Nanchang, 330000, China.
| |
Collapse
|
6
|
Calheiros J, Silva R, Barbosa F, Morais J, Moura SR, Almeida S, Fiorini E, Mulhovo S, Aguiar TQ, Wang T, Ricardo S, Almeida MI, Domingues L, Melo SA, Corbo V, Ferreira MJU, Saraiva L. A first-in-class inhibitor of homologous recombination DNA repair counteracts tumour growth, metastasis and therapeutic resistance in pancreatic cancer. J Exp Clin Cancer Res 2025; 44:129. [PMID: 40275348 DOI: 10.1186/s13046-025-03389-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is among the cancer types with poorest prognosis and survival rates primarily due to resistance to standard-of-care therapies, including gemcitabine (GEM) and olaparib. Particularly, wild-type (wt)BRCA tumours, the most prevalent in PDAC, are more resistant to DNA-targeting agents like olaparib, restraining their clinical application. Recently, we disclosed a monoterpene indole alkaloid derivative (BBIT20) as a new inhibitor of homologous recombination (HR) DNA repair with anticancer activity in breast and ovarian cancer. Since inhibition of DNA repair enhances the sensitivity of cancer cells to chemotherapy, we aimed to investigate the anticancer potential of BBIT20 against PDAC, particularly carrying wtBRCA. METHODS In vitro and in vivo PDAC models, particularly human cell lines (including GEM-resistant PDAC cells), patient-derived organoids and xenograft mice of PDAC were used to evaluate the anticancer potential of BBIT20, alone and in combination with GEM or olaparib. Disruption of the BRCA1-BARD1 interaction by BBIT20 was assessed by co-immunoprecipitation, immunofluorescence and yeast two-hybrid assay. RESULTS The potent antiproliferative activity of BBIT20, superior to olaparib, was demonstrated in PDAC cells regardless of BRCA status, by inducing cell cycle arrest, apoptosis, and DNA damage, while downregulating HR. The disruption of DNA double-strand breaks repair by BBIT20 was further reinforced by non-homologous end joining (NHEJ) suppression. The inhibition of BRCA1-BARD1 heterodimer by BBIT20 was demonstrated in PDAC cells and confirmed in a yeast two-hybrid assay. In GEM-resistant PDAC cells, BBIT20 showed potent antiproliferative, anti-migratory and anti-invasive activity, overcoming GEM resistance by inhibiting the multidrug resistance P-glycoprotein, upregulating the intracellular GEM-transporter ENT1, and downregulating GEM resistance-related microRNA-20a and GEM metabolism enzymes as RRM1/2. Furthermore, BBIT20 did not induce resistance in PDAC cells. It inhibited the growth of patient-derived PDAC organoids, by inducing apoptosis, repressing HR, and potentiating olaparib and GEM cytotoxicity. The enhancement of olaparib antitumor activity by BBIT20 was confirmed in xenograft mice of PDAC. Notably, it hindered tumour growth and liver metastasis formation, improving survival of orthotopic xenograft mice of PDAC. Furthermore, its potential as a stroma-targeting agent, reducing fibrotic extracellular matrix and overcoming desmoplasia, associated with an enhancement of immune cell response by depleting PD-L1 expression in tumour tissues, renders BBIT20 even more appealing for combination therapy, particularly with immunotherapy. CONCLUSION These findings underscore the great potential of BBIT20 as a novel multifaceted anticancer drug candidate for PDAC treatment.
Collapse
Grants
- 2020.04613.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2020.06020.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- NHI; HHSN26100008 NCI NIH HHS
Collapse
Affiliation(s)
- Juliana Calheiros
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Rita Silva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Filipa Barbosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal
| | - João Morais
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Sara Reis Moura
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Sofia Almeida
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Elena Fiorini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Silva Mulhovo
- Centro de Estudos Moçambicanos e de Etnociências (CEMEC), Faculty of Natural Sciences and Mathematics, Pedagogical University, Maputo, 21402161, Mozambique
| | - Tatiana Q Aguiar
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Tao Wang
- Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, China
| | - Sara Ricardo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy and UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, 4585-116, Portugal
| | - Maria Inês Almeida
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Lucília Domingues
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Sonia A Melo
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine University of Porto, Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal
- Porto Comprehensive Cancer Centre (P.CCC) Raquel Seruca, Porto, Portugal
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Maria-José U Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal.
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal.
| |
Collapse
|
7
|
DeLiberty JM, Roach MK, Stalnecker CA, Robb R, Schechter EG, Pieper NL, Taylor KE, Pita LM, Yang R, Bang S, Drizyte-Miller K, Ackermann SE, Peña SRN, Baldelli E, Min SM, Drewry DH, Petricoin EF, Morris JP, Der CJ, Cox AD, Bryant KL. Concurrent Inhibition of the RAS-MAPK Pathway and PIKfyve Is a Therapeutic Strategy for Pancreatic Cancer. Cancer Res 2025; 85:1479-1495. [PMID: 39932818 PMCID: PMC11999774 DOI: 10.1158/0008-5472.can-24-1757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/14/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by KRAS- and autophagy-dependent growth. Inhibition of the KRAS-RAF-MEK-ERK pathway enhances autophagic flux and dependency, and concurrent treatment with the nonspecific autophagy inhibitor chloroquine (CQ) and ERK-MAPK pathway inhibitors can synergistically block PDAC growth. However, CQ is limited in terms of specificity and potency. To find alternative anti-autophagy strategies, in this study, we performed a CRISPR-Cas9 loss-of-function screen in PDAC cell lines that identified the lipid kinase phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) as a growth-promoting gene. PIKfyve inhibition by the small molecule apilimod resulted in durable growth suppression, with much greater potency than CQ treatment. PIKfyve inhibition caused lysosomal dysfunction, reduced autophagic flux, and led to the accumulation of autophagy-related proteins. Furthermore, PIKfyve inhibition blocked the compensatory increases in autophagic flux associated both with MEK inhibition and with direct RAS inhibition. Accordingly, combined inhibition of PIKfyve and the RAS-MAPK pathway showed robust growth suppression across a panel of KRAS-mutant PDAC models. Growth suppression was due, in part, to potentiated cell-cycle arrest and induction of apoptosis following loss of inhibitor of apoptosis proteins. These findings indicate that concurrent inhibition of RAS and PIKfyve is a synergistic, cytotoxic combination that may represent a therapeutic strategy for PDAC. Significance: PIKfyve inhibition effectively blocks autophagy in multiple models of KRAS-mutant pancreatic cancer and can synergize with inhibitors of members of the RAS-MAPK pathway, providing an effective combination strategy for pancreatic cancer.
Collapse
Affiliation(s)
| | - Mallory K. Roach
- Department of Pharmacology, George Mason University, Manassas, VA, USA
| | - Clint A. Stalnecker
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Ryan Robb
- Department of Pharmacology, George Mason University, Manassas, VA, USA
| | - Elyse G. Schechter
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Noah L. Pieper
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Khalilah E. Taylor
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Lily M. Pita
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Runying Yang
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Scott Bang
- Department of Pharmacology, George Mason University, Manassas, VA, USA
| | | | | | | | - Elisa Baldelli
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Sophia M. Min
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David H. Drewry
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - John P. Morris
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Channing J. Der
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| | - Adrienne D. Cox
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, NC, USA
| | - Kirsten L. Bryant
- Department of Pharmacology, George Mason University, Manassas, VA, USA
- Lineberger Comprehensive Cancer Center, George Mason University, Manassas, VA, USA
| |
Collapse
|
8
|
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
|
9
|
Lloyd EG, Jihad M, Manansala JS, Li W, Cheng PS, Mucciolo G, Zaccaria M, Teles SP, Henríquez JA, Harish S, Brais R, Ashworth S, Luo W, Johnson PM, Veghini L, Vallespinos M, Corbo V, Biffi G. SMAD4 and KRAS Status Shapes Cancer Cell-Stromal Cross-Talk and Therapeutic Response in Pancreatic Cancer. Cancer Res 2025; 85:1368-1389. [PMID: 39841099 PMCID: PMC7617379 DOI: 10.1158/0008-5472.can-24-2330] [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: 10/21/2024] [Revised: 12/09/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) contains an extensive stroma that modulates response to therapy, contributing to the dismal prognosis associated with this cancer. Evidence suggests that PDAC stromal composition is shaped by mutations within malignant cells, but most previous work has focused on preclinical models driven by KrasG12D and mutant Trp53. Elucidation of the contribution of additional known oncogenic drivers, including KrasG12V mutation and Smad4 loss, is needed to increase the understanding of malignant cell-stromal cell cross-talk in PDAC. In this study, we used single-cell RNA sequencing to analyze the cellular landscape of Trp53-mutant mouse models driven by KrasG12D or KrasG12V, in which Smad4 was wild type or deleted. KrasG12DSmad4-deleted PDAC developed a fibro-inflammatory rich stroma with increased malignant JAK/STAT cell signaling and enhanced therapeutic response to JAK/STAT inhibition. SMAD4 loss in KrasG12V PDAC differently altered the tumor microenvironment compared with KrasG12D PDAC, and the malignant compartment lacked JAK/STAT signaling dependency. Thus, malignant cell genotype affects cancer cell and stromal cell phenotypes in PDAC, directly affecting therapeutic efficacy. Significance: SMAD4 loss differentially impacts malignant cell-stromal cell signaling and treatment sensitivity of pancreatic tumors driven by KRASG12D or KRASG12V, highlighting the importance of understanding genotype-phenotype relationships for precision therapy.
Collapse
Affiliation(s)
- Eloise G. Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Muntadher Jihad
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Judhell S. Manansala
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Wenlong Li
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Priscilla S.W. Cheng
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Gianluca Mucciolo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Marta Zaccaria
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Sara Pinto Teles
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Sneha Harish
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Rebecca Brais
- Histopathology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke’s Hospital, Cambridge, UK
| | - Sally Ashworth
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Weike Luo
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Paul M. Johnson
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Mireia Vallespinos
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson way, CB2 0RE, Cambridge, UK
| |
Collapse
|
10
|
Tang J, Li X, Tang N, Lin X, Du Y, Zhang S, Li Q, Zhang Y, Zhang Y, Hang H, Qiu T, Qiu Y, Cheng H, Dai Z, Hong H, Wei W, He J, Yan C. CD44 identified as a diagnostic biomarker for highly malignant CA19-9 negative pancreatic cancer. Cancer Lett 2025; 622:217713. [PMID: 40216152 DOI: 10.1016/j.canlet.2025.217713] [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: 02/26/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited diagnostic biomarkers. Carbohydrate antigen 19-9 (CA19-9) is a widely used clinical biomarker and is generally considered to correlate with PDAC malignancy. However, the relationship between CA19-9 expression levels and tumor aggressiveness remains underexplored. In this study, we report a biphasic relationship between CA19-9 expression levels and PDAC malignancy, where both negative (<5 U/mL) and high (>37 U/mL) CA19-9 levels are associated with increased tumor aggressiveness. We defined CA19-9 negative PDAC as tumors that lack CA19-9 expression intracellulary, on the cell membrane, and in secreted form. In PDAC cell lines and patient-derived organoids, CA19-9 negativity, confirmed by immunofluorescence, flow cytometry and ELISA, correlated with more aggressive behaviors. In PDAC patients, tumors from those with serum CA19-9 levels below 5 U/mL exhibited stronger metabolically activity, more immunosuppressive tumor microenvironment, and worse survival than CA19-9 positive tumors, with over 90 % showing absent CA19-9 expression by immunohistochemistry (IHC). Glycoproteomics profiling identified CD44 as a highly expressed biomarker in CA19-9 negative PDAC. Elevated CD44 expression effectively distinguished CA19-9 negative PDAC from both CA19-9 positive PDAC and CA19-9 negative benign pancreatic diseases, suggesting its potential as a diagnostic tool. Furthermore, we developed a radionuclide-labeled CD44 antibody 89Zr-1M2E3, which specifically recognized CA19-9 negative PDAC tumors in preclinical models using PET-CT imaging. These findings highlight CD44 as a promising biomarker and therapeutic target for diagnosing and treating CA19-9 negative PDAC.
Collapse
Affiliation(s)
- Jiatong Tang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaoyang Li
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Neng Tang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiawen Lin
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixiang Du
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China
| | - Shuo Zhang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qi Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yifan Zhang
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hexing Hang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tongtong Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hao Cheng
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhan Dai
- Nanjing Okay Biotechnology Co., Ltd, Nanjing, Jiangsu Provinve, China
| | - Hao Hong
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Wei Wei
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
11
|
Azeez A, Noel C. Global status of research on quality of life in pancreatic cancer patients: A bibliometric and network analysis from 2005-2024. Clin Res Hepatol Gastroenterol 2025; 49:102595. [PMID: 40210107 DOI: 10.1016/j.clinre.2025.102595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is a major global health challenge, with rising incidence and mortality rates, particularly in high-socio-demographic index regions. Given its high mortality and significant morbidity, research on patient quality of life (QoL) has gained momentum, addressing symptom burdens, psychological distress, and treatment-related outcomes. Bibliometric analysis provides a valuable approach to mapping research trends, identifying key contributors, and forecasting future directions. OBJECTIVE This study aimed to map global research on QoL in pancreatic cancer patients, highlighting key findings, challenges, and future directions through bibliometric analysis over the past two decades. METHODS Data for this study were collected from the Web of Science Core Collection (WoSCC) database, using specific search strategies to retrieve relevant documents on the quality of life in pancreatic cancer patients. The data were analysed using the Bibliometrix R package to create knowledge maps and visualize research trends, collaborations, and emerging hotspots in the field. RESULTS A total of 819 articles on pancreatic cancer and quality of life were identified, with an average citation count of 47.13 per article, highlighting moderate academic impact. The research revealed a growing trend in collaborative efforts, with an average of 9.42 co-authors per article and 16 % international collaborations. The United States emerged as the leading contributor, with 203 publications and the highest citation count, followed by France and the United Kingdom. CONCLUSION This bibliometric analysis highlights the growing volume of pancreatic cancer and quality of life research, with a steady annual growth rate of 6.9 % and increasing collaboration, especially from the United States. However, despite the rising number of publications, a decline in citation impact for recent studies suggests a need for continued innovation in therapeutic strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Adeboye Azeez
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa.
| | - Colin Noel
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
12
|
Cramer E, Lopez-Vidal T, Johnson J, Wang V, Bergman D, Weeraratna A, Burkhart R, Fertig EJ, Zimmerman JW, Heiser LM, Chang YH. Automated Quality Control of Time-Course Imaging from 3D in vitro cultures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646437. [PMID: 40236164 PMCID: PMC11996402 DOI: 10.1101/2025.03.31.646437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Longitudinal imaging of 3D cell cultures like tumor organoids and spheroids offers crucial insights into cancer progression and treatment. However, spatial displacement during time-course imaging, caused by matrix detachment or experimental artifacts, can confound analyses. Existing computational methods struggle to address this issue. We present a new algorithm to evaluate data integrity and rectify mislabeling in longitudinal imaging of 3D cell culture. Our algorithm integrates permutation-based optimization with Procrustes analysis. By using X and Y coordinates of images, it accurately reorders, matches, and aligns object positions across time points, correcting for rotation, translation, and small movements. Validation with simulated data confirmed its accuracy and robustness. Applied to longitudinal imaging of tumor spheroids, our algorithm revealed frequent displacement amongst the spheroids between time points and corrected many mislabeled images. This computationally efficient and adaptable method needs no experimental adjustments and presents a readily accessible solution for data quality control. Motivation Three-dimensional (3D) in vitro models, such as tumor organoids and spheroids embedded in an extracellular matrix, are increasingly vital for studying normal and disease biology, including drug responses. 1-3 A key advantage of these models is that imaging platforms can perform continuous longitudinal imaging to track phenotypic changes. However, common issues in 3D techniques, such as matrix shifts during experimental setup or image capture, can introduce technical artifacts that affect downstream analyses. Currently, no automated analytical approaches exist for assessing or correcting technical artifacts. Here, we introduce a robust, automated algorithm for assessing the quality of time-course image data and, in some cases, correcting object mislabeling to enable accurate tracking of individual spheroids over time. This approach relies only on image metadata, requiring no experimental modifications. It offers a readily implementable solution for improving data integrity and reproducibility and enhancing the reliability of longitudinal 3D cell culture studies.
Collapse
|
13
|
Kim SY, van de Wetering M, Clevers H, Sanders K. The future of tumor organoids in precision therapy. Trends Cancer 2025:S2405-8033(25)00073-1. [PMID: 40185656 DOI: 10.1016/j.trecan.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Tumoroids are cultures of patient-derived tumor cells, which are grown in 3D in the presence of an extracellular matrix extract and specific growth factors. Tumoroids can be generated from adult as well as pediatric cancers, including epithelial cancers, sarcomas, and brain cancers. Tumoroids retain multi-omic characteristics of their corresponding tumor and recapitulate interpatient and intratumor heterogeneity. Retrospective and prospective studies have demonstrated that tumoroids predict patient responses to anticancer therapies, making them a promising tool for precision oncology. However, several challenges remain before tumoroids can be fully integrated into clinical decision-making, including success rates of tumoroid establishment and turnaround times. This review discusses the current advances, challenges, and future directions of tumoroid-based models in cancer research and precision therapy.
Collapse
Affiliation(s)
- Seok-Young Kim
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, The Netherlands; Current address: Roche Pharmaceutical Research and Early Development (pRED) of F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Karin Sanders
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Penrose HM, Sinha S, Tindle C, Zablan K, Le HN, Neill J, Ghosh P, Boland BS. A Living Organoid Biobank of Crohn's Disease Patients Reveals Distinct Clinical Correlates of Molecular Subtypes of Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.01.25325058. [PMID: 40236416 PMCID: PMC11998810 DOI: 10.1101/2025.04.01.25325058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Current clinical decision-making lacks reliable preclinical models to predict patient outcomes. Here, we establish patient-derived organoids (PDOs) as predictive tools in Crohn's disease (CD), a complex, heterogeneous disorder. Using a living biobank of adult stem cell-derived colonic-PDOs, we identified two molecular CD subtypes-Immune-Deficient Infectious CD ( IDICD ) and Stress and Senescence-Induced Fibrostenotic CD ( S2FCD )-each with distinct genomic, transcriptomic and functional profiles, along with paired therapeutics. By prospectively linking colonic PDO-derived phenotypes to real-world patient outcomes, we uncovered that while S2FCD associates with severe colonic disease, IDICD associates with severe ileal disease, prior ileocecal surgery, and future disease progression. This approach transforms PDOs from static descriptive models into dynamic tools that capture the past, present, and future of disease behavior and reveals their utility as patient-specific predictive platforms, extending their use beyond oncology to complex inflammatory diseases. Findings also suggest that colonic immune dysfunction may drive ileal-CD, independent of colonic involvement. GRAPHICAL ABSTRACT In Brief In this work, Penrose et al. demonstrate the potential of patient-derived organoids (PDOs) as predictive tools in Crohn's disease (CD) that capture the past, present, and future of disease behavior, thereby advancing PDO-informed precision medicine beyond oncology into complex inflammatory diseases. HIGHLIGHTS A living PDO biobank identified two molecular CD subtypes with distinct functional phenotypes.PDO subtyping tracked severity of ileal disease, prior surgery and future disease progression.Colonic immune dysfunction may drive ileal-CD, independent of colonic involvement.Colonic CD-PDOs are dynamic platforms for outcome-deterministic therapeutic testing.
Collapse
|
15
|
Zhou E, Yang JI, Habowski AN, Deschênes A, Belleau P, Ha T, Tzanavaris CJ, Boyd J, Hollweg CA, Zhu X, Tuveson DA, King DA. GATA6 Amplification Is Associated With Improved Survival in TP53-Mutated Unresectable Pancreatic Cancer. Pancreas 2025; 54:e303-e309. [PMID: 40262102 DOI: 10.1097/mpa.0000000000002431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/01/2024] [Indexed: 04/24/2025]
Abstract
OBJECTIVES GATA6 expression is recognized as a favorable prognostic marker of pancreatic cancer, whereas TP53 is a poor prognostic marker. We evaluated treatment outcomes by genetic alterations in TP53 and GATA6 to determine the prognostic and predictive impact of co-alterations. MATERIALS AND METHODS A single institution retrospective analysis was performed on patients diagnosed with pancreatic ductal adenocarcinoma between 2014 and 2023. TP53 genotype and GATA6 amplification status were included in an analysis of overall survival (OS) and progression-free survival (PFS). Previously published patient-derived organoids were used to investigate correlation between genetic status and drug sensitivity. RESULTS Patients with TP53 mutations had worse OS compared with the wild-type TP53 population. Patients with GATA6 amplification had better OS and a trend toward better PFS than the nonamplified population. Among patients with a TP53 mutation, patients with GATA6 co-alteration had longer OS compared with those who were not GATA6 amplified. In contrast, among patients who were TP53 wild-type, the presence or absence of a GATA6 amplification did not impact OS or PFS. GATA6 genotype was not associated chemotherapy drug response in an organoid pharmacotyping model. CONCLUSIONS We found that GATA6 amplification appeared to attenuate poor prognosis observed in TP53-mutant patients regardless of the type of standard chemotherapy received, suggesting the GATA6 amplification is a prognostic biomarker but not a predictive biomarker of standard-of-care chemotherapy.
Collapse
Affiliation(s)
- Edward Zhou
- Northwell Health Cancer Institute, New York, NY
| | | | | | | | - Pascal Belleau
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Taehoon Ha
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Chris J Tzanavaris
- Division of Pulmonary, Critical Care, and Sleep Medicine, Northwell Health, New York, NY
| | - Jeff Boyd
- Northwell Health Cancer Institute, New York, NY
| | - Christopher A Hollweg
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, NY
| | - Xinhua Zhu
- Northwell Health Cancer Institute, New York, NY
| | | | | |
Collapse
|
16
|
Fiorini E, Malinova A, Schreyer D, Pasini D, Bevere M, Alessio G, Rosa D, D'Agosto S, Azzolin L, Milite S, Andreani S, Lupo F, Veghini L, Grimaldi S, Pedron S, Castellucci M, Nourse C, Salvia R, Malleo G, Ruzzenente A, Guglielmi A, Milella M, Lawlor RT, Luchini C, Agostini A, Carbone C, Pilarsky C, Sottoriva A, Scarpa A, Tuveson DA, Bailey P, Corbo V. MYC ecDNA promotes intratumour heterogeneity and plasticity in PDAC. Nature 2025; 640:811-820. [PMID: 40074906 PMCID: PMC12003172 DOI: 10.1038/s41586-025-08721-9] [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: 10/06/2023] [Accepted: 01/30/2025] [Indexed: 03/14/2025]
Abstract
Intratumour heterogeneity and phenotypic plasticity drive tumour progression and therapy resistance1,2. Oncogene dosage variation contributes to cell-state transitions and phenotypic heterogeneity3, thereby providing a substrate for somatic evolution. Nonetheless, the genetic mechanisms underlying phenotypic heterogeneity are still poorly understood. Here we show that extrachromosomal DNA (ecDNA) is a major source of high-level focal amplification in key oncogenes and a major contributor of MYC heterogeneity in pancreatic ductal adenocarcinoma (PDAC). We demonstrate that ecDNAs drive varying levels of MYC dosage, depending on their regulatory landscape, enabling cancer cells to rapidly and reversibly adapt to microenvironmental changes. In the absence of selective pressure, a high ecDNA copy number imposes a substantial fitness cost on PDAC cells. We also show that MYC dosage affects cell morphology and dependence of cancer cells on stromal niche factors. Our work provides a detailed analysis of ecDNAs in PDAC and describes a new genetic mechanism driving MYC heterogeneity in PDAC.
Collapse
Affiliation(s)
- Elena Fiorini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Antonia Malinova
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Daniel Schreyer
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Davide Pasini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Michele Bevere
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Giorgia Alessio
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Diego Rosa
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Sabrina D'Agosto
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Human Technopole, Milan, Italy
| | - Luca Azzolin
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Salvatore Milite
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Silvia Andreani
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany
| | - Francesca Lupo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Sonia Grimaldi
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Serena Pedron
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Craig Nourse
- Cancer Research UK Beatson Institute, Glasgow, UK
- Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal
| | - Roberto Salvia
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona, Verona, Italy
| | - Giuseppe Malleo
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona, Verona, Italy
| | - Andrea Ruzzenente
- Department of Surgical Sciences, Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Alfredo Guglielmi
- Department of Surgical Sciences, Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Michele Milella
- Section of Medical Oncology, Department of Medicine, University of Verona, Verona, Italy
| | - Rita T Lawlor
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Antonio Agostini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Rome, Italy
| | - Carmine Carbone
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Andrea Sottoriva
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Aldo Scarpa
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Peter Bailey
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
- Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal.
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
- ARC-Net Research Centre, University of Verona, Verona, Italy.
| |
Collapse
|
17
|
Gutmann DH, Boehm JS, Karlsson EK, Padron E, Seshadri M, Wallis D, Snyder JC. Precision preclinical modeling to advance cancer treatment. J Natl Cancer Inst 2025; 117:586-594. [PMID: 39383197 PMCID: PMC11972679 DOI: 10.1093/jnci/djae249] [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: 06/25/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024] Open
Abstract
A new era of cancer management is underway in which treatments are being developed for the entire continuum of the disease process. The availability of genetically engineered and naturally occurring preclinical models serves as instructive platforms for evaluating therapeutic mechanisms. However, a major clinical challenge is that the entire malignancy process occurs across multiple scales including genetic mutations, malignant changes in cell behavior, dysregulated tumor microenvironments, and systemic adaptations in the host. A multidisciplinary group of investigators coalesced at the National Cancer Institute Oncology Models Forum with the overall goal to provide updates on the use of precision preclinical models of cancer. The benefits and limitations of preclinical models were discussed to identify strategies for maximizing opportunities in modeling that could inform future cancer prevention and treatment approaches. Our shared perspective is that the continuum of single cell, multicell, organoid, and in situ models are remarkable resources for the clinical challenges ahead. We provide a roadmap for parsing already available models and include preliminary recommendations for the application of next-generation preclinical modeling in cancer intervention.
Collapse
Affiliation(s)
- David H Gutmann
- Department of Neurology, Washington University, St Louis, MO 63110, United States
| | - Jesse S Boehm
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Elinor K Karlsson
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
- Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Mukund Seshadri
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Deeann Wallis
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Joshua C Snyder
- Department of Surgery, Duke University, Durham, NC 27710, United States
| |
Collapse
|
18
|
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
|
19
|
Mukhare R, Gandhi KA, Kadam A, Raja A, Singh A, Madhav M, Chaubal R, Pandey S, Gupta S. Integration of Organoids With CRISPR Screens: A Narrative Review. Biol Cell 2025; 117:e70006. [PMID: 40223602 PMCID: PMC11995251 DOI: 10.1111/boc.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025]
Abstract
Organoids represent a significant advancement in disease modeling, demonstrated by their capacity to mimic the physiological/pathological structure and functional characteristics of the native tissue. Recently CRISPR/Cas9 technology has emerged as a powerful tool in combination with organoids for the development of novel therapies in preclinical settings. This review explores the current literature on applications of pooled CRISPR screening in organoids and the emerging role of these models in understanding cancer. We highlight the evolution of genome-wide CRISPR gRNA library screens in organoids, noting their increasing adoption in the field over the past decade. Noteworthy studies utilizing these screens to investigate oncogenic vulnerabilities and developmental pathways in various organoid systems are discussed. Despite the promise organoids hold, challenges such as standardization, reproducibility, and the complexity of data interpretation remain. The review also addresses the ideas of assessing tumor organoids (tumoroids) against established cancer hallmarks and the potential of studying intercellular cooperation within these models. Ultimately, we propose that organoids, particularly when personalized for patient-specific applications, could revolutionize drug screening and therapeutic approaches, minimizing the reliance on traditional animal models and enhancing the precision of clinical interventions.
Collapse
Affiliation(s)
- Rushikesh Mukhare
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Khushboo A. Gandhi
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Anushree Kadam
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Aishwarya Raja
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Ankita Singh
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Mrudula Madhav
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Rohan Chaubal
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Surgical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Shwetali Pandey
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Sudeep Gupta
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| |
Collapse
|
20
|
Kashkin K, Kondratyeva L, Kopantzev E, Abramov I, Zhukova L, Chernov I. Deciphering of SOX9 Functions in Pancreatic Cancer Cells. Int J Mol Sci 2025; 26:2652. [PMID: 40141294 PMCID: PMC11941869 DOI: 10.3390/ijms26062652] [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: 01/23/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
SOX9 is widely regarded as a key master regulator of gene transcription, responsible for the development and differentiation programs within tissue and organogenesis, particularly in the pancreas. SOX9 overexpression has been observed in multiple tumor types, including pancreatic cancer, and is discussed as a prognostic marker. In order to gain a more profound understanding of the role of SOX9 in pancreatic cancer, we have performed SOX9 knockdown in the COLO357 and PANC-1 cells using RNA interference, followed by full-transcriptome analysis of the siRNA-transfected cells. The molecular pathway enrichment analysis between SOX9-specific siRNA-transfected cells and control cells reveals the activation of processes associated with cellular signaling, cell differentiation, transcription, and methylation, alongside the suppression of genes involved in various stages of the cell cycle and apoptosis, upon the SOX9 knockdown. Alterations of the expression of transcription factors, epithelial-mesenchymal transition markers, oncogenes, tumor suppressor genes, and drug resistance-related genes upon SOX9 knockdown in comparison of primary and metastatic pancreatic cancer cells are discovered. The expression levels of genes comprising prognostic signatures for pancreatic cancer were also evaluated following SOX9 knockdown. Additional studies are needed to assess the properties and prognostic significance of SOX9 in pancreatic cancer using other biological models.
Collapse
Affiliation(s)
- Kirill Kashkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| | - Liya Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| | - Eugene Kopantzev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| | - Ivan Abramov
- GBUZ Moscow Clinical Scientific and Practical Center Named After A.S. Loginov MHD (MCSC), 111123 Moscow, Russia; (I.A.); (L.Z.)
| | - Lyudmila Zhukova
- GBUZ Moscow Clinical Scientific and Practical Center Named After A.S. Loginov MHD (MCSC), 111123 Moscow, Russia; (I.A.); (L.Z.)
| | - Igor Chernov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, 117997 Moscow, Russia; (E.K.); (I.C.)
| |
Collapse
|
21
|
Binkowski B, Klamer Z, Gao C, Staal B, Repesh A, Tran HL, Brass DM, Bartlett P, Gallinger S, Blomqvist M, Morrow JB, Allen P, Shi C, Singhi A, Brand R, Huang Y, Hostetter G, Haab BB. Multiplexed glycan immunofluorescence identification of pancreatic cancer cell subpopulations in both tumor and blood samples. SCIENCE ADVANCES 2025; 11:eadt0029. [PMID: 40053601 PMCID: PMC11917494 DOI: 10.1126/sciadv.adt0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/30/2025] [Indexed: 03/09/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumor heterogeneity impedes the development of biomarker assays for early disease detection. We hypothesized that PDAC cell subpopulations could be identified by aberrant glycan signatures in both tumor tissue and blood samples. We used multiplexed glycan immunofluorescence to distinguish between PDAC and noncancer cell subpopulations within tumor tissue, and we developed hybrid glycan sandwich assays to determine whether the aberrant glycan signatures could be detected in blood samples. We found that PDAC cells were identified by signatures of glycans detected by four glycan-binding proteins (VVL, CA19-9, sTRA, and GM2) and that there are three types of glycan-defined PDAC tumors: sTRA type, CA19-9 type, and intermixed. In patient-matched tumor and blood samples, the PDAC tumor type could be determined by the aberrant glycans in the blood. As a result, the combined assays of aberrant glycan signatures were more sensitive and specific than any individual assay. Our results demonstrate a methodology to detect and stratify PDAC.
Collapse
Affiliation(s)
| | | | | | - Ben Staal
- Van Andel Institute, Grand Rapids, MI, USA
| | | | | | | | | | | | - Maria Blomqvist
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Peter Allen
- Duke University School of Medicine, Durham, NC, USA
| | - Chanjuan Shi
- Duke University School of Medicine, Durham, NC, USA
| | - Aatur Singhi
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Randall Brand
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ying Huang
- Fred Hutchinson Cancer Research Center; Seattle, WA, USA
| | | | | |
Collapse
|
22
|
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
|
23
|
Ding J, Xie Y, Liu Z, Zhang Z, Ni B, Yan J, Zhou T, Hao J. Hypoxic and Acidic Tumor Microenvironment-Driven AVL9 Promotes Chemoresistance of Pancreatic Ductal Adenocarcinoma via the AVL9-IκBα-SKP1 Complex. Gastroenterology 2025; 168:539-555.e5. [PMID: 39566663 DOI: 10.1053/j.gastro.2024.10.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 09/24/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND & AIMS Gemcitabine combined with albumin-paclitaxel (AG) is a crucial therapeutic option for pancreatic ductal adenocarcinoma (PDAC). However, the response to chemotherapy is relatively poor, with rapid development of resistance. The aim of this study was to explore the mechanism of resistance to AG and to develop strategies that can sensitize the AG regimen. METHODS We used organoid models, patient-derived xenografts, and genetically engineered mouse models in our study. Chromatin immunoprecipitation, double luciferase assay, co-immunoprecipitation, and far-western blotting analysis were performed to investigate the mechanism. The AVL9 inhibitors were identified through protein structure analysis and molecular docking analysis, and their efficacy was verified in patient-derived xenografts, patient-derived organoids-based xenograft, and KPC models. RESULTS Through multistrategy screening, we identified AVL9 as a key target for AG resistance in PDAC. Its tumor-promoting effects were confirmed in our clinical cohorts. Mechanistically, HIF-1α, a hypoxia-related transcription factor, drives the expression of AVL9. AVL9 acts as a scaffold that facilitates the binding of IκBα to SKP1, leading to enhanced ubiquitination and degradation of IκBα, which further activates the nuclear factor-κB pathway. The potential AVL9-targeting inhibitor, Edotecarin, was shown to reverse AG chemo-resistance in PDAC. CONCLUSION AVL9 expression is driven by HIF-1α in PDAC. The physical interaction of AVL9, IκBα, and SKP1 provides a novel molecular mechanism for the abnormal activation of the nuclear factor-κB pathway. Therefore, the AVL9-targeting drug Edotecarin could be a promising therapeutic strategy for sensitizing PDAC to AG.
Collapse
Affiliation(s)
- Jinsheng Ding
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China; Department of Breast Oncoplastic Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, People's Republic of China
| | - Yongjie Xie
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Ziyun Liu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Zhaoyu Zhang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Bo Ni
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Jingrui Yan
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Tianxing Zhou
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
| |
Collapse
|
24
|
Boilève A, Ducreux M, Jaulin F. Reply. Gastroenterology 2025; 168:628-630. [PMID: 39571821 DOI: 10.1053/j.gastro.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 12/22/2024]
Affiliation(s)
- Alice Boilève
- INSERM U1279, Gustave Roussy, Villejuif, France; Département de Médecine, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France
| | - Michel Ducreux
- INSERM U1279, Gustave Roussy, Villejuif, France; Département de Médecine, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France
| | - Fanny Jaulin
- INSERM U1279, Gustave Roussy, Orsay, France; Département de Recherche, Gustave Roussy, Villejuif, France; Université Paris Saclay, Orsay, France
| |
Collapse
|
25
|
Zhao KY, Du YX, Cao HM, Su LY, Su XL, Li X. The biological macromolecules constructed Matrigel for cultured organoids in biomedical and tissue engineering. Colloids Surf B Biointerfaces 2025; 247:114435. [PMID: 39647422 DOI: 10.1016/j.colsurfb.2024.114435] [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: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Matrigel is the most commonly used matrix for 3D organoid cultures. Research on the biomaterial basis of Matrigel for organoid cultures is a highly challenging field. Currently, many studies focus on Matrigel-based biological macromolecules or combinations to construct natural Matrigel and synthetic hydrogel scaffolds based on collagen, peptides, polysaccharides, microbial transglutaminase, DNA supramolecules, and polymers for organoid culture. In this review, we discuss the limitations of both natural and synthetic Matrigel, and describe alternative scaffolds that have been employed for organoid cultures. The patient-derived organoids were constructed in different cancer types and limitations of animal-derived organoids based on the hydrogel or Matrigel. The constructed techniques utilizing 3D bioprinting platforms, air-liquid interface (ALI) culture, microfluidic culture, and organ-on-a-chip platform are summarized. Given the potential of organoids for a wide range of therapeutic, tissue engineering and pharmaceutical applications, it is indeed imperative to develop defined and customized hydrogels in addition to Matrigel.
Collapse
Affiliation(s)
- Ke-Yu Zhao
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Yi-Xiang Du
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Hui-Min Cao
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Li-Ya Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Xiu-Lan Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Xian Li
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China.
| |
Collapse
|
26
|
Tsukamoto S, Huaze Y, Weisheng Z, Machinaga A, Kakiuchi N, Ogawa S, Seno H, Higashiyama S, Matsuda M, Hiratsuka T. Quantitative Live Imaging Reveals Phase Dependency of PDAC Patient-Derived Organoids on ERK and AMPK Activity. Cancer Sci 2025; 116:724-735. [PMID: 39731327 PMCID: PMC11875792 DOI: 10.1111/cas.16439] [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: 08/24/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/29/2024] Open
Abstract
Patient-derived organoids represent a novel platform to recapitulate the cancer cells in the patient tissue. While cancer heterogeneity has been extensively studied by a number of omics approaches, little is known about the spatiotemporal kinase activity dynamics. Here we applied a live imaging approach to organoids derived from 10 pancreatic ductal adenocarcinoma (PDAC) patients to comprehensively understand their heterogeneous growth potential and drug responses. By automated wide-area image acquisitions and analyses, the PDAC cells were non-selectively observed to evaluate their heterogeneous growth patterns. We monitored single-cell ERK and AMPK activities to relate cellular dynamics to molecular dynamics. Furthermore, we evaluated two anti-cancer drugs, a MEK inhibitor, PD0325901, and an autophagy inhibitor, hydroxychloroquine (HCQ), by our analysis platform. Our analyses revealed a phase-dependent regulation of PDAC organoid growth, where ERK activity is necessary for the early phase and AMPK activity is necessary for the late stage of organoid growth. Consistently, we found PD0325901 and HCQ target distinct organoid populations, revealing their combination is widely effective to the heterogeneous cancer cell population in a range of PDAC patient-derived organoid lines. Together, our live imaging quantitatively characterized the growth and drug sensitivity of human PDAC organoids at multiple levels: in single cells, single organoids, and individual patients. This study will pave the way for understanding the cancer heterogeneity and promote the development of new drugs that eradicate intractable cancer.
Collapse
Affiliation(s)
- Shoko Tsukamoto
- Laboratory of Cell Cycle Regulation, Graduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Ye Huaze
- Department of Molecular Oncology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Zhang Weisheng
- Department of Molecular Oncology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Akihito Machinaga
- Oncology Tsukuba Research Department, Discovery, Medicine CreationOBG, Eisai Co. Ltd.TsukubaJapan
| | - Nobuyuki Kakiuchi
- Department of Gastroenterology and Hepatology, Graduate School of MedicineKyoto UniversityKyotoJapan
- The Hakubi Center for Advanced ResearchKyoto UniversityKyotoJapan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroshi Seno
- The Hakubi Center for Advanced ResearchKyoto UniversityKyotoJapan
| | - Shigeki Higashiyama
- Department of Oncogenesis and Growth Regulation, Research CenterOsaka International Cancer InstituteOsakaJapan
| | - Michiyuki Matsuda
- Laboratory of Cell Cycle Regulation, Graduate School of BiostudiesKyoto UniversityKyotoJapan
- Affiliated Graduate School, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Toru Hiratsuka
- Department of Molecular Oncology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Oncogenesis and Growth Regulation, Research CenterOsaka International Cancer InstituteOsakaJapan
| |
Collapse
|
27
|
Hu Y, Peng Z, Qiu M, Xue L, Ren H, Wu X, Zhu X, Ding Y. Developing biotechnologies in organoids for liver cancer. BIOMEDICAL TECHNOLOGY 2025; 9:100067. [DOI: 10.1016/j.bmt.2024.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
28
|
Barcena-Varela M, Monga SP, Lujambio A. Precision models in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2025; 22:191-205. [PMID: 39663463 DOI: 10.1038/s41575-024-01024-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 12/13/2024]
Abstract
Hepatocellular carcinoma (HCC) represents a global health challenge, and ranks among one of the most prevalent and deadliest cancers worldwide. Therapeutic advances have expanded the treatment armamentarium for patients with advanced HCC, but obstacles remain. Precision oncology, which aims to match specific therapies to patients who have tumours with particular features, holds great promise. However, its implementation has been hindered by the existence of numerous 'HCC influencers' that contribute to the high inter-patient heterogeneity. HCC influencers include tumour-related characteristics, such as genetic alterations, immune infiltration, stromal composition and aetiology, and patient-specific factors, such as sex, age, germline variants and the microbiome. This Review delves into the intricate world of HCC, describing the most innovative model systems that can be harnessed to identify precision and/or personalized therapies. We provide examples of how different models have been used to nominate candidate biomarkers, their limitations and strategies to optimize such models. We also highlight the importance of reproducing distinct HCC influencers in a flexible and modular way, with the aim of dissecting their relative contribution to therapy response. Next-generation HCC models will pave the way for faster discovery of precision therapies for patients with advanced HCC.
Collapse
Affiliation(s)
- Marina Barcena-Varela
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satdarshan P Monga
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
29
|
Kim SY, de Weert TAE, Vermeulen M, Ringnalda F, Kester L, Zsiros J, Eising S, Molenaar JJ, Sanders K, van de Wetering M, Clevers H. Organoid drug profiling identifies methotrexate as a therapy for SCCOHT, a rare pediatric cancer. SCIENCE ADVANCES 2025; 11:eadq1724. [PMID: 40009666 PMCID: PMC11864178 DOI: 10.1126/sciadv.adq1724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare and lethal tumor in adolescent and young adult patients. Now, there is no standard-of-care treatment for these patients. Reliable models that represent this disease and can be used for translational research are scarce. To model SCCOHTs, we have established eight patient-derived tumoroid lines from tumor lesions of three patients with SCCOHT. The tumoroids recapitulate genomic and transcriptomic characteristics of the corresponding patient tumors and capture intrapatient tumor heterogeneity. Organoid drug profiling using a library of 153 clinical compounds identified methotrexate as an effective and selective drug against SCCOHTs with a clinically relevant IC50 of 35 nanomolars. RNA sequencing demonstrated that methotrexate induced TP53 pathway activation and apoptosis. These data underscore that organoid technology can support the design of therapeutic strategies for rare cancers.
Collapse
Affiliation(s)
- Seok-Young Kim
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Tamar A. E. de Weert
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Marijn Vermeulen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Femke Ringnalda
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Lennart Kester
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Jozsef Zsiros
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Selma Eising
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Jan J. Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Department of Pharmaceutical Sciences, University Utrecht, Utrecht, Netherlands
| | - Karin Sanders
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Marc van de Wetering
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
30
|
Li P, Huang M, Li M, Li G, Ma Y, Zhao Y, Wang X, Zhang Y, Shi C. Combining molecular characteristics and therapeutic analysis of PDOs predict clinical responses and guide PDAC personalized treatment. J Exp Clin Cancer Res 2025; 44:72. [PMID: 40001264 PMCID: PMC11863571 DOI: 10.1186/s13046-025-03332-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The emergence of targeted therapies and immunotherapy has broadened treatment options for patients with pancreatic ductal adenocarcinoma (PDAC). Despite this, traditional drug selection, predominantly relies on tumor markers and clinical staging, has underutilized these drugs due to ignoring patient genomic diversity. Patient-derived organoids (PDOs) and corresponding patient-derived organoid xenograft (PDOX) models offer a way to better understand and address this. METHODS In this study, we established PDOs and PDOX models from PDAC clinical samples. These models were analyzed using immunohistochemistry, H&E staining, and genomic profiling. Drug screening with 111 FDA-approved drugs was performed on PDOs, and drug responses in PDOs and PDOX models were compared to assess consistency with clinical treatment outcomes. Gene analysis was conducted to explore the molecular mechanisms underlying variations in drug responses. Additionally, by analyzing the sequencing results from various drug-sensitive groups, the identified differential gene-drug metabolism gene UGT1A10 were modulated in PDOs to evaluate its impact on drug efficacy. A co-culture system of PDOs with immune cells was developed to study the efficacy of immunotherapies. RESULTS PDOs and matched PDOX models retain the morphological, biological, and genomic characteristics of the primary tumor. Exome sequencing and RNA sequencing confirmed both the consistency and heterogeneity among the PDOs. High-throughput drug screening revealed significant variability in drug sensitivity across different organoids, yet PDOs and PDOX derived from the same patient exhibited a high degree of concordance in response to clinical chemotherapy agents. The gene expression analysis of PDOs with significant differences in drug sensitivity revealed UGT1A10 as a crucial regulator. The knockdown of UGT1A10 notably increased drug sensitivity. Furthermore, immune cells demonstrated specific cytotoxicity towards the organoids, underscoring the potential of the co-culture system for application in tumor immunotherapy. CONCLUSION Our results highlight the necessity for personalized treatment strategies that consider genomic diversity beyond tumor markers, thus validating the utility of PDOs and PDOX models in advancing PDAC research and personalized medicine.
Collapse
Affiliation(s)
- Peng Li
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, PR China
| | - Minli Huang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Mengyao Li
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Gen Li
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Yifan Ma
- Gansu University of Chinese Medicine, Lanzhou, 730030, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, PR China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, PR China.
| | - Yongbin Zhang
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China.
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, PR China.
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
31
|
Wang MJ, Gao C, Huang X, Wang M, Zhang S, Gao XP, Zhong CQ, Li LY. Establishing Pancreatic Cancer Organoids from EUS-Guided Fine-Needle Biopsy Specimens. Cancers (Basel) 2025; 17:692. [PMID: 40002285 PMCID: PMC11852484 DOI: 10.3390/cancers17040692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic cancer is a highly malignant digestive system tumor characterized by covert onset and rapid progression, with a 5-year survival rate of less than 10%. Most patients have already reached an advanced or metastatic stage at the time of diagnosis. Therefore, it is particularly important to study the occurrence, development, and drug resistance mechanisms of pancreatic cancer. In recent years, the development of 3D tumor cell culture technology has provided new avenues for pancreatic cancer research. Patient-derived organoids (PDOs) are micro-organ structures that are obtained directly from the patient's body and rapidly expand in vitro. PDOs have the ability to self-renew and self-organize and retain the genetic heterogeneity and molecular characteristics of the original tumor. However, the use of organoids is limited because most patients with pancreatic ductal adenocarcinoma (PDAC) are inoperable. Endoscopic ultrasound-guided fine-needle aspiration/biopsy (EUS-FNA/FNB) is an important method for obtaining tissue samples from non-surgical pancreatic cancer patients. This article reviews the factors that affect the formation of pancreatic cancer organoids using EUS-FNA/FNB. High-quality samples, sterile operations, and optimized culture media are key to successfully generating organoids. Additionally, individual patient differences and disease stages can impact the formation of organoids. Pancreatic cancer organoids constructed using EUS-FNA/FNB have significant potential, suggesting new approaches for research and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lian-Yong Li
- Department of Gastroenterology, The Ninth Medical Center of Chinese PLA General Hospital, Beijing 100101, China; (M.-J.W.); (C.G.); (X.H.); (M.W.); (S.Z.); (X.-P.G.); (C.-Q.Z.)
| |
Collapse
|
32
|
Charles S, Jackson-Holmes E, Sun G, Zhou Y, Siciliano B, Niu W, Han H, Nikitina A, Kemp ML, Wen Z, Lu H. Non-Invasive Quality Control of Organoid Cultures Using Mesofluidic CSTR Bioreactors and High-Content Imaging. ADVANCED MATERIALS TECHNOLOGIES 2025; 10:2400473. [PMID: 40248044 PMCID: PMC12002419 DOI: 10.1002/admt.202400473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Indexed: 04/19/2025]
Abstract
Human brain organoids produce anatomically relevant cellular structures and recapitulate key aspects of in vivo brain function, which holds great potential to model neurological diseases and screen therapeutics. However, the long growth time of 3D systems complicates the culturing of brain organoids and results in heterogeneity across samples hampering their applications. We developed an integrated platform to enable robust and long-term culturing of 3D brain organoids. We designed a mesofluidic bioreactor device based on a reaction-diffusion scaling theory, which achieves robust media exchange for sufficient nutrient delivery in long-term culture. We integrated this device with longitudinal tracking and machine learning-based classification tools to enable non-invasive quality control of live organoids. This integrated platform allows for sample pre-selection for downstream molecular analysis. Transcriptome analyses of organoids revealed that our mesofluidic bioreactor promoted organoid development while reducing cell death. Our platform thus offers a generalizable tool to establish reproducible culture standards for 3D cellular systems for a variety of applications beyond brain organoids.
Collapse
Affiliation(s)
- Seleipiri Charles
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Emily Jackson-Holmes
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Gongchen Sun
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Ying Zhou
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Benjamin Siciliano
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, 615 Michael Street, Atlanta, GA, 30322, U.S.A
| | - Weibo Niu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Haejun Han
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Biological Sciences, Georgia Institute of Technology, 310 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Arina Nikitina
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Melissa L Kemp
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Hang Lu
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| |
Collapse
|
33
|
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
|
34
|
Verstegen MMA, Coppes RP, Beghin A, De Coppi P, Gerli MFM, de Graeff N, Pan Q, Saito Y, Shi S, Zadpoor AA, van der Laan LJW. Clinical applications of human organoids. Nat Med 2025; 31:409-421. [PMID: 39901045 DOI: 10.1038/s41591-024-03489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/17/2024] [Indexed: 02/05/2025]
Abstract
Organoids are innovative three-dimensional and self-organizing cell cultures of various lineages that can be used to study diverse tissues and organs. Human organoids have dramatically increased our understanding of developmental and disease biology. They provide a patient-specific model to study known diseases, with advantages over animal models, and can also provide insights into emerging and future health threats related to climate change, zoonotic infections, environmental pollutants or even microgravity during space exploration. Furthermore, organoids show potential for regenerative cell therapies and organ transplantation. Still, several challenges for broad clinical application remain, including inefficiencies in initiation and expansion, increasing model complexity and difficulties with upscaling clinical-grade cultures and developing more organ-specific human tissue microenvironments. To achieve the full potential of organoid technology, interdisciplinary efforts are needed, integrating advances from biology, bioengineering, computational science, ethics and clinical research. In this Review, we showcase pivotal achievements in epithelial organoid research and technologies and provide an outlook for the future of organoids in advancing human health and medicine.
Collapse
Affiliation(s)
- Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Rob P Coppes
- Departments of Biomedical Sciences and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne Beghin
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Research and Engineering in Space Technology, Universite Libre de Bruxelles, Bruxelles, Belgium
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Mattia F M Gerli
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, UK
| | - Nienke de Graeff
- Department of Medical Ethics and Health Law, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Yoshimasa Saito
- Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Shaojun Shi
- Department of Organ Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| |
Collapse
|
35
|
Xu J, Pham MD, Corbo V, Ponz-Sarvise M, Oni T, Öhlund D, Hwang CI. Advancing pancreatic cancer research and therapeutics: the transformative role of organoid technology. Exp Mol Med 2025; 57:50-58. [PMID: 39814914 PMCID: PMC11799150 DOI: 10.1038/s12276-024-01378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 01/18/2025] Open
Abstract
Research on pancreatic cancer has transformed with the advent of organoid technology, providing a better platform that closely mimics cancer biology in vivo. This review highlights the critical advancements facilitated by pancreatic organoid models in understanding disease progression, evaluating therapeutic responses, and identifying biomarkers. These three-dimensional cultures enable the proper recapitulation of the cellular architecture and genetic makeup of the original tumors, providing insights into the complex molecular and cellular dynamics at various stages of pancreatic ductal adenocarcinoma (PDAC). We explore the applications of pancreatic organoids in dissecting the tumor microenvironment (TME); elucidating cancer progression, metastasis, and drug resistance mechanisms; and personalizing therapeutic strategies. By overcoming the limitations of traditional 2D cultures and animal models, the use of pancreatic organoids has significantly accelerated translational research, which is promising for improving diagnostic and therapeutic approaches in clinical settings, ultimately aiming to improve the outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Jihao Xu
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Minh Duc Pham
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Mariano Ponz-Sarvise
- Department of Medical Oncology and Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Pamplona, Spain
| | - Tobiloba Oni
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Daniel Öhlund
- Umeå University, Department of Diagnostics and Intervention, and Wallenberg Centre for Molecular Medicine at Umeå University, Umeå, Sweden
| | - Chang-Il Hwang
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA.
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
36
|
Tapia Contreras C, Falke JD, Seifert D, Schneider C, Krauß L, Fang X, Müller D, Demirdizen E, Spitzner M, De Oliveira T, Schneeweis C, Gaedcke J, Kaulfuß S, Mirzakhani K, Wollnik B, Conrads K, Beißbarth T, Salinas G, Hügel J, Beyer N, Rheinländer S, Sax U, Wirth M, Conradi L, Reichert M, Ellenrieder V, Ströbel P, Ghadimi M, Grade M, Saur D, Hessmann E, Schneider G. KRAS G 12C-inhibitor-based combination therapies for pancreatic cancer: insights from drug screening. Mol Oncol 2025; 19:295-310. [PMID: 39253995 PMCID: PMC11792994 DOI: 10.1002/1878-0261.13725] [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: 11/03/2023] [Revised: 06/06/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has limited treatment options, emphasizing the urgent need for effective therapies. The predominant driver in PDAC is mutated KRAS proto-oncogene, KRA, present in 90% of patients. The emergence of direct KRAS inhibitors presents a promising avenue for treatment, particularly those targeting the KRASG12C mutated allele, which show encouraging results in clinical trials. However, the development of resistance necessitates exploring potent combination therapies. Our objective was to identify effective KRASG12C-inhibitor combination therapies through unbiased drug screening. Results revealed synergistic effects with son of sevenless homolog 1 (SOS1) inhibitors, tyrosine-protein phosphatase non-receptor type 11 (PTPN11)/Src homology region 2 domain-containing phosphatase-2 (SHP2) inhibitors, and broad-spectrum multi-kinase inhibitors. Validation in a novel and unique KRASG12C-mutated patient-derived organoid model confirmed the described hits from the screening experiment. Our findings propose strategies to enhance KRASG12C-inhibitor efficacy, guiding clinical trial design and molecular tumor boards.
Collapse
Affiliation(s)
| | - Jonas Dominik Falke
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Dana‐Magdalena Seifert
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Carolin Schneider
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Lukas Krauß
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Xin Fang
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Denise Müller
- Institute of PathologyUniversity Medical CenterGöttingenGermany
| | - Engin Demirdizen
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Melanie Spitzner
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
| | - Christian Schneeweis
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University MunichGermany
| | - Jochen Gaedcke
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
| | - Silke Kaulfuß
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Institute of Human GeneticsUniversity Medical Center GöttingenGermany
| | - Kimia Mirzakhani
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Institute of Human GeneticsUniversity Medical Center GöttingenGermany
| | - Bernd Wollnik
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Institute of Human GeneticsUniversity Medical Center GöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGermany
| | - Karly Conrads
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical BioinformaticsUniversity Medical Center GöttingenGermany
| | - Tim Beißbarth
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical BioinformaticsUniversity Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
- Campus‐Institute Data Science (CIDAS)GöttingenGermany
| | - Gabriela Salinas
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- NGS Integrative Genomics Core Unit (NIG)University Medical Center Göttingen (UMG)Germany
| | - Jonas Hügel
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Nils Beyer
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Sophia Rheinländer
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Ulrich Sax
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- Campus‐Institute Data Science (CIDAS)GöttingenGermany
- Department of Medical InformaticsUniversity Medical CenterGöttingenGermany
| | - Matthias Wirth
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Department of Hematology, Oncology and Cancer ImmunologyCampus Benjamin Franklin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinGermany
| | - Lena‐Christin Conradi
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic II, Klinikum rechts der IsarTechnical University MunichGermany
- Translational Pancreatic Research Cancer Center, Medical Clinic and Polyclinic II, Klinikum rechts der IsarTechnical University MunichGermany
- Center for Protein Assemblies (CPA)Technical University of MunichGarchingGermany
- Center for Organoid Systems and Tissue Engineering (COS)Technical University MunichGarchingGermany
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership Between DKFZ and University Hospital Klinikum rechts der IsarMunichGermany
| | - Volker Ellenrieder
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
- Department of Gastroenterology, Gastrointestinal Oncology and EndocrinologyUniversity Medical Center GöttingenGermany
| | - Philipp Ströbel
- Institute of PathologyUniversity Medical CenterGöttingenGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Marian Grade
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| | - Dieter Saur
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University MunichGermany
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership Between DKFZ and University Hospital Klinikum rechts der IsarMunichGermany
| | - Elisabeth Hessmann
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
- Department of Gastroenterology, Gastrointestinal Oncology and EndocrinologyUniversity Medical Center GöttingenGermany
| | - Günter Schneider
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGermany
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University MunichGermany
- Clinical Research Unit 5002, KFO5002University Medical Center GöttingenGermany
- CCC‐N (Comprehensive Cancer Center Lower Saxony)GöttingenGermany
| |
Collapse
|
37
|
Lum MA, Jonas KA, Parmar S, Black AR, O’Connor CM, Dobersch S, Yamamoto N, Robertson TM, Schutter A, Giambi M, Avelar RA, DiFeo A, Woods NT, Kugel S, Narla G, Black JD. Small-molecule modulators of B56-PP2A restore 4E-BP function to suppress eIF4E-dependent translation in cancer cells. J Clin Invest 2025; 135:e176093. [PMID: 39869680 PMCID: PMC11827888 DOI: 10.1172/jci176093] [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: 09/25/2023] [Accepted: 12/18/2024] [Indexed: 01/29/2025] Open
Abstract
Dysregulated eIF4E-dependent translation is a central driver of tumorigenesis and therapy resistance. eIF4E-binding proteins (4E-BP1/2/3) are major negative regulators of eIF4E-dependent translation that are inactivated in tumors through inhibitory phosphorylation or downregulation. Previous studies have linked PP2A phosphatase(s) to activation of 4E-BP1. Here, we leveraged biased small-molecule activators of PP2A (SMAPs) to explore the role of B56-PP2A(s) in 4E-BP regulation and the potential of B56-PP2A activation for restoring translational control in tumors. SMAP treatment promoted PP2A-dependent hypophosphorylation of 4E-BP1/2, supporting a role for B56-PP2As (e.g., B56α-PP2A) as 4E-BP phosphatases. Unexpectedly, SMAPs induced transcriptional upregulation of 4E-BP1 through a B56-PP2A→TFE3/TFEB→ATF4 axis. Cap-binding and coimmunoprecipitation assays showed that B56-PP2A(s) activation blocks assembly of the eIF4F translation initiation complex, and cap-dependent translation assays confirmed the translation-inhibitory effects of SMAPs. Thus, B56-PP2A(s) orchestrate a translation-repressive program involving transcriptional induction and activation of 4E-BP1. Notably, SMAPs promoted 4E-BP1-dependent apoptosis in tumor cells and potentiated 4E-BP1 function in the presence of ERK or mTOR inhibitors, agents that rely on inhibition of eIF4E-dependent translation for antitumor activity. These findings, combined with the ability of SMAPs to regulate 4E-BP1 in vivo, highlight the potential of PP2A activators for cancer therapy and overcoming therapy resistance.
Collapse
Affiliation(s)
- Michelle A. Lum
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Kayla A. Jonas
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Shreya Parmar
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Adrian R. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Caitlin M. O’Connor
- Division of Genetic Medicine, Department of Internal Medicine, and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephanie Dobersch
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Naomi Yamamoto
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tess M. Robertson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Aidan Schutter
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Miranda Giambi
- Division of Genetic Medicine, Department of Internal Medicine, and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Rita A. Avelar
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology and
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Analisa DiFeo
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology and
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas T. Woods
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| | - Sita Kugel
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, and
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer D. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, Omaha, Nebraska, USA
| |
Collapse
|
38
|
Xu X, Guo S, Gu H, Cha Z, Shi X, Yin X, Wang H, Gao S, Li B, Zhu L, Jing W, Zheng K, Shao Z, Cheng P, Zheng C, Shih YP, Li Y, Qian B, Gao D, Tran E, Jin G. Identification and validation of a T cell receptor targeting KRAS G12V in HLA-A*11:01 pancreatic cancer patients. JCI Insight 2025; 10:e181873. [PMID: 39846249 PMCID: PMC11790028 DOI: 10.1172/jci.insight.181873] [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] [Indexed: 01/24/2025] Open
Abstract
T cells targeting a KRAS mutation can induce durable tumor regression in some patients with metastatic epithelial cancer. It is unknown whether T cells targeting mutant KRAS that are capable of killing tumor cells can be identified from peripheral blood of patients with pancreatic cancer. We developed an in vitro stimulation approach and identified HLA-A*11:01-restricted KRAS G12V-reactive CD8+ T cells and HLA-DRB1*15:01-restricted KRAS G12V-reactive CD4+ T cells from peripheral blood of 2 out of 6 HLA-A*11:01-positive patients with pancreatic cancer whose tumors expressed KRAS G12V. The HLA-A*11:01-restricted KRAS G12V-reactive T cell receptor (TCR) was isolated and validated to specifically recognize the KRAS G12V8-16 neoepitope. While T cells engineered to express this TCR specifically recognized all 5 tested human HLA-A*11:01+ and KRAS G12V+ pancreatic cancer organoids, the recognition was often modest, and tumor cell killing was observed in only 2 out of 5 organoids. IFN-γ priming of the organoids enhanced the recognition and killing by the TCR-engineered T cells. The TCR-engineered T cells could significantly slow the growth of an established organoid-derived xenograft in immunodeficient mice. Our data suggest that this TCR has potential for use in TCR-gene therapy, but additional strategies that enhance tumor recognition by the TCR-engineered T cells likely will be required to increase clinical activity.
Collapse
Affiliation(s)
- Xiongfei Xu
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| | - Haihui Gu
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhanshan Cha
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery
| | - Xiaoyi Yin
- Department of Hepatobiliary Pancreatic Surgery
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery
| | - Bo Li
- Department of Hepatobiliary Pancreatic Surgery
| | - Lingyu Zhu
- Department of Hepatobiliary Pancreatic Surgery
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery
| | | | - Zhuo Shao
- Department of Hepatobiliary Pancreatic Surgery
| | - Peng Cheng
- Department of Hepatobiliary Pancreatic Surgery
| | - Chunhong Zheng
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
- International Cancer Institute, Peking University, Beijing, China
| | - Yi-Ping Shih
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Baohua Qian
- Department of Transfusion Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Eric Tran
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery
- Shanghai Institute of Pancreatic Diseases, and
| |
Collapse
|
39
|
Sarno F, Tenorio J, Perea S, Medina L, Pazo-Cid R, Juez I, Garcia-Carbonero R, Feliu J, Guillen-Ponce C, Lopez-Casas PP, Guerra C, Duran Y, López-Acosta JF, Alonso C, Esquivel E, Dopazo A, Akshinthala D, Muthuswamy SK, Lapunzina P, Bockorny B, Hidalgo M. A Phase III Randomized Trial of Integrated Genomics and Avatar Models for Personalized Treatment of Pancreatic Cancer: The AVATAR Trial. Clin Cancer Res 2025; 31:278-287. [PMID: 39540844 PMCID: PMC11739777 DOI: 10.1158/1078-0432.ccr-23-4026] [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: 01/04/2024] [Revised: 04/24/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) has limited treatment options. We compared the efficacy of comprehensive precision medicine against that of the conventional treatment in PDAC. PATIENTS AND METHODS We report a phase III trial of advanced PDAC in which patients were randomized (1:2) to a conventional treatment treated at physician's discretion (arm A) or to precision medicine (arm B). Subjects randomized to arm B underwent a tumor biopsy for whole-exome sequencing and to generate avatar mouse models and patient-derived organoids for phenotypic drug screening, with final treatment recommended by the molecular tumor board. The primary objective was median overall survival (OS). RESULTS A total of 137 patients were enrolled with 125 randomized, 44 to arm A and 81 to arm B. Whole-exome sequencing was performed in 80.3% (65/81) patients of arm B, with potentially actionable mutations detected in 21.5% (14/65). Experimental models were generated in 16/81 patients (19.8%). Second-line treatment was administered to 39 patients in the experimental arm, but only four (10.2%) received personalized treatment, whereas 35 could not receive matched therapy because of rapid clinical deterioration, delays in obtaining study results, or the absence of actionable targets. The median OS was 8.7 and 8.6 months (P = 0.849) and the median progression-free survival was 3.8 and 4.3 months (P = 0.563) for the conventional and experimental arms, respectively. Notably, the four patients who received personalized treatment had a median OS of 19.3 months. CONCLUSIONS Personalized medicine was challenging to implement in most patients with PDAC, limiting the interpretation of intention-to-treat analysis. Survival was improved in the subset of patients who did receive matched therapy.
Collapse
Affiliation(s)
| | - Jair Tenorio
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- INGEMM-IdiPAZ Institute of Medical and Molecular Genetics, Madrid, Spain
- ITHACA, European Reference Network, Brussels, Belgium
| | - Sofia Perea
- Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Laura Medina
- UGCI Medical Oncology, Hospital Regional y Virgen de la Victoria, IBIMA, Malaga, Spain
| | | | - Ignacio Juez
- Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | - Jaime Feliu
- Hospital Universitario La Paz, Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Pedro P. Lopez-Casas
- Instituto de Investigación Sanitaria, Hospital 12 de octubre (imas 12), Madrid, Spain
| | - Carmen Guerra
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Experimental Oncology Group, Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Yolanda Duran
- Hospital Universitario de Fuenlabrada, Madrid, Spain
| | | | | | - Estrella Esquivel
- Unidad de Genómica, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Dopazo
- Unidad de Genómica, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Dipikaa Akshinthala
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Pablo Lapunzina
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- INGEMM-IdiPAZ Institute of Medical and Molecular Genetics, Madrid, Spain
- ITHACA, European Reference Network, Brussels, Belgium
| | - Bruno Bockorny
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Manuel Hidalgo
- Division of Hematology-Oncology, Weill Cornell Medical College, New York, New York
- New York-Presbyterian Hospital, New York, New York
| |
Collapse
|
40
|
Dong F, Zhou J, Wu Y, Gao Z, Li W, Song Z. MicroRNAs in pancreatic cancer drug resistance: mechanisms and therapeutic potential. Front Cell Dev Biol 2025; 12:1499111. [PMID: 39882259 PMCID: PMC11774998 DOI: 10.3389/fcell.2024.1499111] [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: 09/20/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal malignancies, primarily due to its intrinsic resistance to conventional therapies. MicroRNAs (miRNAs), key regulators of gene expression, have been identified as crucial modulators of drug resistance mechanisms in this cancer type. This review synthesizes recent advancements in our understanding of how miRNAs influence treatment efficacy in PC. We have thoroughly summarized and discussed the complex role of miRNA in mediating drug resistance in PC treatment. By highlighting specific miRNAs that are implicated in drug resistance pathways, we provide insights into their functional mechanisms and interactions with key molecular targets. We also explore the potential of miRNA-based strategies as novel therapeutic approaches and diagnostic tools to overcome resistance and improve patient outcomes. Despite promising developments, challenges such as specificity, stability, and effective delivery of miRNA-based therapeutics remain. This review aims to offer a critical perspective on current research and propose future directions for leveraging miRNA-based interventions in the fight against PC.
Collapse
Affiliation(s)
- Fangying Dong
- Emergency Department, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jing Zhou
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yijie Wu
- Department of general practice, Taozhuang Branch of the First People’s Hospital of Jiashan, Jiaxing, Zhejiang, China
| | - Zhaofeng Gao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Weiwei Li
- Emergency Department, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
41
|
Razumovskaya A, Silkina M, Poloznikov A, Kulagin T, Raigorodskaya M, Gorban N, Kudryavtseva A, Fedorova M, Alekseev B, Tonevitsky A, Nikulin S. Predicting patient outcomes with gene-expression biomarkers from colorectal cancer organoids and cell lines. Front Mol Biosci 2025; 12:1531175. [PMID: 39886381 PMCID: PMC11774744 DOI: 10.3389/fmolb.2025.1531175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
Introduction Colorectal cancer (CRC) is characterized by an extremely high mortality rate, mainly caused by the high metastatic potential of this type of cancer. To date, chemotherapy remains the backbone of the treatment of metastatic colorectal cancer. Three main chemotherapeutic drugs used for the treatment of metastatic colorectal cancer are 5-fluorouracil, oxaliplatin and irinotecan which is metabolized to an active compound SN-38. The main goal of this study was to find the genes connected to the resistance to the aforementioned drugs and to construct a predictive gene expression-based classifier to separate responders and non-responders. Methods In this study, we analyzed gene expression profiles of seven patient-derived CRC organoids and performed correlation analyses between gene expression and IC50 values for the three standard-of-care chemotherapeutic drugs. We also included in the study publicly available datasets of colorectal cancer cell lines, thus combining two different in vitro models relevant to cancer research. Logistic regression was used to build gene expression-based classifiers for metastatic Stage IV and non-metastatic Stage II/III CRC patients. Prognostic performance was evaluated through Kaplan-Meier survival analysis and log-rank tests, while independent prognostic significance was assessed using multivariate Cox proportional hazards modeling. Results A small set of genes showed consistent correlation with resistance to chemotherapy across different datasets. While some genes were previously implicated in cancer prognosis and drug response, several were linked to drug resistance for the first time. The resulting gene expression signatures successfully stratified Stage II/III and Stage IV CRC patients, with potential clinical utility for improving treatment outcomes after further validation. Discussion This study highlights the advantages of integrating diverse experimental models, such as organoids and cell lines, to identify novel prognostic biomarkers and enhance the understanding of chemotherapy resistance in CRC.
Collapse
Affiliation(s)
- Alexandra Razumovskaya
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Mariia Silkina
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Andrey Poloznikov
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Timur Kulagin
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Maria Raigorodskaya
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nina Gorban
- Central Clinical Hospital with Polyclinic, Administration of the President of the Russian Federation, Moscow, Russia
| | - Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Boris Alekseev
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Art Photonics GmbH, Berlin, Germany
| | - Sergey Nikulin
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
42
|
Taurin S, Alzahrani R, Aloraibi S, Ashi L, Alharmi R, Hassani N. Patient-derived tumor organoids: A preclinical platform for personalized cancer therapy. Transl Oncol 2025; 51:102226. [PMID: 39622151 DOI: 10.1016/j.tranon.2024.102226] [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: 09/13/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/11/2024] Open
Abstract
Patient-derived tumor organoids (PDTOs) represent a significant advancement in cancer research and personalized medicine. These organoids, derived from various cancer types, have shown the ability to retain the genetic and molecular characteristics of the original tumors, allowing for the detailed study of tumor biology and drug responses on an individual basis. The success rates of establishing PDTOs vary widely and are influenced by factors such as cancer type, tissue quality, and media composition. Furthermore, the dynamic nature of organoid cultures may also lead to unique molecular characteristics that deviate from the original tumors, affecting their interpretation in clinical settings without the implementation of rigorous validation and establishment of standardized protocols. Recent studies have supported the correlation between PDTOs and the corresponding patient response. Although these studies involved a small number of patients, they promoted the integration of PDTOs in observational and interventional clinical trials to advance translational cancer therapies.
Collapse
Affiliation(s)
- Sebastien Taurin
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain.
| | - Reem Alzahrani
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Sahar Aloraibi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Layal Ashi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Rawan Alharmi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Noora Hassani
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
43
|
Arafeh R, Shibue T, Dempster JM, Hahn WC, Vazquez F. The present and future of the Cancer Dependency Map. Nat Rev Cancer 2025; 25:59-73. [PMID: 39468210 DOI: 10.1038/s41568-024-00763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/30/2024]
Abstract
Despite tremendous progress in the past decade, the complex and heterogeneous nature of cancer complicates efforts to identify new therapies and therapeutic combinations that achieve durable responses in most patients. Further advances in cancer therapy will rely, in part, on the development of targeted therapeutics matched with the genetic and molecular characteristics of cancer. The Cancer Dependency Map (DepMap) is a large-scale data repository and research platform, aiming to systematically reveal the landscape of cancer vulnerabilities in thousands of genetically and molecularly annotated cancer models. DepMap is used routinely by cancer researchers and translational scientists and has facilitated the identification of several novel and selective therapeutic strategies for multiple cancer types that are being tested in the clinic. However, it is also clear that the current version of DepMap is not yet comprehensive. In this Perspective, we review (1) the impact and current uses of DepMap, (2) the opportunities to enhance DepMap to overcome its current limitations, and (3) the ongoing efforts to further improve and expand DepMap.
Collapse
Affiliation(s)
- Rand Arafeh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | | | | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
44
|
Gayibov E, Sychra T, Spálenková A, Souček P, Oliverius M. The use of patient-derived xenografts and patient-derived organoids in the search for new therapeutic regimens for pancreatic carcinoma. A review. Biomed Pharmacother 2025; 182:117750. [PMID: 39689516 DOI: 10.1016/j.biopha.2024.117750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024] Open
Abstract
Patient-derived organoids (PDOs) and xenografts (PDXs) are powerful tools for personalized medicine in pancreatic cancer (PC) research. This study explores the complementary strengths of PDOs and PDXs in terms of practicality, genetic fidelity, cost, and labor considerations. Among other models like 2D cell cultures, spheroids, cancer-on-chip systems, cell line-derived xenografts (CDX), and genetically engineered mouse models (GEMMs), PDOs and PDXs uniquely balance genetic fidelity and personalized medicine potential, offering distinct advantages over the simplicity of 2D cultures and the advanced, but often resource-intensive, GEMMs and cancer-on-chip systems. PDOs excel in high-throughput drug screening due to their ease of use, lower cost, and shorter experimental timelines. However, they lack a complete tumor microenvironment. Conversely, PDXs offer a more complex microenvironment that closely reflects patient tumors, potentially leading to more clinically relevant results. Despite limitations in size, number of specimens, and engraftment success, PDXs demonstrate significant concordance with patient responses to treatment, highlighting their value in personalized medicine. Both models exhibit significant genetic fidelity, making them suitable for drug sensitivity testing. The choice between PDOs and PDXs depends on the research focus, resource availability, and desired level of microenvironment complexity. PDOs are advantageous for high-throughput screening of a diverse array of potential therapeutic agents due to their relative ease of culture and scalability. PDXs, on the other hand, offer a more physiologically relevant model, allowing for a comprehensive evaluation of drug efficacy and mechanisms of action.
Collapse
Affiliation(s)
- Emin Gayibov
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomáš Sychra
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Alžběta Spálenková
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- Centre of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | - Martin Oliverius
- 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Department of General Surgery, 3rd Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic.
| |
Collapse
|
45
|
Acimovic I, Gabrielová V, Martínková S, Eid M, Vlažný J, Moravčík P, Hlavsa J, Moráň L, Cakmakci RC, Staňo P, Procházka V, Kala Z, Trnka J, Vaňhara P. Ex-Vivo 3D Cellular Models of Pancreatic Ductal Adenocarcinoma: From Embryonic Development to Precision Oncology. Pancreas 2025; 54:e57-e71. [PMID: 39074056 DOI: 10.1097/mpa.0000000000002393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
ABSTRACT Pancreas is a vital gland of gastrointestinal system with exocrine and endocrine secretory functions, interweaved into essential metabolic circuitries of the human body. Pancreatic ductal adenocarcinoma (PDAC) represents one of the most lethal malignancies, with a 5-year survival rate of 11%. This poor prognosis is primarily attributed to the absence of early symptoms, rapid metastatic dissemination, and the limited efficacy of current therapeutic interventions. Despite recent advancements in understanding the etiopathogenesis and treatment of PDAC, there remains a pressing need for improved individualized models, identification of novel molecular targets, and development of unbiased predictors of disease progression. Here we aim to explore the concept of precision medicine utilizing 3-dimensional, patient-specific cellular models of pancreatic tumors and discuss their potential applications in uncovering novel druggable molecular targets and predicting clinical parameters for individual patients.
Collapse
Affiliation(s)
- Ivana Acimovic
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Viktorie Gabrielová
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Stanislava Martínková
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | - Michal Eid
- Departments of Internal Medicine, Hematology and Oncology
| | | | - Petr Moravčík
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Hlavsa
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | | | - Riza Can Cakmakci
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Peter Staňo
- Departments of Internal Medicine, Hematology and Oncology
| | - Vladimír Procházka
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Zdeněk Kala
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Trnka
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | | |
Collapse
|
46
|
Shen S, Liu B, Guan W, Liu Z, Han Y, Hu Y, Chen Y, Liu S, He J, Li Z, Tang W, Zhang P, Ren W, Qiu Y, Zheng H, Li J. Advancing precision medicine in esophageal squamous cell carcinoma using patient-derived organoids. J Transl Med 2024; 22:1168. [PMID: 39741269 DOI: 10.1186/s12967-024-05967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Patient-derived organoids (PDOs) represent a promising approach for replicating the characteristics of original tumors and facilitating drug testing for personalized treatments across diverse cancer types. However, clinical evidence regarding their application to esophageal cancer remains limited. This study aims to evaluate the efficacy of implementing PDOs in clinical practice to benefit patients with esophageal squamous cell carcinoma (ESCC). METHODS Fresh surgical biopsies were obtained from patients with esophageal cancer for the establishment of PDOs. These PDOs were subsequently characterized through histological analysis. A customized drug panel, based on standard-of-care chemotherapy regimens, was applied to the PDOs. The resulting drug sensitivity profiles were then correlated with the clinical responses observed in individual patients undergoing actual treatment. RESULTS A total of 34 PDOs were successfully established with a 61.8% success rate. The classification method based on chemotherapy sensitivity closely corresponded to clinical responses. The paclitaxel plus cisplatin (TP)-sensitive group demonstrated significantly longer progression-free survival (PFS) compared to the resistant groups, Hazard ratio (HR), 5.12; 95% confidence intervals (CI 0.58-44.71; p < 0.05), thus illustrating the potential of this approach for guiding personalized treatment strategies. CONCLUSION Organoid biobanks were established across multiple institutes to facilitate PDOs-based functional precision medicine. The findings demonstrate that this framework offers robust predictive value in clinical settings, enhances precision therapeutics, and advances drug discovery for esophageal cancer.
Collapse
Affiliation(s)
- Suya Shen
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wenyan Guan
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Ziyao Liu
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yuqing Han
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yingzhe Hu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Yiqiang Chen
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Siyuan Liu
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Zhiwen Li
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Weifeng Tang
- Department of Esophageal Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, NanjingJiangsu, 210008, China
| | - Pengju Zhang
- Zhejiang Honray Medical Technology Co., LTD, Taizhou, 318001, Zhejiang, China
| | - Wei Ren
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Hongping Zheng
- Zhejiang Honray Medical Technology Co., LTD, Taizhou, 318001, Zhejiang, China.
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200032, China.
| | - Jingjing Li
- Department of Precision Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
47
|
Li P, Huang M, Ma Y, Zhang Y, Shi C. Novel research model for in vitro immunotherapy: co-culturing tumor organoids with peripheral blood mononuclear cells. Cancer Cell Int 2024; 24:438. [PMID: 39741287 DOI: 10.1186/s12935-024-03628-3] [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: 04/01/2024] [Accepted: 12/24/2024] [Indexed: 01/02/2025] Open
Abstract
Tumor organoids have emerged as powerful tools for in vitro cancer research due to their ability to retain the structural and genetic characteristics of tumors. Nevertheless, the absence of a complete tumor microenvironment (TME) limits the broader application of organoid models in immunological studies. Given the critical role of immune cells in tumor initiation and progression, the co-culture model of organoids and peripheral blood mononuclear cells (PBMCs) may provide an effective platform for simulating the interactions between immune and tumor cells in vitro. This model stands as a robust instrument for dissecting the TME, elucidating the molecular interactions, and exploring the therapeutic applications of chimeric antigen receptor (CAR)-engineered lymphocytes, as well as other cancer treatment modalities. This review systematically evaluates the advantages and disadvantages of the co-culture model, identifies its technical bottlenecks, and proposes corresponding optimization strategies. By summarizing the latest research advancements in this co-culture model, our goal is to provide valuable insights for further model optimization and clinical application, thereby promoting immunological research and bridging the gap between experimental outcomes and clinical practice.
Collapse
Affiliation(s)
- Peng Li
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Minli Huang
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yifan Ma
- Gansu University of traditional Chinese medicine, Lanzhou, 730030, Gansu, China
| | - Yongbin Zhang
- Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
48
|
Chang CJ, Kan KW, Shih YY, Nan YH, Juan YC, Tu CH, Yang CT, Hsu WL, Cho DY, Shie MY, Chen YW, Chen CC. Preliminary study of utilizing a patient derived tumor spheroid model to augment precision therapy in metastatic brain tumors. Sci Rep 2024; 14:31888. [PMID: 39738713 PMCID: PMC11685586 DOI: 10.1038/s41598-024-83409-0] [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: 04/30/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
Treating metastatic brain tumors remains a significant challenge. This study introduces and applies the Patient-Derived Tumor Spheroid (PDTS) system, an ex vivo model for precision drug testing on metastatic brain tumor. The PDTS system utilizes a decellularized extracellular matrix (dECM) derived from adipose tissue, combined with the tumor cells, to form tumor spheroids. These spheroids were subsequently used to test anticancer drugs, with results compared to the clinical outcomes observed after administering these treatments to patients. To assess the validity of the data, the correlation between the drug responses observed in the PDTS model and actual patient outcomes was analyzed. Chi-square tests evaluated the significance of associations between lab predictions and clinical outcomes, using a significance threshold of p < 0.05. In preliminary data, 17 patients met the criteria for final analysis, which showed an overall 57% accuracy (p-value = 0.463), with improvements to 73% accuracy (p-value = 0.072) when patients receiving certain treatments were excluded. This PDTS offers real-time results within three weeks, simultaneous testing of multiple drugs, and the ability to culture and store tumor cells for reproducibility. Despite some limitations, further development of this model could enhance its clinical application and improve patient outcomes.
Collapse
Affiliation(s)
- Chun-Jen Chang
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Kai-Wen Kan
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Yu-Yin Shih
- Research and Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Yu-Han Nan
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Yu-Chung Juan
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Chih-Hsiu Tu
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Chen-Ting Yang
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Wei-Lin Hsu
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Der-Yang Cho
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Ming-You Shie
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung, 404332, Taiwan
- School of Dentistry, China Medical University, Taichung, 406040, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
| | - Yi-Wen Chen
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung, 404332, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, 406040, Taiwan
| | - Chun-Chung Chen
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC.
- Graduate Institute of Acupuncture Science, China Medical University, Taichung City, 40402, Taiwan, ROC.
- Neuroscience and Brain Disease Center, China Medical University, Li-fu Hall 8F, No. 91, Xue Shi Rd., North Dist., Taichung City, 40402, Taiwan, ROC.
- School of Medicine, China Medical University, Taichung, Taiwan.
- Department of Neurosurgery, China Medical University Hospital, 2, Yu-Der Road, Taichung, 40447, Taiwan.
| |
Collapse
|
49
|
Roalsø MTT, Alexeeva M, Oanæs C, Watson M, Lea D, Zaharia C, Hagland HR, Søreide K. Patient-derived organoids from pancreatic cancer after pancreatectomy: Feasibility and organoid take rate in treatment-naïve periampullary tumors. Pancreatology 2024:S1424-3903(24)00846-9. [PMID: 39734118 DOI: 10.1016/j.pan.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND/OBJECTIVE Patient-derived organoids (PDOs) have emerged as essential for ex vivo modelling for pancreatic cancer (PDAC) but reports on efficacy and organoid take rate are scarce. This study aimed to assess the feasibility of establishing PDOs from resected specimens in periampullary tumors. METHODS Patients undergoing surgery for suspected periampullary cancer were included. PDO protocol amendments were tested, with organoid take rate as outcome measure. Samples from resected specimens were processed and expanded per protocol. Pooled estimate of take rates of PDOs in PDAC was derived from literature search. RESULTS 23 specimens were available for PDO, of which 10 were PDAC. In 15 patients other histopathology was found: neuroendocrine tumors (NET; n = 2), neuroendocrine carcinoma (NEC; n = 1), intraductal papillary mucinous neoplasm (IPMN; n = 4), distal cholangiocarcinoma (dCCA; n = 1), ampullary carcinoma (n = 1), duodenal carcinoma (n = 1), intra-ampullary papillary tubular neoplasm (IAPN; n = 1), indeterminate PDAC/ampullary carcinoma(n = 1), and one patient with chronic inflammation/fibrosis. Organoid cultures were grown from 7 of 10 (70 %) PDAC, 1 dCCA, 1 NEC, 1 duodenal carcinoma, 1 indeterminate tumor type and 1 ampullary carcinoma (i.e. 12/18; 66.7 % across periampullary cancers). Overall take rate of PDOs was 12 of 23 (52.2 %) for all tumors. A pooled mean estimate PDO take rate of 62.3 % (95 % CI:54.8-69.3 %) was reported across available studies in the literature. CONCLUSION In the current study, we found that PDOs could be established from resected pancreatic tumors in over half of resected periampullary tumors, and highest in PDACs. As such, generating a pancreatic cancer PDO biobank for translational research was feasible after cryopreservation.
Collapse
Affiliation(s)
- Marcus T T Roalsø
- Department of Quality and Health Technology, University of Stavanger, Stavanger, Norway; Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway; Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway
| | - Marina Alexeeva
- Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway; Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway
| | - Celine Oanæs
- Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway; Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway
| | - Martin Watson
- Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway; Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway
| | - Dordi Lea
- Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway; Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Claudia Zaharia
- Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway; Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Hanne R Hagland
- Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway; Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Kjetil Søreide
- Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway; Gastrointestinal Translational Research Unit, Stavanger University Hospital, Stavanger, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
50
|
Strocchi S, Santandrea G, Zanetti E, Verna G, Cusenza VY, Nicoli D, Fantini V, Grieco A, Paci M, Ciarrocchi A, Sancisi V. A Two-Step Protocol for Isolation and Maintenance of Lung Cancer Primary 3D Cultures. Cancers (Basel) 2024; 17:27. [PMID: 39796658 PMCID: PMC11718983 DOI: 10.3390/cancers17010027] [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: 12/06/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Despite the introduction of innovative therapeutics, lung cancer is still the leading cause of cancer-related death. For this reason, lung cancer still requires deep characterization to identify cellular and molecular targets that can be used to develop novel therapeutic strategies. Three-dimensional cellular models, including patient-derived organoids (PDOs), represent useful tools to study lung cancer biology and may be employed in the future as predictive tools in therapeutic decisions. However, the successful establishment of lung cancer organoids cultures that faithfully represent the respective patient tissues is still challenging due to low success rate and/or overgrowth of normal airway epithelial cells. METHODS We set up a two-step protocol that allows for establishing both short-term and long-term 3D cultures, with different characteristics and success rates. RESULTS Cancer tissue-originated spheroids (CTOSs) show a 100% success rate and allow for the concomitant isolation of autologous tumor infiltrating leukocytes (TILs). On the contrary, PDOs can be expanded for a medium-long term and bio-banked but retain a lower success rate and the possibility of contamination with normal airway epithelial cells. To overcome these problems, we set up an optimal medium formulation and we implemented rigorous quality controls, leading to a substantial improvement in the success rate of tumoral PDO establishment. CONCLUSIONS Overall, this protocol guarantees flexibility and reliability, also providing useful guidelines for quality control checks to support different experimental settings. The setting up of a robust protocol for lung cancer PDO culture establishment and expansion is a key requirement for their employment both in cancer research and as predictive tools in clinical practice.
Collapse
Affiliation(s)
- Silvia Strocchi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (S.S.); (G.V.); (V.F.); (A.G.); (A.C.)
| | - Giacomo Santandrea
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.S.); (E.Z.); (V.Y.C.); (D.N.)
| | - Eleonora Zanetti
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.S.); (E.Z.); (V.Y.C.); (D.N.)
- Biobank, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
| | - Giulio Verna
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (S.S.); (G.V.); (V.F.); (A.G.); (A.C.)
| | - Vincenza Ylenia Cusenza
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.S.); (E.Z.); (V.Y.C.); (D.N.)
| | - Davide Nicoli
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (G.S.); (E.Z.); (V.Y.C.); (D.N.)
| | - Valentina Fantini
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (S.S.); (G.V.); (V.F.); (A.G.); (A.C.)
| | - Alessandra Grieco
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (S.S.); (G.V.); (V.F.); (A.G.); (A.C.)
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Massimiliano Paci
- Thoracic Surgery Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Alessia Ciarrocchi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (S.S.); (G.V.); (V.F.); (A.G.); (A.C.)
| | - Valentina Sancisi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy; (S.S.); (G.V.); (V.F.); (A.G.); (A.C.)
| |
Collapse
|